1
|
Talarmin-Gas C, Smolyakov G, Parisi C, Scandola C, Andrianasolonirina V, Lecoq C, Houtart V, Lee SH, Adle-Biassette H, Thiébot B, Ganderton T, Manivet P. Validation of metaxin-2 deficient C. elegans as a model for MandibuloAcral Dysplasia associated to mtx-2 (MADaM) syndrome. Commun Biol 2024; 7:1398. [PMID: 39462037 PMCID: PMC11513083 DOI: 10.1038/s42003-024-06967-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
MandibuloAcral Dysplasia associated to MTX2 gene (MADaM) is a recently described progeroid syndrome (accelerated aging disease) whose clinical manifestations include skin abnormalities, growth retardation, and cardiovascular diseases. We previously proposed that mtx-2-deficient C. elegans could be used as a model for MADaM and to support this, we present here our comprehensive phenotypic characterization of these worms using atomic force microscopy (AFM), transcriptomic, and oxygen consumption rate analyses. AFM analysis showed that young mtx-2-less worms had a significantly rougher, less elastic cuticle which becomes significantly rougher and less elastic as they age, and abnormal mitochondrial morphology. mtx-2 C. elegans displayed slightly delayed development, decreased pharyngeal pumping, significantly reduced mitochondrial respiratory capacities, and transcriptomic analysis identified perturbations in the aging, TOR, and WNT-signaling pathways. The phenotypic characteristics of mtx-2 worms shown here are analogous to many of the human clinical presentations of MADaM and we believe this validates their use as a model which will allow us to uncover the molecular details of the disease and develop new therapeutics and treatments.
Collapse
Affiliation(s)
- Chloé Talarmin-Gas
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France.
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France.
| | - Georges Smolyakov
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Cleo Parisi
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Cyril Scandola
- Institut Pasteur, Université Paris Cité, Ultrastructural Bioimaging Unit, 75015, Paris, France
| | - Valérie Andrianasolonirina
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Cloé Lecoq
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Valentine Houtart
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | | | - Homa Adle-Biassette
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
- AP-HP, DMU DREAM, Service d'Anatomocytopathologie, Hôpital Lariboisière, Paris, France
| | - Bénédicte Thiébot
- CY Cergy Paris Université, Université d'Evry, Université Paris-Saclay, CNRS, LAMBE, F-95000, Cergy, France
| | - Timothy Ganderton
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Philippe Manivet
- Université Paris Cité, INSERM UMR 1141 "NeuroDiderot", FHU Iio2-D2, Paris, France.
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France.
- CeleScreen SAS, Paris, France.
| |
Collapse
|
2
|
McPhee M, Dellaire G, Ridgway ND. Mechanisms for assembly of the nucleoplasmic reticulum. Cell Mol Life Sci 2024; 81:415. [PMID: 39367888 PMCID: PMC11455740 DOI: 10.1007/s00018-024-05437-3] [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: 06/28/2024] [Revised: 08/22/2024] [Accepted: 09/03/2024] [Indexed: 10/07/2024]
Abstract
The nuclear envelope consists of an outer membrane connected to the endoplasmic reticulum, an inner membrane facing the nucleoplasm and a perinuclear space separating the two bilayers. The inner and outer nuclear membranes are physically connected at nuclear pore complexes that mediate selective communication and transfer of materials between the cytoplasm and nucleus. The spherical shape of the nuclear envelope is maintained by counterbalancing internal and external forces applied by cyto- and nucleo-skeletal networks, and the nuclear lamina and chromatin that underly the inner nuclear membrane. Despite its apparent rigidity, the nuclear envelope can invaginate to form an intranuclear membrane network termed the nucleoplasmic reticulum (NR) consisting of Type-I NR contiguous with the inner nuclear membrane and Type-II NR containing both the inner and outer nuclear membranes. The NR extends deep into the nuclear interior potentially facilitating communication and exchanges between the nuclear interior and the cytoplasm. This review details the evidence that NR intrusions that regulate cytoplasmic communication and genome maintenance are the result of a dynamic interplay between membrane biogenesis and remodelling, and physical forces exerted on the nuclear lamina derived from the cyto- and nucleo-skeletal networks.
Collapse
Affiliation(s)
- Michael McPhee
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, B3H4R2, Canada
| | - Graham Dellaire
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, B3H4R2, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, B3H4R2, Canada
| | - Neale D Ridgway
- Department of Biochemistry & Molecular Biology, Dalhousie University, Halifax, NS, B3H4R2, Canada.
- Department of Pediatrics, Atlantic Research Centre, Dalhousie University, Halifax, NS, B3H4R2, Canada.
| |
Collapse
|
3
|
Ege T, Tao L, North BJ. The Role of Molecular and Cellular Aging Pathways on Age-Related Hearing Loss. Int J Mol Sci 2024; 25:9705. [PMID: 39273652 PMCID: PMC11396656 DOI: 10.3390/ijms25179705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024] Open
Abstract
Aging, a complex process marked by molecular and cellular changes, inevitably influences tissue and organ homeostasis and leads to an increased onset or progression of many chronic diseases and conditions, one of which is age-related hearing loss (ARHL). ARHL, known as presbycusis, is characterized by the gradual and irreversible decline in auditory sensitivity, accompanied by the loss of auditory sensory cells and neurons, and the decline in auditory processing abilities associated with aging. The extended human lifespan achieved by modern medicine simultaneously exposes a rising prevalence of age-related conditions, with ARHL being one of the most significant. While our understanding of the molecular basis for aging has increased over the past three decades, a further understanding of the interrelationship between the key pathways controlling the aging process and the development of ARHL is needed to identify novel targets for the treatment of AHRL. The dysregulation of molecular pathways (AMPK, mTOR, insulin/IGF-1, and sirtuins) and cellular pathways (senescence, autophagy, and oxidative stress) have been shown to contribute to ARHL. However, the mechanistic basis for these pathways in the initiation and progression of ARHL needs to be clarified. Therefore, understanding how longevity pathways are associated with ARHL will directly influence the development of therapeutic strategies to treat or prevent ARHL. This review explores our current understanding of the molecular and cellular mechanisms of aging and hearing loss and their potential to provide new approaches for early diagnosis, prevention, and treatment of ARHL.
Collapse
Affiliation(s)
- Tuba Ege
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Litao Tao
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, NE 68178, USA
| | - Brian J North
- Biomedical Sciences Department, School of Medicine, Creighton University, Omaha, NE 68178, USA
| |
Collapse
|
4
|
Díaz-López EJ, Sánchez-Iglesias S, Castro AI, Cobelo-Gómez S, Prado-Moraña T, Araújo-Vilar D, Fernandez-Pombo A. Lipodystrophic Laminopathies: From Dunnigan Disease to Progeroid Syndromes. Int J Mol Sci 2024; 25:9324. [PMID: 39273270 PMCID: PMC11395136 DOI: 10.3390/ijms25179324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/15/2024] Open
Abstract
Lipodystrophic laminopathies are a group of ultra-rare disorders characterised by the presence of pathogenic variants in the same gene (LMNA) and other related genes, along with an impaired adipose tissue pattern and other features that are specific of each of these disorders. The most fascinating traits include their complex genotype-phenotype associations and clinical heterogeneity, ranging from Dunnigan disease, in which the most relevant feature is precisely adipose tissue dysfunction and lipodystrophy, to the other laminopathies affecting adipose tissue, which are also characterised by the presence of signs of premature ageing (Hutchinson Gilford-progeria syndrome, LMNA-atypical progeroid syndrome, mandibuloacral dysplasia types A and B, Nestor-Guillermo progeria syndrome, LMNA-associated cardiocutaneous progeria). This raises several questions when it comes to understanding how variants in the same gene can lead to similar adipose tissue disturbances and, at the same time, to such heterogeneous phenotypes and variable degrees of metabolic abnormalities. The present review aims to gather the molecular basis of adipose tissue impairment in lipodystrophic laminopathies, their main clinical aspects and recent therapeutic strategies. In addition, it also summarises the key aspects for their differential diagnosis.
Collapse
Affiliation(s)
- Everardo Josué Díaz-López
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Sofía Sánchez-Iglesias
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Ana I Castro
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBERobn), 28029 Madrid, Spain
| | - Silvia Cobelo-Gómez
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Teresa Prado-Moraña
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - David Araújo-Vilar
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| | - Antia Fernandez-Pombo
- UETeM-Molecular Pathology Group, Department of Psychiatry, Radiology, Public Health, Nursing and Medicine, IDIS-CIMUS, University of Santiago de Compostela, 15706 Santiago de Compostela, Spain
- Division of Endocrinology and Nutrition, University Clinical Hospital of Santiago de Compostela, 15706 Santiago de Compostela, Spain
| |
Collapse
|
5
|
Shores KL, Truskey GA. Mechanotransduction of the vasculature in Hutchinson-Gilford Progeria Syndrome. Front Physiol 2024; 15:1464678. [PMID: 39239311 PMCID: PMC11374724 DOI: 10.3389/fphys.2024.1464678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disorder that causes severe cardiovascular disease, resulting in the death of patients in their teenage years. The disease pathology is caused by the accumulation of progerin, a mutated form of the nuclear lamina protein, lamin A. Progerin binds to the inner nuclear membrane, disrupting nuclear integrity, and causes severe nuclear abnormalities and changes in gene expression. This results in increased cellular inflammation, senescence, and overall dysfunction. The molecular mechanisms by which progerin induces the disease pathology are not fully understood. Progerin's detrimental impact on nuclear mechanics and the role of the nucleus as a mechanosensor suggests dysfunctional mechanotransduction could play a role in HGPS. This is especially relevant in cells exposed to dynamic, continuous mechanical stimuli, like those of the vasculature. The endothelial (ECs) and smooth muscle cells (SMCs) within arteries rely on physical forces produced by blood flow to maintain function and homeostasis. Certain regions within arteries produce disturbed flow, leading to an impaired transduction of mechanical signals, and a reduction in cellular function, which also occurs in HGPS. In this review, we discuss the mechanics of nuclear mechanotransduction, how this is disrupted in HGPS, and what effect this has on cell health and function. We also address healthy responses of ECs and SMCs to physiological mechanical stimuli and how these responses are impaired by progerin accumulation.
Collapse
Affiliation(s)
- Kevin L Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| |
Collapse
|
6
|
Kim SJ, Park SH, Myung K, Lee KY. Lamin A/C facilitates DNA damage response by modulating ATM signaling and homologous recombination pathways. Anim Cells Syst (Seoul) 2024; 28:401-416. [PMID: 39176289 PMCID: PMC11340224 DOI: 10.1080/19768354.2024.2393820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/05/2024] [Accepted: 08/11/2024] [Indexed: 08/24/2024] Open
Abstract
Lamin A/C, a core component of the nuclear lamina, forms a mesh-like structure beneath the inner nuclear membrane. While its structural role is well-studied, its involvement in DNA metabolism remains unclear. We conducted sequential protein fractionation to determine the subcellular localization of early DNA damage response (DDR) proteins. Our findings indicate that most DDR proteins, including ATM and the MRE11-RAD50-NBS1 (MRN) complex, are present in the nuclease - and high salt-resistant pellet fraction. Notably, ATM and MRN remain stably associated with these structures throughout the cell cycle, independent of ionizing radiation (IR)-induced DNA damage. Although Lamin A/C interacts with ATM and MRN, its depletion does not disrupt their association with nuclease-resistant structures. However, it impairs the IR-enhanced association of ATM with the nuclear matrix and ATM-mediated DDR signaling, as well as the interaction between ATM and MRN. This disruption impedes the recruitment of MRE11 to damaged DNA and the association of damaged DNA with the nuclear matrix. Additionally, Lamin A/C depletion results in reduced protein levels of CtIP and RAD51, which is mediated by transcriptional regulation. This, in turn, impairs the efficiency of homologous recombination (HR). Our findings indicate that Lamin A/C plays a pivotal role in DNA damage repair (DDR) by orchestrating ATM-mediated signaling, maintaining HR protein levels, and ensuring efficient DNA repair processes.
Collapse
Affiliation(s)
- Seong-jung Kim
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Korea
- Department of Biological Sciences, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Su Hyung Park
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Korea
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Korea
- Department of Biomedical Engineering, College of Information-Bio Convergence Engineering, Ulsan National Institute of Science and Technology, Ulsan, Korea
| | - Kyoo-young Lee
- Center for Genomic Integrity, Institute for Basic Science, Ulsan, Korea
- Department of Biochemistry, College of Medicine, Hallym University, Chuncheon, Korea
| |
Collapse
|
7
|
Zych MG, Hatch EM. Small spaces, big problems: The abnormal nucleoplasm of micronuclei and its consequences. Curr Opin Struct Biol 2024; 87:102839. [PMID: 38763098 DOI: 10.1016/j.sbi.2024.102839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/29/2024] [Accepted: 04/26/2024] [Indexed: 05/21/2024]
Abstract
Micronuclei (MN) form from missegregated chromatin that recruits its own nuclear envelope during mitotic exit and are a common consequence of chromosomal instability. MN are unstable due to errors in nuclear envelope organization and frequently rupture, leading to loss of compartmentalization, loss of nuclear functions, and major changes in genome stability and gene expression. However, recent work found that, even prior to rupture, nuclear processes can be severely defective in MN, which may contribute to rupture-associated defects and have lasting consequences for chromatin structure and function. In this review we discuss work that highlights nuclear function defects in intact MN, including their mechanisms and consequences, and how biases in chromosome missegregation into MN may affect the penetrance of these defects. Illuminating the nuclear environment of MN demonstrates that MN formation alone has major consequences for both the genome and cell and provides new insight into how nuclear content is regulated.
Collapse
Affiliation(s)
- Molly G Zych
- Molecular and Cellular Biology PhD Program, University of Washington, Seattle, WA, USA; Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA. https://twitter.com/ZychMolly
| | - Emily M Hatch
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA; Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
8
|
Joudeh LA, Logan Schuck P, Van NM, DiCintio AJ, Stewart JA, Waldman AS. Progerin Can Induce DNA Damage in the Absence of Global Changes in Replication or Cell Proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.02.601729. [PMID: 39005395 PMCID: PMC11244969 DOI: 10.1101/2024.07.02.601729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare genetic condition characterized by features of accelerated aging, and individuals with HGPS seldom live beyond their mid-teens. The syndrome is commonly caused by a point mutation in the LMNA gene which codes for lamin A and its splice variant lamin C, components of the nuclear lamina. The mutation causing HGPS leads to production of a truncated, farnesylated form of lamin A referred to as "progerin." Progerin is also expressed at low levels in healthy individuals and appears to play a role in normal aging. HGPS is associated with an accumulation of genomic DNA double-strand breaks (DSBs) and alterations in the nature of DSB repair. The source of DSBs in HGPS is often attributed to stalling and subsequent collapse of replication forks in conjunction with faulty recruitment of repair factors to damage sites. In this work, we used a model system involving immortalized human cell lines to investigate progerin-induced genomic damage. Using an immunofluorescence approach to visualize phosphorylated histone H2AX foci which mark sites of genomic damage, we report that cells engineered to express progerin displayed a significant elevation of endogenous damage in the absence of any change in the cell cycle profile or doubling time of cells. Genomic damage was enhanced and persistent in progerin-expressing cells treated with hydroxyurea. Overexpression of wild-type lamin A did not elicit the outcomes associated with progerin expression. Our results show that DNA damage caused by progerin can occur independently from global changes in replication or cell proliferation.
Collapse
Affiliation(s)
- Liza A. Joudeh
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208
| | - P. Logan Schuck
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208
| | - Nina M. Van
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208
| | - Alannah J. DiCintio
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208
| | - Jason A. Stewart
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208
- Department of Biology, Western Kentucky University, Bowling Green, KY 42101
| | - Alan S. Waldman
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208
| |
Collapse
|
9
|
Wang M, Zhang J, Qiu J, Ma X, Xu C, Wu Q, Xing S, Chen X, Liu B. Doxycycline decelerates aging in progeria mice. Aging Cell 2024; 23:e14188. [PMID: 38686927 PMCID: PMC11258430 DOI: 10.1111/acel.14188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/26/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024] Open
Abstract
Beyond the antimicrobial activity, doxycycline (DOX) exhibits longevity-promoting effect in nematodes, while its effect on mammals is unclear. Here, we applied a mouse model of Hutchinson-Gilford progeria syndrome (HGPS), Zmpste24 knockout (KO) mice, and analyzed the antiaging effect of DOX. We found that the DOX treatment prolongs lifespan and ameliorates progeroid features of Zmpste24 KO mice, including the decline of body and tissue weight, exercise capacity and cortical bone density, and the shortened colon length. DOX treatment alleviates the abnormal nuclear envelope in multiple tissues, and attenuates cellular senescence and cell death of Zmpste24 KO and HGPS fibroblasts. DOX downregulates the level of proinflammatory IL6 in both serum and tissues. Moreover, the elevated α-tubulin (K40) acetylation mediated by NAT10 in progeria, is rescued by DOX treatment in the aorta tissues in Zmpste24 KO mice and fibroblasts. Collectively, our study uncovers that DOX can decelerate aging in progeria mice via counteracting IL6 expression and NAT10-mediated acetylation of α-tubulin.
Collapse
Affiliation(s)
- Ming Wang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jie Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Jiangping Qiu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xuan Ma
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Chenzhong Xu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Qiuhuan Wu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Shaojun Xing
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Xinchun Chen
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL‐SAI), Marshall Laboratory of Biomedical Engineering, International Cancer Center, School of Basic Medical SciencesShenzhen University Medical SchoolShenzhenChina
| |
Collapse
|
10
|
Ngubo M, Chen Z, McDonald D, Karimpour R, Shrestha A, Yockell‐Lelièvre J, Laurent A, Besong OTO, Tsai EC, Dilworth FJ, Hendzel MJ, Stanford WL. Progeria-based vascular model identifies networks associated with cardiovascular aging and disease. Aging Cell 2024; 23:e14150. [PMID: 38576084 PMCID: PMC11258467 DOI: 10.1111/acel.14150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 03/01/2024] [Accepted: 03/05/2024] [Indexed: 04/06/2024] Open
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) is a lethal premature aging disorder caused by a de novo heterozygous mutation that leads to the accumulation of a splicing isoform of Lamin A termed progerin. Progerin expression deregulates the organization of the nuclear lamina and the epigenetic landscape. Progerin has also been observed to accumulate at low levels during normal aging in cardiovascular cells of adults that do not carry genetic mutations linked with HGPS. Therefore, the molecular mechanisms that lead to vascular dysfunction in HGPS may also play a role in vascular aging-associated diseases, such as myocardial infarction and stroke. Here, we show that HGPS patient-derived vascular smooth muscle cells (VSMCs) recapitulate HGPS molecular hallmarks. Transcriptional profiling revealed cardiovascular disease remodeling and reactive oxidative stress response activation in HGPS VSMCs. Proteomic analyses identified abnormal acetylation programs in HGPS VSMC replication fork complexes, resulting in reduced H4K16 acetylation. Analysis of acetylation kinetics revealed both upregulation of K16 deacetylation and downregulation of K16 acetylation. This correlates with abnormal accumulation of error-prone nonhomologous end joining (NHEJ) repair proteins on newly replicated chromatin. The knockdown of the histone acetyltransferase MOF recapitulates preferential engagement of NHEJ repair activity in control VSMCs. Additionally, we find that primary donor-derived coronary artery vascular smooth muscle cells from aged individuals show similar defects to HGPS VSMCs, including loss of H4K16 acetylation. Altogether, we provide insight into the molecular mechanisms underlying vascular complications associated with HGPS patients and normative aging.
Collapse
Affiliation(s)
- Mzwanele Ngubo
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
| | - Zhaoyi Chen
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
| | - Darin McDonald
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Rana Karimpour
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - Amit Shrestha
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Julien Yockell‐Lelièvre
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
| | - Aurélie Laurent
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Université de StrasbourgStrasbourgFrance
| | - Ojong Tabi Ojong Besong
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- School of BioscienceUniversity of SkövdeSkövdeSweden
| | - Eve C. Tsai
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Division of Neurosurgery, Department of Surgery, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - F. Jeffrey Dilworth
- Department of Cell and Regenerative BiologyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Michael J. Hendzel
- Cross Cancer Institute and the Department of Experimental Oncology, Faculty of Medicine and DentistryUniversity of AlbertaEdmontonAlbertaCanada
| | - William L. Stanford
- The Sprott Centre for Stem Cell ResearchOttawa Hospital Research InstituteOttawaOntarioCanada
- Ottawa Institute of Systems BiologyOttawaOntarioCanada
- Department of Cellular and Molecular MedicineUniversity of OttawaOttawaOntarioCanada
- Department of Biochemistry, Microbiology & ImmunologyUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
11
|
Zhang J, Sun P, Wu Z, Wu J, Jia J, Zou H, Mo Y, Zhou Z, Liu B, Ao Y, Wang Z. Targeting CK2 eliminates senescent cells and prolongs lifespan in Zmpste24-deficient mice. Cell Death Dis 2024; 15:380. [PMID: 38816370 PMCID: PMC11139886 DOI: 10.1038/s41419-024-06760-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/01/2024]
Abstract
Senescent cell clearance is emerging as a promising strategy for treating age-related diseases. Senolytics are small molecules that promote the clearance of senescent cells; however, senolytics are uncommon and their underlying mechanisms remain largely unknown. Here, we investigated whether genomic instability is a potential target for senolytic. We screened small-molecule kinase inhibitors involved in the DNA damage response (DDR) in Zmpste24-/- mouse embryonic fibroblasts, a progeroid model characterized with impaired DDR and DNA repair. 4,5,6,7-tetrabromo-2-azabenzamidazole (TBB), which specifically inhibits casein kinase 2 (CK2), was selected and discovered to preferentially trigger apoptosis in Zmpste24-/- cells. Mechanistically, inhibition of CK2 abolished the phosphorylation of heterochromatin protein 1α (HP1α), which retarded the dynamic HP1α dissociation from repressive histone mark H3K9me3 and its relocalization with γH2AX to DNA damage sites, suggesting that disrupting heterochromatin remodeling in the initiation of DDR accelerates apoptosis in senescent cells. Furthermore, feeding Zmpste24-deficient mice with TBB alleviated progeroid features and extended their lifespan. Our study identified TBB as a new class senolytic compound that can reduce age-related symptoms and prolong lifespan in progeroid mice.
Collapse
Affiliation(s)
- Jie Zhang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Shenzhen University, Shenzhen, 518055, China
| | - Pengfei Sun
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Zhuping Wu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Jie Wu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Jiali Jia
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Haoman Zou
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Yanzhen Mo
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Baohua Liu
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Shenzhen University, Shenzhen, 518055, China
| | - Ying Ao
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Shenzhen University, Shenzhen, 518055, China.
| | - Zimei Wang
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Carson International Cancer Center, Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518055, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention, National Engineering Research Center for Biotechnology (Shenzhen), Shenzhen University, Shenzhen, 518055, China.
| |
Collapse
|
12
|
Chen WA, Boskovic DS. Neutrophil Extracellular DNA Traps in Response to Infection or Inflammation, and the Roles of Platelet Interactions. Int J Mol Sci 2024; 25:3025. [PMID: 38474270 DOI: 10.3390/ijms25053025] [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: 11/30/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Neutrophils present the host's first line of defense against bacterial infections. These immune effector cells are mobilized rapidly to destroy invading pathogens by (a) reactive oxygen species (ROS)-mediated oxidative bursts and (b) via phagocytosis. In addition, their antimicrobial service is capped via a distinct cell death mechanism, by the release of their own decondensed nuclear DNA, supplemented with a variety of embedded proteins and enzymes. The extracellular DNA meshwork ensnares the pathogenic bacteria and neutralizes them. Such neutrophil extracellular DNA traps (NETs) have the potential to trigger a hemostatic response to pathogenic infections. The web-like chromatin serves as a prothrombotic scaffold for platelet adhesion and activation. What is less obvious is that platelets can also be involved during the initial release of NETs, forming heterotypic interactions with neutrophils and facilitating their responses to pathogens. Together, the platelet and neutrophil responses can effectively localize an infection until it is cleared. However, not all microbial infections are easily cleared. Certain pathogenic organisms may trigger dysregulated platelet-neutrophil interactions, with a potential to subsequently propagate thromboinflammatory processes. These may also include the release of some NETs. Therefore, in order to make rational intervention easier, further elucidation of platelet, neutrophil, and pathogen interactions is still needed.
Collapse
Affiliation(s)
- William A Chen
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Pharmaceutical and Administrative Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA 92350, USA
| | - Danilo S Boskovic
- Division of Biochemistry, Department of Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
- Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA
| |
Collapse
|
13
|
Escudeiro-Lopes S, Filimonenko VV, Jarolimová L, Hozák P. Lamin A/C and PI(4,5)P2-A Novel Complex in the Cell Nucleus. Cells 2024; 13:399. [PMID: 38474363 DOI: 10.3390/cells13050399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024] Open
Abstract
Lamins, the nuclear intermediate filaments, are important regulators of nuclear structural integrity as well as nuclear functional processes such as DNA transcription, replication and repair, and epigenetic regulations. A portion of phosphorylated lamin A/C localizes to the nuclear interior in interphase, forming a lamin A/C pool with specific properties and distinct functions. Nucleoplasmic lamin A/C molecular functions are mainly dependent on its binding partners; therefore, revealing new interactions could give us new clues on the lamin A/C mechanism of action. In the present study, we show that lamin A/C interacts with nuclear phosphoinositides (PIPs), and with nuclear myosin I (NM1). Both NM1 and nuclear PIPs have been previously reported as important regulators of gene expression and DNA damage/repair. Furthermore, phosphorylated lamin A/C forms a complex with NM1 in a phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2)-dependent manner in the nuclear interior. Taken together, our study reveals a previously unidentified interaction between phosphorylated lamin A/C, NM1, and PI(4,5)P2 and suggests new possible ways of nucleoplasmic lamin A/C regulation, function, and importance for the formation of functional nuclear microdomains.
Collapse
Affiliation(s)
- Sara Escudeiro-Lopes
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Vlada V Filimonenko
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
- Electron Microscopy Core Facility, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Lenka Jarolimová
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics of the Czech Academy of Sciences, Vídeňská 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
14
|
Chen Y, Zhen Z, Chen L, Wang H, Wang X, Sun X, Song Z, Wang H, Lin Y, Zhang W, Wu G, Jiang Y, Mao Z. Androgen signaling stabilizes genomes to counteract senescence by promoting XRCC4 transcription. EMBO Rep 2023; 24:e56984. [PMID: 37955230 PMCID: PMC10702805 DOI: 10.15252/embr.202356984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 10/16/2023] [Accepted: 10/24/2023] [Indexed: 11/14/2023] Open
Abstract
Aging is accompanied by a decreased DNA repair capacity, which might contribute to age-associated functional decline in multiple tissues. Disruption in hormone signaling, associated with reproductive organ dysfunction, is an early event of age-related tissue degeneration, but whether it impacts DNA repair in nonreproductive organs remains elusive. Using skin fibroblasts derived from healthy donors with a broad age range, we show here that the downregulation of expression of XRCC4, a factor involved in nonhomologous end-joining (NHEJ) repair, which is the dominant pathway to repair somatic double-strand breaks, is mediated through transcriptional mechanisms. We show that the androgen receptor (AR), whose expression is also reduced during aging, directly binds to and enhances the activity of the XRCC4 promoter, facilitating XRCC4 transcription and thus stabilizing the genome. We also demonstrate that dihydrotestosterone (DHT), a powerful AR agonist, restores XRCC4 expression and stabilizes the genome in different models of cellular aging. Moreover, DHT treatment reverses senescence-associated phenotypes, opening a potential avenue to aging interventions in the future.
Collapse
Affiliation(s)
- Yu Chen
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhengyi Zhen
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lingjiang Chen
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hao Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xuhui Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoxiang Sun
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhiwei Song
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Haiyan Wang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yizi Lin
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wenjun Zhang
- Department of Plastic Surgery, Changzheng Hospital, Shanghai, China
| | - Guizhu Wu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, Shanghai Tongji University School of Medicine, Shanghai, China
| | - Ying Jiang
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
15
|
Lian J, Du L, Li Y, Yin Y, Yu L, Wang S, Ma H. Hutchinson-Gilford progeria syndrome: Cardiovascular manifestations and treatment. Mech Ageing Dev 2023; 216:111879. [PMID: 37832833 DOI: 10.1016/j.mad.2023.111879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/04/2023] [Accepted: 10/09/2023] [Indexed: 10/15/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), also known as hereditary progeria syndrome, is caused by mutations in the LMNA gene and the expression of progerin, which causes accelerated aging and premature death, with most patients dying of heart failure or other cardiovascular complications in their teens. HGPS patients are able to exhibit cardiovascular phenotypes similar to physiological aging, such as extensive atherosclerosis, smooth muscle cell loss, vascular lesions, and electrical and functional abnormalities of the heart. It also excludes the traditional risk causative factors of cardiovascular disease, making HGPS a new model for studying aging-related cardiovascular disease. Here, we analyzed the pathogenesis and pathophysiological characteristics of HGPS and the relationship between HGPS and cardiovascular disease, provided insight into the molecular mechanisms of cardiovascular disease pathogenesis in HGPS patients and treatment strategies for this disease. Moreover, we summarize the disease models used in HGPS studies to improve our understanding of the pathological mechanisms of cardiovascular aging in HGPS patients.
Collapse
Affiliation(s)
- Jing Lian
- Medical School of Yan'an University, Yan'an, China
| | - Linfang Du
- Medical School of Yan'an University, Yan'an, China
| | - Yang Li
- School of Basic Medical Sciences, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Lu Yu
- Department of Pathology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | | | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
16
|
Worman HJ, Michaelis S. Prelamin A and ZMPSTE24 in premature and physiological aging. Nucleus 2023; 14:2270345. [PMID: 37885131 PMCID: PMC10730219 DOI: 10.1080/19491034.2023.2270345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/06/2023] [Indexed: 10/28/2023] Open
Abstract
As human longevity increases, understanding the molecular mechanisms that drive aging becomes ever more critical to promote health and prevent age-related disorders. Premature aging disorders or progeroid syndromes can provide critical insights into aspects of physiological aging. A major cause of progeroid syndromes which result from mutations in the genes LMNA and ZMPSTE24 is disruption of the final posttranslational processing step in the production of the nuclear scaffold protein lamin A. LMNA encodes the lamin A precursor, prelamin A and ZMPSTE24 encodes the prelamin A processing enzyme, the zinc metalloprotease ZMPSTE24. Progeroid syndromes resulting from mutations in these genes include the clinically related disorders Hutchinson-Gilford progeria syndrome (HGPS), mandibuloacral dysplasia-type B, and restrictive dermopathy. These diseases have features that overlap with one another and with some aspects of physiological aging, including bone defects resembling osteoporosis and atherosclerosis (the latter primarily in HGPS). The progeroid syndromes have ignited keen interest in the relationship between defective prelamin A processing and its accumulation in normal physiological aging. In this review, we examine the hypothesis that diminished processing of prelamin A by ZMPSTE24 is a driver of physiological aging. We review features a new mouse (LmnaL648R/L648R) that produces solely unprocessed prelamin A and provides an ideal model for examining the effects of its accumulation during aging. We also discuss existing data on the accumulation of prelamin A or its variants in human physiological aging, which call out for further validation and more rigorous experimental approaches to determine if prelamin A contributes to normal aging.
Collapse
Affiliation(s)
- Howard J. Worman
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Susan Michaelis
- Department of Cell Biology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Chen Y, Huang S, Cui Z, Sun X, Tang Y, Zhang H, Chen Z, Jiang R, Zhang W, Li X, Chen J, Liu B, Jiang Y, Wei K, Mao Z. Impaired end joining induces cardiac atrophy in a Hutchinson-Gilford progeria mouse model. Proc Natl Acad Sci U S A 2023; 120:e2309200120. [PMID: 37967221 PMCID: PMC10666128 DOI: 10.1073/pnas.2309200120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/14/2023] [Indexed: 11/17/2023] Open
Abstract
Patients with Hutchinson-Gilford progeria syndrome (HGPS) present with a number of premature aging phenotypes, including DNA damage accumulation, and many of them die of cardiovascular complications. Although vascular pathologies have been reported, whether HGPS patients exhibit cardiac dysfunction and its underlying mechanism is unclear, rendering limited options for treating HGPS-related cardiomyopathy. In this study, we reported a cardiac atrophy phenotype in the LmnaG609G/G609G mice (hereafter, HGPS mice). Using a GFP-based reporter system, we demonstrated that the efficiency of nonhomologous end joining (NHEJ) declined by 50% in HGPS cardiomyocytes in vivo, due to the attenuated interaction between γH2AX and Progerin, the causative factor of HGPS. As a result, genomic instability in cardiomyocytes led to an increase of CHK2 protein level, promoting the LKB1-AMPKα interaction and AMPKα phosphorylation, which further led to the activation of FOXO3A-mediated transcription of atrophy-related genes. Moreover, inhibiting AMPK enlarged cardiomyocyte sizes both in vitro and in vivo. Most importantly, our proof-of-concept study indicated that isoproterenol treatment significantly reduced AMPKα and FOXO3A phosphorylation in the heart, attenuated the atrophy phenotype, and extended the mean lifespan of HGPS mice by ~21%, implying that targeting cardiac atrophy may be an approach to HGPS treatment.
Collapse
Affiliation(s)
- Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Shiqi Huang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Zhen Cui
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Xiaoxiang Sun
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Yansong Tang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Hongjie Zhang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Zhixi Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Rui Jiang
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Weina Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Xue Li
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Jiayu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Baohua Liu
- National Engineering Research Center for Biotechnology (Shenzhen), Carson International Cancer Center, Medical Research Center, Shenzhen University Health Science Center, Shenzhen518055, China
| | - Ying Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Ke Wei
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai200092, China
- Tsingtao Advanced Research Institute, Tongji University, Qingdao266071, China
| |
Collapse
|
18
|
Kong K, Jin M, Zhao C, Qiao H, Chen X, Li B, Rong K, Zhang P, Shan Y, Xu Z, Chang Y, Li H, Zhai Z. Mechanical overloading leads to chondrocyte degeneration and senescence via Zmpste24-mediated nuclear membrane instability. iScience 2023; 26:108119. [PMID: 37965144 PMCID: PMC10641493 DOI: 10.1016/j.isci.2023.108119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 08/28/2023] [Accepted: 09/29/2023] [Indexed: 11/16/2023] Open
Abstract
Patients with OA and varus knees are subject to abnormal mechanical environment and objective of this study was to investigate the molecular mechanisms underlying chondrocyte senescence caused by mechanical overloading and the role of Zmpste24-mediated nuclear membrane instability in varus knees. Finite element analysis showed that anteromedial region of tibial plateau experienced the most mechanical stress in an osteoarthritis patient with a varus knee. Immunohistochemistry exhibited lower Zmpste24 expression and higher expression of senescence marker p21 in the anteromedial region. Animal experiments and cell-stretch models also demonstrated an inverse relationship between Zmpste24 and mechanically induced senescence. Zmpste24 overexpression rescued cartilage degeneration and senescence in vitro by scavenging ROS. In conclusion, anteromedial tibial plateau is exposed to abnormal stress in varus knees, downregulation of Zmpste24, and nuclear membrane stability may explain increased senescence in this region. Zmpste24 and nuclear membrane stability are potential targets for treating osteoarthritis caused by abnormal alignment.
Collapse
Affiliation(s)
- Keyu Kong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Minghao Jin
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chen Zhao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hua Qiao
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuzhuo Chen
- Shanghai Key Laboratory of Stomatology, Department of Oral Surgery, College of Stomatology, Ninth People’s Hospital, Shanghai Research Institute of Stomatology, National Clinical Research Center of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baixing Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kewei Rong
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Pu Zhang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yu Shan
- Suzhou Ninth People’s Hospital, Department of Orthopedics, Suzhou Ninth Hospital affiliated to Soochow University, Suzhou, China
| | - Zhengquan Xu
- Suzhou Municipal Hospital, Department of Orthopedics, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Yongyun Chang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huiwu Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zanjing Zhai
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Yang R, Cao D, Suo J, Zhang L, Mo C, Wang M, Niu N, Yue R, Zou W. Premature aging of skeletal stem/progenitor cells rather than osteoblasts causes bone loss with decreased mechanosensation. Bone Res 2023; 11:35. [PMID: 37407584 DOI: 10.1038/s41413-023-00269-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 04/03/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
A distinct population of skeletal stem/progenitor cells (SSPCs) has been identified that is indispensable for the maintenance and remodeling of the adult skeleton. However, the cell types that are responsible for age-related bone loss and the characteristic changes in these cells during aging remain to be determined. Here, we established models of premature aging by conditional depletion of Zmpste24 (Z24) in mice and found that Prx1-dependent Z24 deletion, but not Osx-dependent Z24 deletion, caused significant bone loss. However, Acan-associated Z24 depletion caused only trabecular bone loss. Single-cell RNA sequencing (scRNA-seq) revealed that two populations of SSPCs, one that differentiates into trabecular bone cells and another that differentiates into cortical bone cells, were significantly decreased in Prx1-Cre; Z24f/f mice. Both premature SSPC populations exhibited apoptotic signaling pathway activation and decreased mechanosensation. Physical exercise reversed the effects of Z24 depletion on cellular apoptosis, extracellular matrix expression and bone mass. This study identified two populations of SSPCs that are responsible for premature aging-related bone loss. The impairment of mechanosensation in Z24-deficient SSPCs provides new insight into how physical exercise can be used to prevent bone aging.
Collapse
Affiliation(s)
- Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Dandan Cao
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jinlong Suo
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lingli Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Chunyang Mo
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Miaomiao Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ningning Niu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
20
|
Kell L, Simon AK, Alsaleh G, Cox LS. The central role of DNA damage in immunosenescence. FRONTIERS IN AGING 2023; 4:1202152. [PMID: 37465119 PMCID: PMC10351018 DOI: 10.3389/fragi.2023.1202152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/22/2023] [Indexed: 07/20/2023]
Abstract
Ageing is the biggest risk factor for the development of multiple chronic diseases as well as increased infection susceptibility and severity of diseases such as influenza and COVID-19. This increased disease risk is linked to changes in immune function during ageing termed immunosenescence. Age-related loss of immune function, particularly in adaptive responses against pathogens and immunosurveillance against cancer, is accompanied by a paradoxical gain of function of some aspects of immunity such as elevated inflammation and increased incidence of autoimmunity. Of the many factors that contribute to immunosenescence, DNA damage is emerging as a key candidate. In this review, we discuss the evidence supporting the hypothesis that DNA damage may be a central driver of immunosenescence through senescence of both immune cells and cells of non-haematopoietic lineages. We explore why DNA damage accumulates during ageing in a major cell type, T cells, and how this may drive age-related immune dysfunction. We further propose that existing immunosenescence interventions may act, at least in part, by mitigating DNA damage and restoring DNA repair processes (which we term "genoprotection"). As such, we propose additional treatments on the basis of their evidence for genoprotection, and further suggest that this approach may provide a viable therapeutic strategy for improving immunity in older people.
Collapse
Affiliation(s)
- Loren Kell
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| | - Anna Katharina Simon
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ghada Alsaleh
- Botnar Institute for Musculoskeletal Sciences, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, United Kingdom
| | - Lynne S. Cox
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Cisneros B, García-Aguirre I, De Ita M, Arrieta-Cruz I, Rosas-Vargas H. Hutchinson-Gilford Progeria Syndrome: Cellular Mechanisms and Therapeutic Perspectives. Arch Med Res 2023; 54:102837. [PMID: 37390702 DOI: 10.1016/j.arcmed.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 05/26/2023] [Accepted: 06/14/2023] [Indexed: 07/02/2023]
Abstract
In humans, aging is characterized by a gradual decline of physical and psychological functions, with the concomitant onset of chronic-degenerative diseases, which ultimately lead to death. The study of Hutchinson-Gilford progeria syndrome (HGPS), a premature aging disorder that recapitulates several features of natural aging, has provided important insights into deciphering the aging process. The genetic origin of HGPS is a de novo point mutation in the LMNA gene that drives the synthesis of progerin, mutant version of lamin A. Progerin is aberrantly anchored to the nuclear envelope disrupting a plethora of molecular processes; nonetheless, how progerin exerts a cascade of deleterious alterations at the cellular and systemic levels is not fully understood. Over the past decade, the use of different cellular and animal models for HGPS has allowed the identification of the molecular mechanisms underlying HGPS, paving the way towards the development of therapeutic treatments against the disease. In this review, we present an updated overview of the biology of HGPS, including its clinical features, description of key cellular processes affected by progerin (nuclear morphology and function, nucleolar activity, mitochondrial function, protein nucleocytoplasmic trafficking and telomere homeostasis), as well as discussion of the therapeutic strategies under development.
Collapse
Affiliation(s)
- Bulmaro Cisneros
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico
| | - Ian García-Aguirre
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico; Bioengineering Department, School of Engineering and Sciences, Tecnológico de Monterrey, Mexico City, Mexico
| | - Marlon De Ita
- Genetics and Molecular Biology Department, Research and Advanced Studies Center, National Polytechnical Institute, Mexico City, Mexico; Medical Research Unit in Human Genetics, Pediatrics Hospital, 21st Century National Medical Center, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Isabel Arrieta-Cruz
- Basic Research Department, Research Direction, National Institute of Geriatrics, Ministry of Health, Mexico City, Mexico
| | - Haydeé Rosas-Vargas
- Medical Research Unit in Human Genetics, Pediatrics Hospital, 21st Century National Medical Center, Instituto Mexicano del Seguro Social, Mexico City, Mexico.
| |
Collapse
|
22
|
Porter J, Ward LC, Nguo K, Davidson Z, Gibson S, Prentice R, Neuhouser ML, Truby H. Development and validation of new predictive equations for the resting metabolic rate of older adults aged ≥65 y. Am J Clin Nutr 2023; 117:1164-1173. [PMID: 37054885 PMCID: PMC10447471 DOI: 10.1016/j.ajcnut.2023.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 03/23/2023] [Accepted: 04/10/2023] [Indexed: 04/15/2023] Open
Abstract
BACKGROUND The aging process alters the resting metabolic rate (RMR), but it still accounts for 50%-70% of the total energy needs. The rising proportion of older adults, especially those over 80 y of age, underpins the need for a simple, rapid method to estimate the energy needs of older adults. OBJECTIVES This research aimed to generate and validate new RMR equations specifically for older adults and to report their performance and accuracy. METHODS Data were sourced to form an international dataset of adults aged ≥65 y (n = 1686, 38.5% male) where RMR was measured using the reference method of indirect calorimetry. Multiple regression was used to predict RMR from age (y), sex, weight (kg), and height (cm). Double cross-validation in a randomized, sex-stratified, age-matched 50:50 split and leave one out cross-validation were performed. The newly generated prediction equations were compared with the existing commonly used equations. RESULTS The new prediction equation for males and females aged ≥65 y had an overall improved performance, albeit marginally, when compared with the existing equations. It is described as follows: RMR (kJ/d) = 31.524 × W (kg) + 25.851 × H (cm) - 24.432 × Age (y) + 486.268 × Sex (M = 1, F = 0) + 530.557. Equations stratified by age (65-79.9 y and >80 y) and sex are also provided. The newly created equation estimates RMR within a population mean prediction bias of ∼50 kJ/d (∼1%) for those aged ≥65 y. Accuracy was reduced in adults aged ≥80 y (∼100 kJ/d, ∼2%) but was still within the clinically acceptable range for both males and females. Limits of agreement indicated a poorer performance at an individual level with 1.96-SD limits of approximately ±25%. CONCLUSIONS The new equations, using simple measures of weight, height, and age, improved the accuracy in the prediction of RMR in populations in clinical practice. However, no equation performs optimally at the individual level.
Collapse
Affiliation(s)
- Judi Porter
- Institute of Physical Activity and Nutrition, School of Exercise and Nutrition Science, Deakin University, Burwood, Melbourne, Victoria, Australia.
| | - Leigh C Ward
- Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Kay Nguo
- Department of Nutrition, Dietetics and Food, Monash University, Notting Hill, Melbourne, Victoria, Australia
| | - Zoe Davidson
- Department of Nutrition, Dietetics and Food, Monash University, Notting Hill, Melbourne, Victoria, Australia
| | - Simone Gibson
- Department of Nutrition, Dietetics and Food, Monash University, Notting Hill, Melbourne, Victoria, Australia
| | - Ross Prentice
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Marian L Neuhouser
- Fred Hutchinson Cancer Research Center and School of Public Health and Community Medicine, University of Washington, Seattle, Washington, USA
| | - Helen Truby
- School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
23
|
Joudeh LA, DiCintio AJ, Ries MR, Gasperson AS, Griffin KE, Robbins VP, Bonner M, Nolan S, Black E, Waldman AS. Corruption of DNA end-joining in mammalian chromosomes by progerin expression. DNA Repair (Amst) 2023; 126:103491. [PMID: 37018982 PMCID: PMC10133198 DOI: 10.1016/j.dnarep.2023.103491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/23/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023]
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare genetic condition characterized by features of accelerated aging and a life expectancy of about 14 years. HGPS is commonly caused by a point mutation in the LMNA gene which codes for lamin A, an essential component of the nuclear lamina. The HGPS mutation alters splicing of the LMNA transcript, leading to a truncated, farnesylated form of lamin A termed "progerin." Progerin is also produced in small amounts in healthy individuals by alternative splicing of RNA and has been implicated in normal aging. HGPS is associated with an accumulation of genomic DNA double-strand breaks (DSBs), suggesting alteration of DNA repair. DSB repair normally occurs by either homologous recombination (HR), an accurate, templated form of repair, or by nonhomologous end-joining (NHEJ), a non-templated rejoining of DNA ends that can be error-prone; however a good portion of NHEJ events occurs precisely with no alteration to joined sequences. Previously, we reported that over-expression of progerin correlated with increased NHEJ relative to HR. We now report on progerin's impact on the nature of DNA end-joining. We used a model system involving a DNA end-joining reporter substrate integrated into the genome of cultured thymidine kinase-deficient mouse fibroblasts. Some cells were engineered to express progerin. Two closely spaced DSBs were induced in the integrated substrate through expression of endonuclease I-SceI, and DSB repair events were recovered through selection for thymidine kinase function. DNA sequencing revealed that progerin expression correlated with a significant shift away from precise end-joining between the two I-SceI sites and toward imprecise end-joining. Additional experiments revealed that progerin did not reduce HR fidelity. Our work suggests that progerin suppresses interactions between complementary sequences at DNA termini, thereby shifting DSB repair toward low-fidelity DNA end-joining and perhaps contributing to accelerated and normal aging through compromised genome stability.
Collapse
Affiliation(s)
- Liza A Joudeh
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Alannah J DiCintio
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Madeline R Ries
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Andrew S Gasperson
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Kennedy E Griffin
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Victoria P Robbins
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Makenzie Bonner
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Sarah Nolan
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Emma Black
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Alan S Waldman
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA.
| |
Collapse
|
24
|
Bao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, Wang S, Wang X, Wang X, Wang YJ, Wang Y, Wong CCL, Xiang AP, Xiao Y, Xie Z, Xu D, Ye J, Yue R, Zhang C, Zhang H, Zhang L, Zhang W, Zhang Y, Zhang YW, Zhang Z, Zhao T, Zhao Y, Zhu D, Zou W, Pei G, Liu GH. Biomarkers of aging. SCIENCE CHINA. LIFE SCIENCES 2023; 66:893-1066. [PMID: 37076725 PMCID: PMC10115486 DOI: 10.1007/s11427-023-2305-0] [Citation(s) in RCA: 108] [Impact Index Per Article: 108.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023]
Abstract
Aging biomarkers are a combination of biological parameters to (i) assess age-related changes, (ii) track the physiological aging process, and (iii) predict the transition into a pathological status. Although a broad spectrum of aging biomarkers has been developed, their potential uses and limitations remain poorly characterized. An immediate goal of biomarkers is to help us answer the following three fundamental questions in aging research: How old are we? Why do we get old? And how can we age slower? This review aims to address this need. Here, we summarize our current knowledge of biomarkers developed for cellular, organ, and organismal levels of aging, comprising six pillars: physiological characteristics, medical imaging, histological features, cellular alterations, molecular changes, and secretory factors. To fulfill all these requisites, we propose that aging biomarkers should qualify for being specific, systemic, and clinically relevant.
Collapse
Affiliation(s)
- Hainan Bao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Jiani Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Min Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yingjie Ding
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junlin Feng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mengmeng Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Chuting He
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Yujuan Jia
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Haiping Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Ying Jing
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyi Li
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Qinhao Liang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jianwei Lv
- School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jingyi Ma
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kehang Mao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China
| | - Jiawei Nie
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Siyuan Wang
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xuan Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuhan Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Rimo Wu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Kai Xia
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fu-Hui Xiao
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Xu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Haoteng Yan
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuanxin Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Le Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiwei Zhang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China
| | - Wenwan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qingchen Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China.
| | - Zhongwei Cao
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou, 510000, China.
| | - Hou-Zao Chen
- Department of Biochemistryand Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China.
| | - Weimin Ci
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Feng Gao
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| | - Qing-Peng Kong
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Xin Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Baohua Liu
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518060, China.
| | - Feng Liu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South Unversity, Changsha, 410011, China.
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China.
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, 300000, China.
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Institute of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
| | - Yong Liu
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Shuai Ma
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jing Nie
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yaojin Peng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Center for Aging and Cancer, Hainan Medical University, Haikou, 571199, China.
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China.
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China.
| | - Si Wang
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Xia Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Xiaoning Wang
- Institute of Geriatrics, The second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Yunfang Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Catherine C L Wong
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China.
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, 100101, China.
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Cuntai Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China.
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Liang Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yong Zhang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Zhuohua Zhang
- Key Laboratory of Molecular Precision Medicine of Hunan Province and Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Dahai Zhu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Gang Pei
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, 200070, China.
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
25
|
Kristiani L, Kim Y. The Interplay between Oxidative Stress and the Nuclear Lamina Contributes to Laminopathies and Age-Related Diseases. Cells 2023; 12:cells12091234. [PMID: 37174634 PMCID: PMC10177617 DOI: 10.3390/cells12091234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Oxidative stress is a physiological condition that arises when there is an imbalance between the production of reactive oxygen species (ROS) and the ability of cells to neutralize them. ROS can damage cellular macromolecules, including lipids, proteins, and DNA, leading to cellular senescence and physiological aging. The nuclear lamina (NL) is a meshwork of intermediate filaments that provides structural support to the nucleus and plays crucial roles in various nuclear functions, such as DNA replication and transcription. Emerging evidence suggests that oxidative stress disrupts the integrity and function of the NL, leading to dysregulation of gene expression, DNA damage, and cellular senescence. This review highlights the current understanding of the interplay between oxidative stress and the NL, along with its implications for human health. Specifically, elucidation of the mechanisms underlying the interplay between oxidative stress and the NL is essential for the development of effective treatments for laminopathies and age-related diseases.
Collapse
Affiliation(s)
- Lidya Kristiani
- Department of Biomedicine, School of Life Science, Indonesia International Institute for Life Science, Jakarta 13210, Indonesia
| | - Youngjo Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bioscience, Soonchunhyang University, Cheonan 31151, Republic of Korea
| |
Collapse
|
26
|
Cho SJ, Pronko A, Yang J, Stout-Delgado H. Impact of Senolytic Treatment on Gene Expression in Aged Lung. Int J Mol Sci 2023; 24:7628. [PMID: 37108795 PMCID: PMC10145650 DOI: 10.3390/ijms24087628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Cellular senescence plays a key role in mediating tissue remodeling and modulation of host responses to pathogenic stimuli. Our current study was designed to gain a better understanding of the impact of short-term senolytic treatment or inflammatory stimulation on lung senescence. The results of our study demonstrate that short term treatment of aged adult mice (20 months of age) with senolytics, quercetin, and dasatinib decreases p16 and p21 expression in lung tissue. Short-term treatment with senolytics also significantly improved the expression of genes associated with genomic instability, telomere attrition, mitochondrial dysfunction, DNA binding, and the inflammatory response. In contrast, in response to low-dose LPS administration, there was increased expression of genes associated with genomic instability, mitochondrial dysfunction, and heightened inflammatory responses in young adult murine lung (3 months of age). Taken together, the results of our current study illustrate the efficacy of senolytic treatment on modulating responses in aged lung and the potential role of chronic low dose inflammation on senescence induction in the lung.
Collapse
Affiliation(s)
| | | | | | - Heather Stout-Delgado
- Pulmonary and Critical Medicine, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
27
|
Abutaleb NO, Atchison L, Choi L, Bedapudi A, Shores K, Gete Y, Cao K, Truskey GA. Lonafarnib and everolimus reduce pathology in iPSC-derived tissue engineered blood vessel model of Hutchinson-Gilford Progeria Syndrome. Sci Rep 2023; 13:5032. [PMID: 36977745 PMCID: PMC10050176 DOI: 10.1038/s41598-023-32035-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, fatal genetic disease that accelerates atherosclerosis. With a limited pool of HGPS patients, clinical trials face unique challenges and require reliable preclinical testing. We previously reported a 3D tissue engineered blood vessel (TEBV) microphysiological system fabricated with iPSC-derived vascular cells from HGPS patients. HGPS TEBVs exhibit features of HGPS atherosclerosis including loss of smooth muscle cells, reduced vasoactivity, excess extracellular matrix (ECM) deposition, inflammatory marker expression, and calcification. We tested the effects of HGPS therapeutics Lonafarnib and Everolimus separately and together, currently in Phase I/II clinical trial, on HGPS TEBVs. Everolimus decreased reactive oxygen species levels, increased proliferation, reduced DNA damage in HGPS vascular cells, and improved vasoconstriction in HGPS TEBVs. Lonafarnib improved shear stress response of HGPS iPSC-derived endothelial cells (viECs) and reduced ECM deposition, inflammation, and calcification in HGPS TEBVs. Combination treatment with Lonafarnib and Everolimus produced additional benefits such as improved endothelial and smooth muscle marker expression and reduced apoptosis, as well as increased TEBV vasoconstriction and vasodilation. These results suggest that a combined trial of both drugs may provide cardiovascular benefits beyond Lonafarnib, if the Everolimus dose can be tolerated.
Collapse
Affiliation(s)
- Nadia O Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leigh Atchison
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Leandro Choi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Akhil Bedapudi
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Kevin Shores
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Yantenew Gete
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
28
|
Elevated Levels of Lamin A Promote HR and NHEJ-Mediated Repair Mechanisms in High-Grade Ovarian Serous Carcinoma Cell Line. Cells 2023; 12:cells12050757. [PMID: 36899893 PMCID: PMC10001195 DOI: 10.3390/cells12050757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/15/2023] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Extensive research for the last two decades has significantly contributed to understanding the roles of lamins in the maintenance of nuclear architecture and genome organization which is drastically modified in neoplasia. It must be emphasized that alteration in lamin A/C expression and distribution is a consistent event during tumorigenesis of almost all tissues of human bodies. One of the important signatures of a cancer cell is its inability to repair DNA damage which befalls several genomic events that transform the cells to be sensitive to chemotherapeutic agents. This genomic and chromosomal instability is the most common feature found in cases of high-grade ovarian serous carcinoma. Here, we report elevated levels of lamins in OVCAR3 cells (high-grade ovarian serous carcinoma cell line) in comparison to IOSE (immortalised ovarian surface epithelial cells) and, consequently, altered damage repair machinery in OVCAR3. We have analysed the changes in global gene expression as a sequel to DNA damage induced by etoposide in ovarian carcinoma where lamin A is particularly elevated in expression and reported some differentially expressed genes associated with pathways conferring cellular proliferation and chemoresistance. We hereby establish the role of elevated lamin A in neoplastic transformation in the context of high-grade ovarian serous cancer through a combination of HR and NHEJ mechanisms.
Collapse
|
29
|
Zhang J, Wang S, Liu B. New Insights into the Genetics and Epigenetics of Aging Plasticity. Genes (Basel) 2023; 14:329. [PMID: 36833255 PMCID: PMC9956228 DOI: 10.3390/genes14020329] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/14/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
Biological aging is characterized by irreversible cell cycle blockade, a decreased capacity for tissue regeneration, and an increased risk of age-related diseases and mortality. A variety of genetic and epigenetic factors regulate aging, including the abnormal expression of aging-related genes, increased DNA methylation levels, altered histone modifications, and unbalanced protein translation homeostasis. The epitranscriptome is also closely associated with aging. Aging is regulated by both genetic and epigenetic factors, with significant variability, heterogeneity, and plasticity. Understanding the complex genetic and epigenetic mechanisms of aging will aid the identification of aging-related markers, which may in turn aid the development of effective interventions against this process. This review summarizes the latest research in the field of aging from a genetic and epigenetic perspective. We analyze the relationships between aging-related genes, examine the possibility of reversing the aging process by altering epigenetic age.
Collapse
Affiliation(s)
- Jie Zhang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University, Shenzhen 518000, China
| | - Shixiao Wang
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University, Shenzhen 518000, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SKL-SAI), School of Basic Medical Sciences, Shenzhen University, Shenzhen 518000, China
- Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Medical School, Lihu Campus, Shenzhen University, Shenzhen 518000, China
| |
Collapse
|
30
|
Fragoso-Luna A, Askjaer P. The Nuclear Envelope in Ageing and Progeria. Subcell Biochem 2023; 102:53-75. [PMID: 36600129 DOI: 10.1007/978-3-031-21410-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Development from embryo to adult, organismal homeostasis and ageing are consecutive processes that rely on several functions of the nuclear envelope (NE). The NE compartmentalises the eukaryotic cells and provides physical stability to the genetic material in the nucleus. It provides spatiotemporal regulation of gene expression by controlling nuclear import and hence access of transcription factors to target genes as well as organisation of the genome into open and closed compartments. In addition, positioning of chromatin relative to the NE is important for DNA replication and repair and thereby also for genome stability. We discuss here the relevance of the NE in two classes of age-related human diseases. Firstly, we focus on the progeria syndromes Hutchinson-Gilford (HGPS) and Nestor-Guillermo (NGPS), which are caused by mutations in the LMNA and BANF1 genes, respectively. Both genes encode ubiquitously expressed components of the nuclear lamina that underlines the nuclear membranes. HGPS and NGPS patients manifest symptoms of accelerated ageing and cells from affected individuals show similar defects as cells from healthy old donors, including signs of increased DNA damage and epigenetic alternations. Secondly, we describe how several age-related neurodegenerative diseases, such as amyotrophic lateral sclerosis and Huntington's disease, are related with defects in nucleocytoplasmic transport. A common feature of this class of diseases is the accumulation of nuclear pore proteins and other transport factors in inclusions. Importantly, genetic manipulations of the nucleocytoplasmic transport machinery can alleviate disease-related phenotypes in cell and animal models, paving the way for potential therapeutic interventions.
Collapse
Affiliation(s)
- Adrián Fragoso-Luna
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Sevilla, Spain
| | - Peter Askjaer
- Andalusian Centre for Developmental Biology, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Pablo de Olavide, Sevilla, Spain.
| |
Collapse
|
31
|
González-Amor M, Dorado B, Andrés V. Emerging roles of interferon-stimulated gene-15 in age-related telomere attrition, the DNA damage response, and cardiovascular disease. Front Cell Dev Biol 2023; 11:1128594. [PMID: 37025175 PMCID: PMC10071045 DOI: 10.3389/fcell.2023.1128594] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/08/2023] [Indexed: 04/08/2023] Open
Abstract
Population aging and age-related cardiovascular disease (CVD) are becoming increasingly prevalent worldwide, generating a huge medical and socioeconomic burden. The complex regulation of aging and CVD and the interaction between these processes are crucially dependent on cellular stress responses. Interferon-stimulated gene-15 (ISG15) encodes a ubiquitin-like protein expressed in many vertebrate cell types that can be found both free and conjugated to lysine residues of target proteins via a post-translational process termed ISGylation. Deconjugation of ISG15 (deISGylation) is catalyzed by the ubiquitin-specific peptidase 18 (USP18). The ISG15 pathway has mostly been studied in the context of viral and bacterial infections and in cancer. This minireview summarizes current knowledge on the role of ISG15 in age-related telomere shortening, genomic instability, and DNA damage accumulation, as well as in hypertension, diabetes, and obesity, major CVD risk factors prevalent in the elderly population.
Collapse
Affiliation(s)
- María González-Amor
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Molecular and Genetic Cardiovascular Pathophysiology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Beatriz Dorado
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Molecular and Genetic Cardiovascular Pathophysiology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Vicente Andrés
- CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Molecular and Genetic Cardiovascular Pathophysiology Laboratory, Novel Mechanisms of Atherosclerosis Program, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- *Correspondence: Vicente Andrés,
| |
Collapse
|
32
|
Kirkland NJ, Skalak SH, Whitehead AJ, Hocker JD, Beri P, Vogler G, Hum B, Wang M, Lakatta EG, Ren B, Bodmer R, Engler AJ. Age-dependent Lamin changes induce cardiac dysfunction via dysregulation of cardiac transcriptional programs. NATURE AGING 2023; 3:17-33. [PMID: 36845078 PMCID: PMC9956937 DOI: 10.1038/s43587-022-00323-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
As we age, structural changes contribute to progressive decline in organ function, which in the heart act through poorly characterized mechanisms. Taking advantage of the short lifespan and conserved cardiac proteome of the fruit fly, we found that cardiomyocytes exhibit progressive loss of Lamin C (mammalian Lamin A/C homologue) with age, coincident with decreasing nuclear size and increasing nuclear stiffness. Premature genetic reduction of Lamin C phenocopies aging's effects on the nucleus, and subsequently decreases heart contractility and sarcomere organization. Surprisingly, Lamin C reduction downregulates myogenic transcription factors and cytoskeletal regulators, possibly via reduced chromatin accessibility. Subsequently, we find a role for cardiac transcription factors in regulating adult heart contractility and show that maintenance of Lamin C, and cardiac transcription factor expression, prevents age-dependent cardiac decline. Our findings are conserved in aged non-human primates and mice, demonstrating that age-dependent nuclear remodeling is a major mechanism contributing to cardiac dysfunction.
Collapse
Affiliation(s)
- Natalie J. Kirkland
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - Scott H. Skalak
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - Alexander J. Whitehead
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - James D. Hocker
- Cell and Molecular Medicine, University California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Program, University California San Diego; La Jolla, CA, USA 92093
| | - Pranjali Beri
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| | - Geo Vogler
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute; La Jolla, CA, USA 92037
| | - Bill Hum
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute; La Jolla, CA, USA 92037
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA 21224
| | - Edward G. Lakatta
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA 21224
| | - Bing Ren
- Cell and Molecular Medicine, University California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Program, University California San Diego; La Jolla, CA, USA 92093
- Ludwig Institute for Cancer Research; La Jolla, CA, USA 92037
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute; La Jolla, CA, USA 92037
| | - Adam J. Engler
- Department of Bioengineering, University California San Diego; La Jolla, CA, USA 92093
- Biomedical Sciences Program, University California San Diego; La Jolla, CA, USA 92093
- Sanford Consortium for Regenerative Medicine; La Jolla, CA, USA 92037
| |
Collapse
|
33
|
Hu Q, Zhang N, Sui T, Li G, Wang Z, Liu M, Zhu X, Huang B, Lu J, Li Z, Zhang Y. Anti-hsa-miR-59 alleviates premature senescence associated with Hutchinson-Gilford progeria syndrome in mice. EMBO J 2022; 42:e110937. [PMID: 36382717 PMCID: PMC9811625 DOI: 10.15252/embj.2022110937] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/17/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a lethal premature aging disorder without an effective therapeutic regimen. Because of their targetability and influence on gene expression, microRNAs (miRNAs) are attractive therapeutic tools to treat diseases. Here we identified that hsa-miR-59 (miR-59) was markedly upregulated in HGPS patient cells and in multiple tissues of an HGPS mouse model (LmnaG609G/G609G ), which disturbed the interaction between RNAPII and TFIIH, resulting in abnormal expression of cell cycle genes by targeting high-mobility group A family HMGA1 and HMGA2. Functional inhibition of miR-59 alleviated the cellular senescence phenotype of HGPS cells. Treatment with AAV9-mediated anti-miR-59 reduced fibrosis in the quadriceps muscle, heart, and aorta, suppressed epidermal thinning and dermal fat loss, and yielded a 25.5% increase in longevity of LmnaG609G/G609G mice. These results identify a new strategy for the treatment of HGPS and provide insight into the etiology of HGPS disease.
Collapse
Affiliation(s)
- Qianying Hu
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Na Zhang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Tingting Sui
- The Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal ScienceJilin UniversityChangchunChina
| | - Guanlin Li
- The Institute of Genetics and CytologyNortheast Normal UniversityChangchunChina
| | - Zhiyao Wang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Mingyue Liu
- The Institute of Genetics and CytologyNortheast Normal UniversityChangchunChina
| | - Xiaojuan Zhu
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Baiqu Huang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| | - Jun Lu
- The Institute of Genetics and CytologyNortheast Normal UniversityChangchunChina
| | - Zhanjun Li
- The Key Laboratory of Zoonosis Research, Ministry of Education, College of Animal ScienceJilin UniversityChangchunChina
| | - Yu Zhang
- The Key Laboratory of Molecular Epigenetics of the Ministry of Education (MOE)Northeast Normal UniversityChangchunChina
| |
Collapse
|
34
|
Hu C, Zhou A, Hu X, Xiang Y, Huang M, Huang J, Yang D, Tang Y. LMNA Reduced Acquired Resistance to Erlotinib in NSCLC by Reversing the Epithelial-Mesenchymal Transition via the FGFR/MAPK/c-fos Signaling Pathway. Int J Mol Sci 2022; 23:13237. [PMID: 36362025 PMCID: PMC9658955 DOI: 10.3390/ijms232113237] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/17/2022] [Accepted: 10/25/2022] [Indexed: 09/17/2023] Open
Abstract
For patients exhibiting non-small-cell lung cancer (NSCLC) with activating epidermal growth factor receptor (EGFR) mutations, epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) are a first-line treatment. However, most patients who initially responded to EGFR-TKIs eventually developed acquired resistance, limiting the effectiveness of therapy. It has long been known that epithelial-mesenchymal transition (EMT) leads to acquired resistance to EGFR-TKIs in NSCLC. However, the mechanisms underlying the resistance dependent on EMT are unknown. This research aimed to reveal the effects of LMNA in the regulation of acquired resistance to erlotinib by EMT in NSCLC. The acquired erlotinib-resistant cells (HCC827/ER) were induced by gradual increase of concentrations of erlotinib in erlotinib-sensitive HCC827 cells. RNA sequencing and bioinformatics analysis were performed to uncover the involvement of LMNA in the EMT process that induced acquired resistance to erlotinib. The effect of LMNA on cell proliferation and migration was measured by clone-formation, wound-healing, and transwell assays, respectively. The EMT-related protein, nuclear shape and volume, and cytoskeleton changes were examined by immunofluorescence. Western blot was used to identify the underlying molecular mechanism of LMNA regulation of EMT. HCC827/ER cells with acquired resistance to erlotinib underwent EMT and exhibited lower LMNA expression compared to parental sensitive cells. LMNA negatively regulated the expression of EMT markers; HCC827/ER cells showed a significant up-regulation of mesenchymal markers, such as CDH2, SNAI2, VIM, ZEB1, and TWIST1. The overexpression of LMNA in HCC827/ER cells significantly inhibited EMT and cell proliferation, and this inhibitory effect of LMNA was enhanced in the presence of 2.5 μM erlotinib. Furthermore, a decrease in LMNA expression resulted in a higher nuclear deformability and cytoskeletal changes. In HCC827/ER cells, AKT, FGFR, ERK1/2, and c-fos phosphorylation levels were higher than those in HCC827 cells; Furthermore, overexpression of LMNA in HCC827/ER cells reduced the phosphorylation of AKT, ERK1/2, c-fos, and FGFR. In conclusion, our findings first demonstrated that downregulation of LMNA promotes acquired EGFR-TKI resistance in NSCLC with EGFR mutations by EMT. LMNA inhibits cell proliferation and migration of erlotinib-resistant cells via inhibition of the FGFR/MAPK/c-fos signaling pathway. These findings indicated LMNA as a driver of acquired resistance to erlotinib and provided important information about the development of resistance to erlotinib treatment in NSCLC patients with EGFR mutations.
Collapse
Affiliation(s)
- Chunsheng Hu
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing 402160, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Anting Zhou
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Xin Hu
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Yu Xiang
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Mengjun Huang
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Jiuhong Huang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Donglin Yang
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing University of Arts and Sciences, Chongqing 402160, China
| | - Yan Tang
- College of Pharmacy (International Academy of Targeted Therapeutics and Innovation), Chongqing University of Arts and Sciences, Chongqing 402160, China
- National & Local Joint Engineering Research Center of Targeted and Innovative Therapeutics, Chongqing University of Arts and Sciences, Chongqing 402160, China
| |
Collapse
|
35
|
Primmer SR, Liao CY, Kummert OMP, Kennedy BK. Lamin A to Z in normal aging. Aging (Albany NY) 2022; 14:8150-8166. [PMID: 36260869 DOI: 10.18632/aging.204342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/31/2022] [Indexed: 11/25/2022]
Abstract
Almost since the discovery that mutations in the LMNA gene, encoding the nuclear structure components lamin A and C, lead to Hutchinson-Gilford progeria syndrome, people have speculated that lamins may have a role in normal aging. The most common HPGS mutation creates a splice variant of lamin A, progerin, which promotes accelerated aging pathology. While some evidence exists that progerin accumulates with normal aging, an increasing body of work indicates that prelamin A, a precursor of lamin A prior to C-terminal proteolytic processing, accumulates with age and may be a driver of normal aging. Prelamin A shares properties with progerin and is also linked to a rare progeroid disease, restrictive dermopathy. Here, we describe mechanisms underlying changes in prelamin A with aging and lay out the case that this unprocessed protein impacts normative aging. This is important since intervention strategies can be developed to modify this pathway as a means to extend healthspan and lifespan.
Collapse
Affiliation(s)
| | - Chen-Yu Liao
- The Buck Institute for Research on Aging, Novato, CA 94945, USA
| | | | - Brian K Kennedy
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Centre for Healthy Longevity, National University Health System, Singapore.,Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
36
|
Maynard S, Hall A, Galanos P, Rizza S, Yamamoto T, Gram H, Munk SHN, Shoaib M, Sørensen CS, Bohr V, Lerdrup M, Maya-Mendoza A, Bartek J. Lamin A/C impairments cause mitochondrial dysfunction by attenuating PGC1α and the NAMPT-NAD+ pathway. Nucleic Acids Res 2022; 50:9948-9965. [PMID: 36099415 PMCID: PMC9508839 DOI: 10.1093/nar/gkac741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 07/30/2022] [Accepted: 08/30/2022] [Indexed: 11/14/2022] Open
Abstract
Mutations in the lamin A/C gene (LMNA) cause laminopathies such as the premature aging Hutchinson Gilford progeria syndrome (HGPS) and altered lamin A/C levels are found in diverse malignancies. The underlying lamin-associated mechanisms remain poorly understood. Here we report that lamin A/C-null mouse embryo fibroblasts (Lmna-/- MEFs) and human progerin-expressing HGPS fibroblasts both display reduced NAD+ levels, unstable mitochondrial DNA and attenuated bioenergetics. This mitochondrial dysfunction is associated with reduced chromatin recruitment (Lmna-/- MEFs) or low levels (HGPS) of PGC1α, the key transcription factor for mitochondrial homeostasis. Lmna-/- MEFs showed reduced expression of the NAD+-biosynthesis enzyme NAMPT and attenuated activity of the NAD+-dependent deacetylase SIRT1. We find high PARylation in lamin A/C-aberrant cells, further decreasing the NAD+ pool and consistent with impaired DNA base excision repair in both cell models, a condition that fuels DNA damage-induced PARylation under oxidative stress. Further, ATAC-sequencing revealed a substantially altered chromatin landscape in Lmna-/- MEFs, including aberrantly reduced accessibility at the Nampt gene promoter. Thus, we identified a new role of lamin A/C as a key modulator of mitochondrial function through impairments of PGC1α and the NAMPT-NAD+ pathway, with broader implications for the aging process.
Collapse
Affiliation(s)
- Scott Maynard
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Arnaldur Hall
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | | | - Salvatore Rizza
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | - Tatsuro Yamamoto
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
| | | | | | - Muhammad Shoaib
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Claus Storgaard Sørensen
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Vilhelm A Bohr
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mads Lerdrup
- The DNRF Center for Chromosome Stability, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | - Jiri Bartek
- Danish Cancer Society Research Center, DK-2100 Copenhagen, Denmark
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Life Laboratory, Karolinska Institute, SE-17177 Stockholm, Sweden
| |
Collapse
|
37
|
Lin H, Mensch J, Haschke M, Jäger K, Köttgen B, Dernedde J, Orsó E, Walter M. Establishment and Characterization of hTERT Immortalized Hutchinson–Gilford Progeria Fibroblast Cell Lines. Cells 2022; 11:cells11182784. [PMID: 36139359 PMCID: PMC9497314 DOI: 10.3390/cells11182784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/22/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare premature aging syndrome caused by a dominant mutation in the LMNA gene. Previous research has shown that the ectopic expression of the catalytic subunit of telomerase (hTERT) can elongate the telomeres of the patients’ fibroblasts. Here, we established five immortalized HGP fibroblast cell lines using retroviral infection with the catalytic subunit of hTERT. Immortalization enhanced the proliferative life span by at least 50 population doublings (PDs). The number of cells with typical senescence signs was reduced by 63 + 17%. Furthermore, the growth increase and phenotype improvement occurred with a lag phase of 50–100 days and was not dependent on the degree of telomere elongation. The initial telomeric stabilization after hTERT infection and relatively low amounts of hTERT mRNA were sufficient for the phenotype improvement but the retroviral infection procedure was associated with transient cell stress. Our data have implications for therapeutic strategies in HGP and other premature aging syndromes.
Collapse
Affiliation(s)
- Haihuan Lin
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
| | - Juliane Mensch
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Maria Haschke
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Kathrin Jäger
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Brigitte Köttgen
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
| | - Jens Dernedde
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
| | - Evelyn Orsó
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Michael Walter
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, 13353 Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Rostock University Medical Center, 18057 Rostock, Germany
- Correspondence:
| |
Collapse
|
38
|
Impact of MnTBAP and Baricitinib Treatment on Hutchinson–Gilford Progeria Fibroblasts. Pharmaceuticals (Basel) 2022; 15:ph15080945. [PMID: 36015093 PMCID: PMC9415676 DOI: 10.3390/ph15080945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/11/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare premature aging disease. It is caused by a mutation in the LMNA gene, which results in a 50-amino-acid truncation of prelamin A. The resultant truncated prelamin A (progerin) lacks the cleavage site for the zinc-metallopeptidase ZMPSTE24. Progerin is permanently farnesylated, carboxymethylated, and strongly anchored to the nuclear envelope. This leads to abnormalities, such as altered nuclear shape, mitochondrial dysfunction, and inflammation. HGPS patients display symptoms of physiological aging, including atherosclerosis, alopecia, lipodystrophy, and arthritis. Currently, no cure for HGPS exists. Here we focus on a drug combination consisting of the superoxide dismutase mimetic MnTBAP and JAK1/2 inhibitor baricitinib (Bar) to restore phenotypic alterations in HGPS fibroblasts. Treating HGPS fibroblasts with the MnTBAP/Bar combination improved mitochondrial functions and sustained Bar’s positive effects on reducing progerin and pro-inflammatory factor levels. Collectively, MnTBAP/Bar combination treatment ameliorates the aberrant phenotype of HGPS fibroblasts and is a potential treatment strategy for patients with HGPS.
Collapse
|
39
|
Jiang B, Wu X, Meng F, Si L, Cao S, Dong Y, Sun H, Lv M, Xu H, Bai N, Guo Q, Song X, Yu Y, Guo W, Yi F, Zhou T, Li X, Feng Y, Wang Z, Zhang D, Guan Y, Ma M, Liu J, Li X, Zhao W, Liu B, Finkel T, Cao L. Progerin modulates the IGF-1R/Akt signaling involved in aging. SCIENCE ADVANCES 2022; 8:eabo0322. [PMID: 35857466 PMCID: PMC9269893 DOI: 10.1126/sciadv.abo0322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Progerin, a product of LMNA mutation, leads to multiple nuclear abnormalities in patients with Hutchinson-Gilford progeria syndrome (HGPS), a devastating premature aging disorder. Progerin also accumulates during physiological aging. Here, we demonstrate that impaired insulin-like growth factor 1 receptor (IGF-1R)/Akt signaling pathway results in severe growth retardation and premature aging in Zmpste24-/- mice, a mouse model of progeria. Mechanistically, progerin mislocalizes outside of the nucleus, interacts with the IGF-1R, and down-regulates its expression, leading to inhibited mitochondrial respiration, retarded cell growth, and accelerated cellular senescence. Pharmacological treatment with the PTEN (phosphatase and tensin homolog deleted on chromosome 10) inhibitor bpV (HOpic) increases Akt activity and improves multiple abnormalities in Zmpste24-deficient mice. These findings provide previously unidentified insights into the role of progerin in regulating the IGF-1R/Akt signaling in HGPS and might be useful for treating LMNA-associated progeroid disorders.
Collapse
Affiliation(s)
- Bo Jiang
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
- Institute of Health Sciences, China Medical University, Shenyang, China
| | - Xuan Wu
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
- Institute of Health Sciences, China Medical University, Shenyang, China
| | - Fang Meng
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Limiao Si
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Sunrun Cao
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yuqing Dong
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Huayi Sun
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Mengzhu Lv
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| | - Hongde Xu
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Ning Bai
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Qiqiang Guo
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xiaoyu Song
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yang Yu
- Institute of Health Sciences, China Medical University, Shenyang, China
| | - Wendong Guo
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Fei Yi
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Tingting Zhou
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xiaoman Li
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yanling Feng
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Zhuo Wang
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Dan Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi Guan
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Mengtao Ma
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Jingwei Liu
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xining Li
- Department of Pathology, School of Medicine, Huzhou University, Zhejiang Province, China
| | - Weidong Zhao
- Department of Developmental Cell Biology, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Baohua Liu
- Center for Anti-Aging and Regenerative Medicine, Shenzhen University Health Science Center, Shenzhen 518060, China
| | - Toren Finkel
- Aging Institute, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Corresponding author. (T.F.); (L.C.)
| | - Liu Cao
- College of Basic Medical Sciences, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
- Institute of Health Sciences, China Medical University, Shenyang, China
- Corresponding author. (T.F.); (L.C.)
| |
Collapse
|
40
|
Díez-Díez M, Amorós-Pérez M, de la Barrera J, Vázquez E, Quintas A, Pascual-Figal DA, Dopazo A, Sánchez-Cabo F, Kleinman ME, Gordon LB, Fuster V, Andrés V, Fuster JJ. Clonal hematopoiesis is not prevalent in Hutchinson-Gilford progeria syndrome. GeroScience 2022; 45:1231-1236. [PMID: 35752705 PMCID: PMC9886702 DOI: 10.1007/s11357-022-00607-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/13/2022] [Indexed: 02/03/2023] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP), defined as the presence of somatic mutations in cancer-related genes in blood cells in the absence of hematological cancer, has recently emerged as an important risk factor for several age-related conditions, especially cardiovascular disease. CHIP is strongly associated with normal aging, but its role in premature aging syndromes is unknown. Hutchinson-Gilford progeria syndrome (HGPS) is an ultra-rare genetic condition driven by the accumulation of a truncated form of the lamin A protein called progerin. HGPS patients exhibit several features of accelerated aging and typically die from cardiovascular complications in their early teens. Previous studies have shown normal hematological parameters in HGPS patients, except for elevated platelets, and low levels of lamin A expression in hematopoietic cells relative to other cell types in solid tissues, but the prevalence of CHIP in HGPS remains unexplored. To investigate the potential role of CHIP in HGPS, we performed high-sensitivity targeted sequencing of CHIP-related genes in blood DNA samples from a cohort of 47 HGPS patients. As a control, the same sequencing strategy was applied to blood DNA samples from middle-aged and elderly individuals, expected to exhibit a biological age and cardiovascular risk profile similar to HGPS patients. We found that CHIP is not prevalent in HGPS patients, in marked contrast to our observations in individuals who age normally. Thus, our study unveils a major difference between HGPS and normal aging and provides conclusive evidence that CHIP is not frequent in HGPS and, therefore, is unlikely to contribute to the pathophysiology of this accelerated aging syndrome.
Collapse
Affiliation(s)
- Miriam Díez-Díez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Marta Amorós-Pérez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Jorge de la Barrera
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Enrique Vázquez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Domingo A. Pascual-Figal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain ,Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares, (CIBERCV) 28029 Madrid, Spain ,Hospital Virgen de La Arrixaca, Universidad de Murcia, 30120 Murcia, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain
| | - Monica E. Kleinman
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115 USA
| | - Leslie B. Gordon
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115 USA ,Department of Pediatrics, Division of Genetics, Hasbro Children’s Hospital and Warren Alpert Medical School of Brown University, Providence, RI 02903 USA ,The Progeria Research Foundation, Peabody, MA 01960 USA
| | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain ,Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029, Madrid, Spain. .,Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares, (CIBERCV), 28029, Madrid, Spain.
| | - José J. Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3., 28029 Madrid, Spain ,Centro de Investigacion Biomedica en Red de Enfermedades Cardiovasculares, (CIBERCV) 28029 Madrid, Spain
| |
Collapse
|
41
|
Meqbel BRM, Gomes M, Omer A, Gallouzi IE, Horn HF. LINCing Senescence and Nuclear Envelope Changes. Cells 2022; 11:1787. [PMID: 35681483 PMCID: PMC9179861 DOI: 10.3390/cells11111787] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 01/27/2023] Open
Abstract
The nuclear envelope (NE) has emerged as a nexus for cellular organization, signaling, and survival. Beyond its role as a barrier to separate the nucleoplasm from the cytoplasm, the NE's role in supporting and maintaining a myriad of other functions has made it a target of study in many cellular processes, including senescence. The nucleus undergoes dramatic changes in senescence, many of which are driven by changes in the NE. Indeed, Lamin B1, a key NE protein that is consistently downregulated in senescence, has become a marker for senescence. Other NE proteins have also been shown to play a role in senescence, including LINC (linker of nucleoskeleton and cytoskeleton) complex proteins. LINC complexes span the NE, forming physical connections between the cytoplasm to the nucleoplasm. In this way, they integrate nuclear and cytoplasmic mechanical signals and are essential not only for a variety of cellular functions but are needed for cell survival. However, LINC complex proteins have been shown to have a myriad of functions in addition to forming a LINC complex, often existing as nucleoplasmic or cytoplasmic soluble proteins in a variety of isoforms. Some of these proteins have now been shown to play important roles in DNA repair, cell signaling, and nuclear shape regulation, all of which are important in senescence. This review will focus on some of these roles and highlight the importance of LINC complex proteins in senescence.
Collapse
Affiliation(s)
- Bakhita R. M. Meqbel
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| | - Matilde Gomes
- KAUST Smart-Health Initiative and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Jeddah 21589, Saudi Arabia; (M.G.); (I.E.G.)
| | - Amr Omer
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Imed E. Gallouzi
- KAUST Smart-Health Initiative and Biological and Environmental Science and Engineering (BESE) Division, King Abdullah University of Science and Technology (KAUST), Jeddah 21589, Saudi Arabia; (M.G.); (I.E.G.)
- Department of Biochemistry, McGill University, 3655 Promenade Sir William Osler, Montreal, QC H3G 1Y6, Canada;
| | - Henning F. Horn
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar;
| |
Collapse
|
42
|
Gao S, Wang S, Zhao Z, Zhang C, Liu Z, Ye P, Xu Z, Yi B, Jiao K, Naik GA, Wei S, Rais-Bahrami S, Bae S, Yang WH, Sonpavde G, Liu R, Wang L. TUBB4A interacts with MYH9 to protect the nucleus during cell migration and promotes prostate cancer via GSK3β/β-catenin signalling. Nat Commun 2022; 13:2792. [PMID: 35589707 PMCID: PMC9120517 DOI: 10.1038/s41467-022-30409-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/28/2022] [Indexed: 01/22/2023] Open
Abstract
Human tubulin beta class IVa (TUBB4A) is a member of the β-tubulin family. In most normal tissues, expression of TUBB4A is little to none, but it is highly expressed in human prostate cancer. Here we show that high expression levels of TUBB4A are associated with aggressive prostate cancers and poor patient survival, especially for African-American men. Additionally, in prostate cancer cells, TUBB4A knockout (KO) reduces cell growth and migration but induces DNA damage through increased γH2AX and 53BP1. Furthermore, during constricted cell migration, TUBB4A interacts with MYH9 to protect the nucleus, but either TUBB4A KO or MYH9 knockdown leads to severe DNA damage and reduces the NF-κB signaling response. Also, TUBB4A KO retards tumor growth and metastasis. Functional analysis reveals that TUBB4A/GSK3β binds to the N-terminal of MYH9, and that TUBB4A KO reduces MYH9-mediated GSK3β ubiquitination and degradation, leading to decreased activation of β-catenin signaling and its relevant epithelial-mesenchymal transition. Likewise, prostate-specific deletion of Tubb4a reduces spontaneous tumor growth and metastasis via inhibition of NF-κB, cyclin D1, and c-MYC signaling activation. Our results suggest an oncogenic role of TUBB4A and provide a potentially actionable therapeutic target for prostate cancers with TUBB4A overexpression. The β-tubulin family protein TUBB4A is highly expressed in cancer but it’s molecular role is unclear. Here, the authors show that TUBB4A is required to protect the nucleus from genomic instability during migration and that it’s over expression promotes cancer progression.
Collapse
Affiliation(s)
- Song Gao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shuaibin Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhiying Zhao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chao Zhang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhicao Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ping Ye
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhifang Xu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Baozhu Yi
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kai Jiao
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gurudatta A Naik
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shi Wei
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Soroush Rais-Bahrami
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sejong Bae
- Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wei-Hsiung Yang
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA, USA
| | | | - Runhua Liu
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Lizhong Wang
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA. .,Department of O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
43
|
Zheng M, Jin G, Zhou Z. Post-Translational Modification of Lamins: Mechanisms and Functions. Front Cell Dev Biol 2022; 10:864191. [PMID: 35656549 PMCID: PMC9152177 DOI: 10.3389/fcell.2022.864191] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/17/2022] [Indexed: 12/22/2022] Open
Abstract
Lamins are the ancient type V intermediate filament proteins contributing to diverse biological functions, such as the maintenance of nuclear morphology, stabilization of chromatin architecture, regulation of cell cycle progression, regulation of spatial-temporal gene expressions, and transduction of mechano-signaling. Deregulation of lamins is associated with abnormal nuclear morphology and chromatin disorganization, leading to a variety of diseases such as laminopathy and premature aging, and might also play a role in cancer. Accumulating evidence indicates that lamins are functionally regulated by post-translational modifications (PTMs) including farnesylation, phosphorylation, acetylation, SUMOylation, methylation, ubiquitination, and O-GlcNAcylation that affect protein stabilization and the association with chromatin or associated proteins. The mechanisms by which these PTMs are modified and the relevant functionality become increasingly appreciated as understanding of these changes provides new insights into the molecular mechanisms underlying the laminopathies concerned and novel strategies for the management. In this review, we discussed a range of lamin PTMs and their roles in both physiological and pathological processes, as well as potential therapeutic strategies by targeting lamin PTMs.
Collapse
Affiliation(s)
- Mingyue Zheng
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guoxiang Jin
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhongjun Zhou
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
44
|
Ribeiro R, Macedo JC, Costa M, Ustiyan V, Shindyapina AV, Tyshkovskiy A, Gomes RN, Castro JP, Kalin TV, Vasques-Nóvoa F, Nascimento DS, Dmitriev SE, Gladyshev VN, Kalinichenko VV, Logarinho E. In vivo cyclic induction of the FOXM1 transcription factor delays natural and progeroid aging phenotypes and extends healthspan. NATURE AGING 2022; 2:397-411. [PMID: 37118067 DOI: 10.1038/s43587-022-00209-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 03/15/2022] [Indexed: 04/30/2023]
Abstract
The FOXM1 transcription factor exhibits pleiotropic C-terminal transcriptional and N-terminal non-transcriptional functions in various biological processes critical for cellular homeostasis. We previously found that FOXM1 repression during cellular aging underlies the senescence phenotypes, which were vastly restored by overexpressing transcriptionally active FOXM1. Yet, it remains unknown whether increased expression of FOXM1 can delay organismal aging. Here, we show that in vivo cyclic induction of an N-terminal truncated FOXM1 transgene on progeroid and naturally aged mice offsets aging-associated repression of full-length endogenous Foxm1, reinstating both transcriptional and non-transcriptional functions. This translated into mitigation of several cellular aging hallmarks, as well as molecular and histopathological progeroid features of the short-lived Hutchison-Gilford progeria mouse model, significantly extending its lifespan. FOXM1 transgene induction also reinstated endogenous Foxm1 levels in naturally aged mice, delaying aging phenotypes while extending their lifespan. Thus, we disclose that FOXM1 genetic rewiring can delay senescence-associated progeroid and natural aging pathologies.
Collapse
Affiliation(s)
- Rui Ribeiro
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Graduate Program in Areas of Basic and Applied Biology (GABBA), ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Joana C Macedo
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Madalena Costa
- Anatomy Department, Unit for Multidisciplinary Biomedical Research, ICBAS - Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Vladimir Ustiyan
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anastasia V Shindyapina
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alexander Tyshkovskiy
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Rita N Gomes
- INEB - Instituto Nacional de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - José Pedro Castro
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Francisco Vasques-Nóvoa
- INEB - Instituto Nacional de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Cardiovascular Research and Development Center, Faculty of Medicine of the University of Porto, Porto, Portugal
| | - Diana S Nascimento
- INEB - Instituto Nacional de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Sergey E Dmitriev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Vladimir V Kalinichenko
- Center for Lung Regenerative Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Elsa Logarinho
- Aging and Aneuploidy Laboratory, i3S - Instituto de Investigação e Inovação em Saúde, IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
45
|
HiIDDD: a high-throughput imaging pipeline for the quantitative detection of DNA damage in primary human immune cells. Sci Rep 2022; 12:6335. [PMID: 35428779 PMCID: PMC9022135 DOI: 10.1038/s41598-022-10018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/25/2022] [Indexed: 11/21/2022] Open
Abstract
DNA damage is a prominent biomarker for numerous diseases, including cancer, as well as for the aging process. Detection of DNA damage routinely relies on traditional microscopy or cytometric methods. However, these techniques are typically of limited throughput and are not ideally suited for large-scale longitudinal and population studies that require analysis of large sample sets. We have developed HiIDDD (High-throughput Immune cell DNA Damage Detection), a robust, quantitative and single-cell assay that measures DNA damage by high-throughput imaging using the two major DNA damage markers 53BP1 and \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$$\upgamma$$\end{document}γ-H2AX. We demonstrate sensitive detection with low inter-assay variability of DNA damage in various types of freshly isolated and cryopreserved primary human immune cells, including CD4 + and CD8 + T cells, B cells and monocytes. As proof of principle, we demonstrate parallel batch processing of several immune cell types from multiple donors. We find common patterns of DNA damage in multiple immune cell types of donors of varying ages, suggesting that immune cell properties are specific to individuals. These results establish a novel high-throughput assay for the evaluation of DNA damage in large-scale studies.
Collapse
|
46
|
Wu G, Tian Q, Liu J, Zhou Q, Zou D, Chen Z, Wu T, Wang W, Xia H, Zhou J. Comprehensive analysis of expression and prognosis for LMNB family genes in human sarcoma. Medicine (Baltimore) 2022; 101:e28933. [PMID: 35356902 PMCID: PMC10513290 DOI: 10.1097/md.0000000000028933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 02/08/2022] [Indexed: 11/26/2022] Open
Abstract
ABSTRACT Previous studies indicated that lamin proteins were thought to be related to gene expression, chromatin structure, and unclear stability. There are 2 types of vertebrate lamins, including A and B. The 2 B type proteins are encoded by lamin B1 (LMNB1) and lamin B2 (LMNB2). The LMNBs factor has been found to be associated with the development of multiple tumors, but its association with sarcoma has been barely mentioned.The transcription levels of LMNBs were analyzed via Oncomine database. Gene Expression Profiling Interactive Analysis (GEPIA) dataset was adopted to analyze the differential expression of LMNBs in sarcoma. Cancer Cell Line Encyclopedia dataset was used to explore the expression of LMNBs in sarcoma cell line. We analyzed the prognostic value of LMNBs in GEPIA and Kaplan-Meier Plotter. Oncomine and GEPIA datasets were also used to detect the relationship between LMNBs and their co-expressed genes. We used the Database for Annotation, Visualization and Integrated Discovery to conduct the Gene Ontology analysis of LMNBs and their co-expressed genes. Kyoto Encyclopedia of Genes and Genomes was also used to analyze the pathway of LMNBs.LMNB1 and LMNB2 were reported to be hyperexpressed in sarcoma. The expression of LMNBs was elevated in various sarcoma cell lines. According to the results, we observed that LMNBs were connected to the poor overall survival, recurrence-free survival, and disease-free survival of sarcoma patients.This study indicated that hyperexpression of LMNBs was significantly related to worse outcome of sarcoma, LMNB1 and LMNB2 were expected to become potential biomarkers for human.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hong Xia
- * Correspondence: Hong Xia, Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan 411100, China (e-mail: ); Jian Zhou,Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China (e-mail: ).
| | - Jian Zhou
- * Correspondence: Hong Xia, Department of Orthopedics, Xiangtan Central Hospital, Xiangtan, Hunan 411100, China (e-mail: ); Jian Zhou,Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China (e-mail: ).
| |
Collapse
|
47
|
González-Blanco C, Marqués P, Burillo J, Jiménez B, García G, Benito M, Guillén C. Cell immortalization facilitates prelamin A clearance by increasing both cell proliferation and autophagic flux. Aging (Albany NY) 2022; 14:2047-2061. [PMID: 35306483 PMCID: PMC8954962 DOI: 10.18632/aging.203943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/25/2022] [Indexed: 12/03/2022]
Abstract
Hutchinson-Gilford Progeria Syndrome is an ultrarare disease which is characterized by an accelerated senescence phenotype with deleterious consequences to people suffering this pathology. The production of an abnormal protein derived from lamin A, called progerin, presents a farnesylated domain, which is not eliminated by the causal mutation of the disease, and accumulates in the interior of the nucleus, provoking a disruption of nuclear membrane, chromatin organization and an altered gene expression. The mutation in these patients occurs in a single nucleotide change, which creates a de novo splicing site, producing a shorter version of the protein. Apart from this mutation, an alteration in the metalloproteinase Zmpste24, involved in the maturation of lamin A, causing a similar alteration than in progeria. However, in this case, patients accumulate a protein, called prelamin A, which generates similar alterations in the nucleus than progerin. The reduction of prelamin A protein levels facilitates the recovery of the phenotype in different mice models of the disease, reducing the aging process. Different strategies have been studied for eliminating this toxic protein. Here, we report that immortalization of primary cells derived from the Zmpste24 KO mice, facilitates prelamin A degradation by different mechanisms, being essential, the enhancing proliferative capacity that the immortalized cells present. Then, these data suggest that using different treatments for increasing proliferative capacity of these cells, potentially could have a beneficial effect, facilitating prelamin A toxicity.
Collapse
Affiliation(s)
| | - Patricia Marqués
- Department of Biochemistry, Complutense University, Madrid, Spain
| | - Jesús Burillo
- Department of Biochemistry, Complutense University, Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), Madrid, Spain
| | - Beatriz Jiménez
- Department of Biochemistry, Complutense University, Madrid, Spain
| | - Gema García
- Department of Biochemistry, Complutense University, Madrid, Spain
| | - Manuel Benito
- Department of Biochemistry, Complutense University, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), Madrid, Spain
| | - Carlos Guillén
- Department of Biochemistry, Complutense University, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Madrid, Spain
- Mechanisms of Insulin Resistance (MOIR2), General Direction of Universities and Investigation (CCMM), Madrid, Spain
| |
Collapse
|
48
|
The Expression of the Senescence-Associated Biomarker Lamin B1 in Human Breast Cancer. Diagnostics (Basel) 2022; 12:diagnostics12030609. [PMID: 35328162 PMCID: PMC8947068 DOI: 10.3390/diagnostics12030609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/27/2022] [Accepted: 02/27/2022] [Indexed: 12/04/2022] Open
Abstract
Senescence is a major response to cancer chemotherapy and has been linked to unfavorable therapy outcomes. Lamin B1 is a component of the nuclear lamina that plays a pivotal role in chromatin stability. Downregulation of lamin B1 represents an established biomarker for cellular senescence. However, the protein expression level of lamin B1 in malignant tissue, particularly of the breast, has not been previously described. In this work, we investigated lamin B1 protein expression in normal breast epithelium, malignant breast tissue (including adjacent non-malignant tissue) and in malignant tissue exposed to neoadjuvant chemotherapy (NAC) using immunohistochemistry (IHC) in three patient groups (n = 15, n = 87, and n = 43, respectively). Our results indicate that lamin B1 mean positive expression was 93% in normal breast epithelium and 88% in malignant breast cells, but significantly decreased (mean: 55%, p < 0.001) in malignant breast tissue after exposure to NAC, suggestive of senescence induction. No significant association between lamin B1 expression and other clinicopathological characteristics or survival of breast cancer patients was recorded. To our knowledge, this is the first report that established the baseline protein expression level of lamin B1 in normal and malignant breast tissue, and its reduction following exposure to chemotherapy. In conclusion, lamin B1 downregulation can be used reliably as a component of multiple biomarker batteries to identify therapy-induced senescence (TIS) in clinical cancer.
Collapse
|
49
|
Li T, Lu D, Yao C, Li T, Dong H, Li Z, Xu G, Chen J, Zhang H, Yi X, Zhu H, Liu G, Wen K, Zhao H, Gao J, Zhang Y, Han Q, Li T, Zhang W, Zhao J, Li T, Bai Z, Song M, He X, Zhou T, Xia Q, Li A, Pan X. Kansl1 haploinsufficiency impairs autophagosome-lysosome fusion and links autophagic dysfunction with Koolen-de Vries syndrome in mice. Nat Commun 2022; 13:931. [PMID: 35177641 PMCID: PMC8854428 DOI: 10.1038/s41467-022-28613-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/15/2022] [Indexed: 12/11/2022] Open
Abstract
Koolen-de Vries syndrome (KdVS) is a rare disorder caused by haploinsufficiency of KAT8 regulatory NSL complex subunit 1 (KANSL1), which is characterized by intellectual disability, heart failure, hypotonia, and congenital malformations. To date, no effective treatment has been found for KdVS, largely due to its unknown pathogenesis. Using siRNA screening, we identified KANSL1 as an essential gene for autophagy. Mechanistic study shows that KANSL1 modulates autophagosome-lysosome fusion for cargo degradation via transcriptional regulation of autophagosomal gene, STX17. Kansl1+/− mice exhibit impairment in the autophagic clearance of damaged mitochondria and accumulation of reactive oxygen species, thereby resulting in defective neuronal and cardiac functions. Moreover, we discovered that the FDA-approved drug 13-cis retinoic acid can reverse these mitophagic defects and neurobehavioral abnormalities in Kansl1+/− mice by promoting autophagosome-lysosome fusion. Hence, these findings demonstrate a critical role for KANSL1 in autophagy and indicate a potentially viable therapeutic strategy for KdVS. Here the authors show that the Koolen-de Vries syndrome associated gene KANSL1 modulates autophagosome-lysosome fusion via transcriptional regulation of autophagosomal gene Syntaxin17, and that 13-cis retinoic acid can reverses mitophagic defects and neurobehavioural abnormalities of mice lacking Kansl1.
Collapse
Affiliation(s)
- Ting Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Dingyi Lu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Chengcheng Yao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Tingting Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Hua Dong
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Zhan Li
- Nanhu Laboratory, Jiaxing, Zhejiang Province, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Guang Xu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Military Institute of Chinese Materia, the Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Jiayi Chen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Hao Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xiaoyu Yi
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Haizhen Zhu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Guangqin Liu
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kaiqing Wen
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Haixin Zhao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,State Key Laboratory of Experimental Haematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jun Gao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Yakun Zhang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Qiuying Han
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Teng Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Weina Zhang
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Jie Zhao
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China
| | - Tao Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Zhaofang Bai
- Military Institute of Chinese Materia, the Fifth Medical Centre of Chinese PLA General Hospital, Beijing, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Xinhua He
- Nanhu Laboratory, Jiaxing, Zhejiang Province, China.,State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and Toxicology, Beijing, China
| | - Tao Zhou
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China.,Nanhu Laboratory, Jiaxing, Zhejiang Province, China
| | - Qing Xia
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China. .,Nanhu Laboratory, Jiaxing, Zhejiang Province, China.
| | - Ailing Li
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China. .,Nanhu Laboratory, Jiaxing, Zhejiang Province, China. .,School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Xin Pan
- State Key Laboratory of Proteomics, Institute of Basic Medical Sciences, National Center of Biomedical Analysis, Beijing, China. .,Nanhu Laboratory, Jiaxing, Zhejiang Province, China. .,School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
50
|
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: 1.0] [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
|