1
|
Joo EY, Park JS, Shin HT, Yoo M, Kim SJ, Lee JE, Choi GS. Mesenchymal Stem Cell Therapy for Hutchinson-Gilford Progeria: Improvements in Arterial Stiffness and Bone Mineral Density in a Single Case. CHILDREN (BASEL, SWITZERLAND) 2025; 12:523. [PMID: 40310235 PMCID: PMC12025413 DOI: 10.3390/children12040523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/24/2025] [Accepted: 04/02/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND/OBJECTIVES Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disorder that cause premature aging due to LMNA mutations and progerin accumulation. Although lonafarnib, an FDA-approved farnesyltransferase inhibitor, offers modest extension of life, the disease remains progressive. As progeria is associated with stem cell depletion and mesenchymal stem cell (MSC) therapy has shown efficacy in treating atherosclerosis, we aimed to evaluate its efficacy and safety in HGPS. METHODS A 7-year-old male with classic HGPS and preexisting severe cerebrovascular disease received four intravenous infusion of bone marrow-derived MSCs (2.5 × 10⁵ cells/kg) over 8 months. Growth, metabolic, cardiovascular, musculoskeletal, auditory, and inflammatory cytokines were monitored throughout the study. Prophylactic enoxaparin was administered to prevent vascular complications. RESULTS MSC therapy was associated with improved lean body mass (11.5%), bone mineral density (L-spine z-score: 0.55 → 2.03), reduced arterial stiffness (9.98% reductionin pulse wave velocity), joint range of motion, dentition, and decreased sICAM-1 levels. However, Cardiovascular deterioration continued, and the patient passed away 10 months after the fourth dose, likely due to progression of the underlying vascular disease. No severe adverse effects were attributed to MSC therapy. CONCLUSIONS MSC therapy may offer short-term benefits in arterial stiffness, bone health and inflammation in HGPS without notable safety concerns. Further studies are warranted to validate these findings, explore earlier intervention, and determine long-term efficacy and optimal dosing strategies.
Collapse
Affiliation(s)
- Eun-Young Joo
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Ji-Sun Park
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Hyun-Tae Shin
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
- Department of Dermatology, Inha University College of Medicine, Incheon 22212, Republic of Korea
| | - Myungji Yoo
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Su-Jin Kim
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Ji-Eun Lee
- Department of Pediatrics, Inha University College of Medicine, Incheon 22212, Republic of Korea; (E.-Y.J.); (J.-S.P.)
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Advanced Regenerative Medicine Clinical Trial Center, Inha University Hospital, Incheon 22332, Republic of Korea
| | - Gwang-Seong Choi
- Gyeonggi-Incheon Regional Specialized Rare Disease Institute, Inha University Hospital, Incheon 22332, Republic of Korea
- Department of Dermatology, Inha University College of Medicine, Incheon 22212, Republic of Korea
| |
Collapse
|
2
|
Xiong J, Ma R, Xie K, Shan C, Chen H, Wang Y, Liao Y, Deng Y, Ye G, Wang Y, Zhu Q, Zhang Y, Cai H, Guo W, Yin Y, Li Z. Recapitulation of endochondral ossification by hPSC-derived SOX9 + sclerotomal progenitors. Nat Commun 2025; 16:2781. [PMID: 40118845 PMCID: PMC11928506 DOI: 10.1038/s41467-025-58122-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/11/2025] [Indexed: 03/24/2025] Open
Abstract
Endochondral ossification generates most of the load-bearing bones, recapitulating it in human cells remains a challenge. Here, we report generation of SOX9+ sclerotomal progenitors (scl-progenitors), a mesenchymal precursor at the pre-condensation stage, from human pluripotent stem cells and development of osteochondral induction methods for these cells. Upon lineage-specific induction, SOX9+ scl-progenitors have not only generated articular cartilage but have also undergone spontaneous condensation, cartilaginous anlagen formation, chondrocyte hypertrophy, vascular invasion, and finally bone formation with stroma, thereby recapitulating key stages during endochondral ossification. Moreover, self-organized growth plate-like structures have also been induced using SOX9+ scl-progenitor-derived fusion constructs with chondro- and osteo-spheroids, exhibiting molecular and cellular similarities to the primary growth plates. Furthermore, we have identified ITGA9 as a specific surface marker for reporter-independent isolation of SOX9+ scl-progenitors and established a culture system to support their expansion. Our work highlights SOX9+ scl-progenitors as a promising tool for modeling human skeletal development and bone/cartilage bioengineering.
Collapse
Affiliation(s)
- Jingfei Xiong
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Runxin Ma
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kun Xie
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Ce Shan
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Hanyi Chen
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuqing Wang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuansong Liao
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yanhui Deng
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Guogen Ye
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yifu Wang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Qing Zhu
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
- Department of Anesthesiology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, Sichuan University, Chengdu, China
| | - Yunqiu Zhang
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Haoyang Cai
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China
| | - Weihua Guo
- Yunnan Key Laboratory of Stomatology, Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming Medical University, Kunming, China
| | - Yike Yin
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China.
| | - Zhonghan Li
- Center of Growth Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, Animal Disease Prevention and Food Safety Key Laboratory of Sichuan Province, College of Life Sciences, Sichuan University, Chengdu, China.
- Yunnan Key Laboratory of Stomatology, Department of Pediatric Dentistry, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming Medical University, Kunming, China.
| |
Collapse
|
3
|
Zeng M, Chen L, Wang Y. Nuclear membrane: A key potential therapeutic target for lipid metabolism. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2024; 194:10-15. [PMID: 39433092 DOI: 10.1016/j.pbiomolbio.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/22/2024] [Accepted: 10/15/2024] [Indexed: 10/23/2024]
Abstract
Lipid homeostasis plays a pivotal role in cellular growth, necessitating the engagement of numerous lipid metabolism genes and the cohesive functioning of organelles. While the nucleus is traditionally recognized for its genetic roles, emerging evidence highlights its significant contribution to lipid homeostasis maintenance. Certain nuclear membrane proteins or associated proteins have the capacity to directly catalyze lipid synthesis or modification processes. Mutations in the genes encoding these proteins can lead to disrupted lipid metabolism, contributing to a spectrum of metabolic disorders. This article provides a comprehensive reviews of the investigations exploring the interplay between nuclear membrane proteins and lipid metabolism. Additionally, it delves into the heterogeneity of the nuclear membrane, positioning it as a novel therapeutic target for managing metabolic disorders and mitigating adverse drug reactions.
Collapse
Affiliation(s)
- Min Zeng
- Department of Gastroenterology, Liuyang Hospital of Chinese Medicine, Liuyang, Hunan, China
| | - Longgui Chen
- Department of Gastroenterology, Liuyang Hospital of Chinese Medicine, Liuyang, Hunan, China.
| | - YaZhu Wang
- Department of Cardiovascular Medicine, Liuyang Hospital of Chinese Medicine, Liuyang, Hunan, China.
| |
Collapse
|
4
|
Kim PH, Kim JR, Heizer PJ, Jung H, Tu Y, Presnell A, Scheithauer J, Yu RG, Young SG, Fong LG. The Accumulation of Progerin Underlies the Loss of Aortic Smooth Muscle Cells in Hutchinson-Gilford Progeria Syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620896. [PMID: 39554077 PMCID: PMC11565845 DOI: 10.1101/2024.10.29.620896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a progeroid disorder characterized by multiple aging-like phenotypes, including disease in large arteries. HGPS is caused by an internally truncated prelamin A (progerin) that cannot undergo the ZMPSTE24-mediated processing step that converts farnesyl-prelamin A to mature lamin A; consequently, progerin retains a carboxyl-terminal farnesyl lipid anchor. In cultured cells, progerin and full-length farnesyl-prelamin A (produced in Zmpste24 -/- cells) form an abnormal nuclear lamin meshwork accompanied by nuclear membrane ruptures and cell death; however, these proteins differ in their capacity to cause arterial disease. In a mouse model of HGPS (Lmna G609G), progerin causes loss of aortic smooth muscle cells (SMCs) by ~12 weeks of age. In contrast, farnesyl-prelamin A in Zmpste24 -/- mice does not cause SMC loss-even at 21 weeks of age. In young mice, aortic levels of farnesyl-prelamin A in Zmpste24 -/- mice and aortic levels of progerin in Lmna G609G/+ mice are the same. However, the levels of progerin and other A-type lamins increase with age in Lmna G609G/+ mice, whereas farnesyl-prelamin A and lamin C levels in Zmpste24 -/- mice remain stable. Lmna transcript levels are similar, implying that progerin influences nuclear lamin turnover. We identified a likely mechanism. In cultured SMCs, the phosphorylation of Ser-404 by AKT (which triggers prelamin A degradation) is reduced in progerin. In mice, AKT activity is significantly lower in Lmna G609G/+ aortas than in wild-type or Zmpste24 -/- aortas. Our studies identify that the accumulation of progerin in Lmna G609G aortas underlies the hallmark arterial pathology in HGPS.
Collapse
Affiliation(s)
- Paul H. Kim
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Joonyoung R. Kim
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Patrick J. Heizer
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Hyesoo Jung
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Yiping Tu
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Ashley Presnell
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Julia Scheithauer
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Rachel G. Yu
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Stephen G. Young
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
- Department of Human Genetics, University of California, Los Angeles; Los Angeles, CA 90095, USA
| | - Loren G. Fong
- Department of Medicine, University of California, Los Angeles; Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Hartinger R, Singh K, Leverett J, Djabali K. Enhancing Cellular Homeostasis: Targeted Botanical Compounds Boost Cellular Health Functions in Normal and Premature Aging Fibroblasts. Biomolecules 2024; 14:1310. [PMID: 39456243 PMCID: PMC11506649 DOI: 10.3390/biom14101310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The human skin, the body's largest organ, undergoes continuous renewal but is significantly impacted by aging, which impairs its function and leads to visible changes. This study aimed to identify botanical compounds that mimic the anti-aging effects of baricitinib, a known JAK1/2 inhibitor. Through in silico screening of a botanical compound library, 14 potential candidates were identified, and 7 were further analyzed for their effects on cellular aging. The compounds were tested on both normal aged fibroblasts and premature aging fibroblasts derived from patients with Hutchinson-Gilford Progeria Syndrome (HGPS). Results showed that these botanical compounds effectively inhibited the JAK/STAT pathway, reduced the levels of phosphorylated STAT1 and STAT3, and ameliorated phenotypic changes associated with cellular aging. Treatments improved cell proliferation, reduced senescence markers, and enhanced autophagy without inducing cytotoxicity. Compounds, such as Resveratrol, Bisdemethoxycurcumin, Pinosylvin, Methyl P-Hydroxycinnamate, cis-Pterostilbene, and (+)-Gallocatechin, demonstrated significant improvements in both control and HGPS fibroblasts. These findings suggest that these botanical compounds have the potential to mitigate age-related cellular alterations, offering promising strategies for anti-aging therapies, particularly for skin health. Further in vivo studies are warranted to validate these results and explore their therapeutic applications.
Collapse
Affiliation(s)
- Ramona Hartinger
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany;
| | - Khushboo Singh
- Amway Corporation, Innovation and Science, 7575 Fulton Street East, Ada, MI 49355, USA
| | - Jesse Leverett
- Amway Corporation, Innovation and Science, 7575 Fulton Street East, Ada, MI 49355, USA
| | - Karima Djabali
- Epigenetics of Aging, Department of Dermatology and Allergy, TUM School of Medicine, Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich (TUM), 85748 Garching, Germany;
| |
Collapse
|
6
|
Yu R, Xue H, Lin W, Collins F, Mount S, Cao K. Progerin mRNA expression in non-HGPS patients is correlated with widespread shifts in transcript isoforms. NAR Genom Bioinform 2024; 6:lqae115. [PMID: 39211333 PMCID: PMC11358823 DOI: 10.1093/nargab/lqae115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/06/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is a premature aging disease caused primarily by a C1824T mutation in LMNA. This mutation activates a cryptic splice donor site, producing a lamin variant called progerin. Interestingly, progerin has also been detected in cells and tissues of non-HGPS patients. Here, we investigated progerin expression using publicly available RNA-seq data from non-HGPS patients in the GTEx project. We found that progerin expression is present across all tissue types in non-HGPS patients and correlated with telomere shortening in the skin. Transcriptome-wide correlation analyses suggest that the level of progerin expression is correlated with switches in gene isoform expression patterns. Differential expression analyses show that progerin expression is correlated with significant changes in genes involved in splicing regulation and mitochondrial function. Interestingly, 5' splice sites whose use is correlated with progerin expression have significantly altered frequencies of consensus trinucleotides within the core 5' splice site. Furthermore, introns whose alternative splicing correlates with progerin have reduced GC content. Our study suggests that progerin expression in non-HGPS patients is part of a global shift in splicing patterns.
Collapse
Affiliation(s)
- Reynold Yu
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Huijing Xue
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Wanru Lin
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Francis S Collins
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephen M Mount
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD, USA
| |
Collapse
|
7
|
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
|
8
|
Jyothi S, Prashanth RR, Nair S, Haribalakrishna A. A Growth-Restricted Neonate with Abnormal Facies and Lax Skin. Neoreviews 2024; 25:e286-e289. [PMID: 38688889 DOI: 10.1542/neo.25-5-e286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Affiliation(s)
- S Jyothi
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - R R Prashanth
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Sruthi Nair
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| | - Anitha Haribalakrishna
- Department of Neonatology, Seth G.S. Medical College and King Edward Memorial Hospital, Mumbai, India
| |
Collapse
|
9
|
Gordon LB, Basso S, Maestranzi J, Aikawa E, Clift CL, Cammardella AG, Danesi TH, del Nido PJ, Edelman ER, Hamdy A, Hegde SM, Kleinman ME, Maschietto N, Mehra MR, Mukundan S, Musumeci F, Russo M, Rybicki FJ, Shah PB, Suarez WA, Tuminelli K, Zaleski K, Prakash A, Gerhard-Herman M. Intervention for critical aortic stenosis in Hutchinson-Gilford progeria syndrome. Front Cardiovasc Med 2024; 11:1356010. [PMID: 38725831 PMCID: PMC11079313 DOI: 10.3389/fcvm.2024.1356010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare genetic premature aging disease that is historically fatal in teenage years, secondary to severe accelerated atherosclerosis. The only approved treatment is the farnesyltransferase inhibitor lonafarnib, which improves vascular structure and function, extending average untreated lifespan of 14.5 years by 4.3 years (30%). With this longer lifespan, calcific aortic stenosis (AS) was identified as an emerging critical risk factor for cardiac death in older patients. Intervention to relieve critical AS has the potential for immediate improvement in healthspan and lifespan. However, HGPS patient-device size mismatch, pervasive peripheral arterial disease, skin and bone abnormalities, and lifelong failure to thrive present unique challenges to intervention. An international group of experts in HGPS, pediatric and adult cardiology, cardiac surgery, and pediatric critical care convened to identify strategies for successful treatment. Candidate procedures were evaluated by in-depth examination of 4 cases that typify HGPS clinical pathology. Modified transcatheter aortic valve replacement (TAVR) and left ventricular Apico-Aortic Conduit (AAC) placement were deemed high risk but viable options. Two cases received TAVR and 2 received AAC post-summit. Three were successful and 1 patient died perioperatively due to cardiovascular disease severity, highlighting the importance of intervention timing and comparative risk stratification. These breakthrough interventions for treating critical aortic stenosis in HGPS patients could rewrite the current clinical perspective on disease course by greatly improving late-stage quality of life and increasing lifespan. Expanding worldwide medical and surgical competency for this ultra-rare disease through expert information-sharing could have high impact on treatment success.
Collapse
Affiliation(s)
- Leslie B. Gordon
- Division of Genetics, Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI, United States
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- The Progeria Research Foundation, Peabody, MA, United States
| | - Sammy Basso
- The Progeria Research Foundation, Peabody, MA, United States
- Associazione Italiana Progeria Sammy Basso, Tezze sul Brenta, Vicenza
- CNR - National Research Council of Italy, Institute of Molecular Genetics Luigi Luca Cavalli-Sforza,Unit 9 of Bologna, Bologna, Italy
- IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Elena Aikawa
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
| | - Cassandra L. Clift
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
| | | | - Tommaso Hinna Danesi
- Department of Surgery, Division of Cardiac Surgery, Brigham and Women's Hospital, Boston, MA, United States
| | - Pedro J. del Nido
- Department of Cardiac Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Elazer R. Edelman
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Abeer Hamdy
- Department of Pediatrics, Division of Pediatric Cardiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sheila M. Hegde
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Monica E. Kleinman
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Nicola Maschietto
- Department of Cardiology, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Mandeep R. Mehra
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Srinivasan Mukundan
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, United States
- Department of Medicine, Boston Children's Hospital, Boston, MA, United States
| | - Francesco Musumeci
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, Roma, Italy
| | - Marco Russo
- Department of Cardiac Surgery and Heart Transplantation, San Camillo Forlanini Hospital, Roma, Italy
| | - Frank J. Rybicki
- Department of Radiology, University of Arizona - Phoenix, Phoenix, AZ, United States
| | - Pinak Bipin Shah
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, United States
| | - William A. Suarez
- Division of Pediatric Cardiology, Department of Pediatrics, University of Toledo Medical Center, Toledo, OH, United States
| | | | - Katherine Zaleski
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Ashwin Prakash
- Department of Cardiology, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Marie Gerhard-Herman
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
10
|
Arun A, Nath AR, Thankachan B, Unnikrishnan MK. Hutchinson-Gilford progeria syndrome: unraveling the genetic basis, symptoms, and advancements in therapeutic approaches. THERAPEUTIC ADVANCES IN RARE DISEASE 2024; 5:26330040241305144. [PMID: 39691184 PMCID: PMC11650505 DOI: 10.1177/26330040241305144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024]
Abstract
Hutchinson-Gilford Progeria syndrome (HGPS) serves as a prominent model for Progeroid syndromes, a group of rare genetic disorders characterized by accelerated aging. This review explores the genetic basis, clinical presentation, and complications of HGPS. HGPS is caused by mutations in the LMNA gene, resulting in the production of a defective structural protein, prelamin A. This protein contains a "CAAX" motif, where C represents cysteine, and its abnormal processing is central to the disease's pathology. HGPS leads to multiple organ systems being affected, including cardiovascular, skeletal, neurological, and dermatological systems, causing severe disability and increased mortality. Cardiovascular issues are particularly significant in HGPS and are crucial for developing therapeutic strategies. Recent advances in treatment modalities offer promise for managing HGPS. Farnesyltransferase inhibitors and genetic interventions, such as CRISPR-Cas9, have shown potential in mitigating progerin-associated symptoms, with encouraging results observed in preclinical and clinical studies. Additionally, emerging therapies such as rapamycin, sulforaphane, and MG132 hold promise in targeting underlying disease mechanisms. Comprehensive management approaches, including growth hormone therapy, retinoids, and dental care, are emphasized to enhance overall patient well-being. Despite progress, further research is essential to unravel the complex pathophysiology of Progeroid syndromes and develop effective treatments. Continued focus on therapies that address progerin accumulation and its downstream effects is vital for improving patient care and outcomes for individuals affected by HGPS and related disorders. This review highlights ongoing efforts to understand and combat Progeroid syndromes, aiming to alleviate the burdens imposed by these debilitating conditions.
Collapse
Affiliation(s)
- Akhil Arun
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, AIMS Ponekkara P.O., Kochi, KL 682041, India
| | - Athira Rejith Nath
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, KL, India
| | - Bonny Thankachan
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, KL, India
| | - M. K. Unnikrishnan
- Department of Pharmacy Practice Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Kochi, KL, India
| |
Collapse
|
11
|
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
|
12
|
Cabral WA, Stephan C, Terajima M, Thaivalappil AA, Blanchard O, Tavarez UL, Narisu N, Yan T, Wincovitch S, Taga Y, Yamauchi M, Kozloff KM, Erdos MR, Collins FS. Bone dysplasia in Hutchinson-Gilford progeria syndrome is associated with dysregulated differentiation and function of bone cell populations. Aging Cell 2023; 22:e13903. [PMID: 37365004 PMCID: PMC10497813 DOI: 10.1111/acel.13903] [Citation(s) in RCA: 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: 03/21/2022] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disorder affecting tissues of mesenchymal origin. Most individuals with HGPS harbor a de novo c.1824C > T (p.G608G) mutation in the gene encoding lamin A (LMNA), which activates a cryptic splice donor site resulting in production of the toxic "progerin" protein. Clinical manifestations include growth deficiency, lipodystrophy, sclerotic dermis, cardiovascular defects, and bone dysplasia. Here we utilized the LmnaG609G knock-in (KI) mouse model of HGPS to further define mechanisms of bone loss associated with normal and premature aging disorders. Newborn skeletal staining of KI mice revealed altered rib cage shape and spinal curvature, and delayed calvarial mineralization with increased craniofacial and mandibular cartilage content. MicroCT analysis and mechanical testing of adult femurs indicated increased fragility associated with reduced bone mass, recapitulating the progressive bone deterioration that occurs in HGPS patients. We investigated mechanisms of bone loss in KI mice at the cellular level in bone cell populations. Formation of wild-type and KI osteoclasts from marrow-derived precursors was inhibited by KI osteoblast-conditioned media in vitro, suggesting a secreted factor(s) responsible for decreased osteoclasts on KI trabecular surfaces in vivo. Cultured KI osteoblasts exhibited abnormal differentiation characterized by reduced deposition and mineralization of extracellular matrix with increased lipid accumulation compared to wild-type, providing a mechanism for altered bone formation. Furthermore, quantitative analyses of KI transcripts confirmed upregulation of adipogenic genes both in vitro and in vivo. Thus, osteoblast phenotypic plasticity, inflammation and altered cellular cross-talk contribute to abnormal bone formation in HGPS mice.
Collapse
Affiliation(s)
- Wayne A. Cabral
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Chris Stephan
- Departments of Orthopedic Surgery and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Masahiko Terajima
- Division of Oral and Craniofacial Health Sciences, Adams School of DentistryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Abhirami A. Thaivalappil
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Owen Blanchard
- Departments of Orthopedic Surgery and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Urraca L. Tavarez
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Narisu Narisu
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Tingfen Yan
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Stephen M. Wincovitch
- Cytogenetics and Microscopy CoreNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Yuki Taga
- Nippi Research Institute of BiomatrixIbarakiJapan
| | - Mitsuo Yamauchi
- Division of Oral and Craniofacial Health Sciences, Adams School of DentistryUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Kenneth M. Kozloff
- Departments of Orthopedic Surgery and Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Michael R. Erdos
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| | - Francis S. Collins
- Molecular Genetics Section, Center for Precision Health ResearchNational Human Genome Research Institute, NIHBethesdaMarylandUSA
| |
Collapse
|
13
|
İPEK İ, DERDİYOK C, ÖZNURHAN F. WIEDEMANN–RAUTENSTRAUCH SYNDROME: CASE REPORT. CUMHURIYET DENTAL JOURNAL 2022. [DOI: 10.7126/cumudj.1127344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- İrem İPEK
- FIRAT ÜNİVERSİTESİ, DİŞ HEKİMLİĞİ FAKÜLTESİ
| | | | | |
Collapse
|
14
|
Lamis A, Siddiqui SW, Ashok T, Patni N, Fatima M, Aneef AN. Hutchinson-Gilford Progeria Syndrome: A Literature Review. Cureus 2022; 14:e28629. [PMID: 36196312 PMCID: PMC9524302 DOI: 10.7759/cureus.28629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2022] [Indexed: 11/05/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging condition that involves genetic mutations, resulting in debilitating phenotypic features. The present state of knowledge on the molecular pathways that contribute to the pathophysiology of HGPS and the techniques being tested in vitro and in vivo to combat progerin toxicity have been discussed here. Nuclear morphological abnormalities, dysregulated gene expression, DNA repair deficiencies, telomere shortening, and genomic instability are all caused by progerin accumulation, all of which impair cellular proliferative capability. In addition, HGPS cells and preclinical animal models have revealed new information about the disease's molecular and cellular pathways and putative mechanisms involved in normal aging. This article has discussed the understanding of the molecular pathways by which progerin expression leads to HGPS and how the advanced therapy options for HGPS patients can help us understand and treat the condition.
Collapse
|
15
|
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
|
16
|
Sengupta D, Sengupta K. Lamin A and telomere maintenance in aging: Two to Tango. Mutat Res 2022; 825:111788. [PMID: 35687934 DOI: 10.1016/j.mrfmmm.2022.111788] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 06/15/2023]
Abstract
Lamin proteins which constitute the nuclear lamina in almost all higher eukaryotes, are mainly of two types A & B encoded by LMNA and LMNB1/B2 genes respectively. While lamin A remains the principal product of LMNA gene, variants like lamin C, C2 and A∆10 are also formed as alternate splice products. Role of lamin A in aging has been highlighted in recent times due to its association with progeroid or premature aging syndromes which is classified as a type of laminopathy. Progeria caused by accelerated accumulation of lamin A Δ50 or progerin occurs due to a mutation in this LMNA gene leading to defects in post translational modification of lamin A. One of the most common and severe symptoms of progeroid laminopathy is accelerated cellular senescence or aging along with bone resorption, muscle weakness, lipodystrophy and cardiovascular disorders. On the other hand, progerin accumulation and telomere dysfunction merge as common traits in the process of chronological aging. Two major hallmarks of physiological aging in humans include loss of genomic integrity and telomere attrition which can result from defective laminar organization leading to deformed nuclear architecture and culminates into replicative senescence. This also adversely affects epigenetic landscape, mitochondrial dysfunction and several signalling pathways like DNA repair, mTOR, MAPK, TGFβ. In this review, we discuss the telomere-lamina interplay in the context of physiological aging and progeria.
Collapse
Affiliation(s)
- Duhita Sengupta
- Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, West Bengal, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India
| | - Kaushik Sengupta
- Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, West Bengal, India; Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai 400094, India.
| |
Collapse
|
17
|
Wang J, Yu Q, Ma X, Yuan Z, Mao J. Hutchinson-Gilford progeria syndrome complicated with stroke: A report of 2 cases and literature review. Front Pediatr 2022; 10:1056225. [PMID: 36523395 PMCID: PMC9745312 DOI: 10.3389/fped.2022.1056225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/24/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Hutchinson-Gilford Progeria Syndrome (HGPS) is a ultrarare, fatal autosomal dominant disorder. The pathogenesis of the disease is a mutation in LMNA, which leads to the accumulation of progerin in cells, impairing the normal physiological functions. Stroke and transient ischemic attack seriously affect the survival rate and quality of life of HGPS children, although the literature of this aspect is limited. This study summarizes the clinical manifestations and related imaging features of HGPS children with stroke to improve pediatric clinicians' understanding of this disease. CASE PRESENTATION Both children have a de novo heterozygous mutation of LMNA [c.1824C > T ( p.G608G)]. Case 1. At the age of 4 years, the child had a cerebral infarction, which manifested as blurred vision and communication disturbance. Multiple abnormal signals were observed on the head MRI in the bilateral frontoparietal cortex, bilateral semiovale center, lateral ventricle, and deep frontal and parietal lobes. Multiple abnormal white matter signals on head MRA: bilateral internal carotid artery stenosis with basilar artery, and bilateral thickening of the posterior communicating artery. Case 2. At the age of 8.5 years, the child presented with cerebral infarction, which manifested as decreased muscle strength and choking after drinking water. MRI of the head showed that the bilateral frontal lobes were small with multiple abnormal signal shadows in the bilateral center of the semiovale and the lateral ventricle. Brain MRA revealed that the bilateral internal carotid arteries (C5-7) were narrow and uneven in thickness, and the A1 segment of the left anterior cerebral artery was narrower than the contralateral one. After symptomatic and supportive treatment, the two children improved. CONCLUSION Hemiplegia and physical weakness are the most prevalent stroke symptoms in children with HGPS, followed by headache, epilepsy, dysarthria, and psychosis as the primary manifestation in some children. Stroke in children with HGPS is mostly ischemic cerebral infarction caused by an insufficient cerebral blood supply. Pediatric cerebral infarction mainly occurs in the large vascular area, involving all vascular areas, with the internal carotid artery and middle cerebral artery being the most commonly accumulated.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qinmei Yu
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Xiaohui Ma
- Department of Radiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhefeng Yuan
- Department of Neurology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
18
|
Alfahaad H. A 3-year-old girl with old face appearance: Case report. JOURNAL OF DERMATOLOGY & DERMATOLOGIC SURGERY 2022. [DOI: 10.4103/jdds.jdds_51_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
19
|
Efficacy of Cord Blood Cell Therapy for Hutchinson-Gilford Progeria Syndrome-A Case Report. Int J Mol Sci 2021; 22:ijms222212316. [PMID: 34830197 PMCID: PMC8619635 DOI: 10.3390/ijms222212316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 02/07/2023] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is an extremely rare premature aging disorder characterized by short stature and atherosclerosis-induced death within teenage years. A 13-year-old male diagnosed with HGPS was administered three intravenous infusions of allogeneic cord blood (CB) cells from unrelated donors at four-month intervals to evaluate the safety and its therapeutic efficacy. Adverse events were monitored in addition to height, weight, laboratory blood tests, joint range of motion (ROM), and carotid Doppler. Cytokine and receptor assays were also performed. The patient exhibited an increase in growth rate for both height and weight. One year after therapy initiation, evident amelioration in pulse wave velocity, bilateral maximal intima-media thickness, and dyslipidemic status were observed, which were in abrupt aggravation prior to treatment. Further, an increase in flexibility occurred in some joints of the upper extremities. No serious adverse events were observed throughout the study period and one year beyond. A molecular assay revealed downregulation of proinflammatory and atherosclerosis, representing cytokine expressions following the administration of CB cells. This is the first reported case of an allogeneic CB trial in a patient with HGPS showing therapeutic effects of CB with improvements in anthropometric measures, joint ROM with amelioration of atherosclerosis, and dyslipidemia induced by anti-inflammatory and anti-atherosclerotic responses.
Collapse
|
20
|
Mojiri A, Walther BK, Jiang C, Matrone G, Holgate R, Xu Q, Morales E, Wang G, Gu J, Wang R, Cooke JP. Telomerase therapy reverses vascular senescence and extends lifespan in progeria mice. Eur Heart J 2021; 42:4352-4369. [PMID: 34389865 PMCID: PMC8603239 DOI: 10.1093/eurheartj/ehab547] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/29/2021] [Accepted: 08/12/2021] [Indexed: 12/28/2022] Open
Abstract
AIMS Hutchinson-Gilford progeria syndrome (HGPS) is an accelerated ageing syndrome associated with premature vascular disease and death due to heart attack and stroke. In HGPS a mutation in lamin A (progerin) alters nuclear morphology and gene expression. Current therapy increases the lifespan of these children only modestly. Thus, greater understanding of the underlying mechanisms of HGPS is required to improve therapy. Endothelial cells (ECs) differentiated from induced pluripotent stem cells (iPSCs) derived from these patients exhibit hallmarks of senescence including replication arrest, increased expression of inflammatory markers, DNA damage, and telomere erosion. We hypothesized that correction of shortened telomeres may reverse these measures of vascular ageing. METHODS AND RESULTS We generated ECs from iPSCs belonging to children with HGPS and their unaffected parents. Telomerase mRNA (hTERT) was used to treat HGPS ECs. Endothelial morphology and functions were assessed, as well as proteomic and transcriptional profiles with attention to inflammatory markers, DNA damage, and EC identity genes. In a mouse model of HGPS, we assessed the effects of lentiviral transfection of mTERT on measures of senescence, focusing on the EC phenotype in various organs. hTERT treatment of human HGPS ECs improved replicative capacity; restored endothelial functions such as nitric oxide generation, acetylated low-density lipoprotein uptake and angiogenesis; and reduced the elaboration of inflammatory cytokines. In addition, hTERT treatment improved cellular and nuclear morphology, in association with a normalization of the transcriptional profile, effects that may be mediated in part by a reduction in progerin expression and an increase in sirtuin 1 (SIRT1). Progeria mice treated with mTERT lentivirus manifested similar improvements, with a reduction in inflammatory and DNA damage markers and increased SIRT1 in their vasculature and other organs. Furthermore, mTERT therapy increased the lifespan of HGPS mice. CONCLUSION Vascular rejuvenation using telomerase mRNA is a promising approach for progeria and other age-related diseases.
Collapse
Affiliation(s)
- Anahita Mojiri
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Brandon K Walther
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
- Department of Biomedical Engineering, Texas A&M University, 101 Bizzell St., College Station, TX 77840, USA
| | - Chongming Jiang
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gianfranco Matrone
- British Heart Foundation Centre for Cardiovascular Science, Queen’s Medical Research Institute, The University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Rhonda Holgate
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Qiu Xu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Elisa Morales
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
- Center for Bioinformatics and Computational Biology, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Jianhua Gu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| | - Rongfu Wang
- Department of Medicine, and Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - John P Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6670 Bertner Ave., R10-South, Houston, TX 77030, USA
| |
Collapse
|
21
|
Najdi F, Krüger P, Djabali K. Impact of Progerin Expression on Adipogenesis in Hutchinson-Gilford Progeria Skin-Derived Precursor Cells. Cells 2021; 10:cells10071598. [PMID: 34202258 PMCID: PMC8306773 DOI: 10.3390/cells10071598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/19/2021] [Accepted: 06/23/2021] [Indexed: 01/10/2023] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a segmental premature aging disease caused by a mutation in LMNA. The mutation generates a truncated and farnesylated form of prelamin A, called progerin. Affected individuals develop several features of normal aging, including lipodystrophy caused by the loss of general subcutaneous fat. To determine whether premature cellular senescence is responsible for the altered adipogenesis in patients with HGPS, we evaluated the differentiation of HGPS skin-derived precursor stem cells (SKPs) into adipocytes. The SKPs were isolated from primary human HGPS and normal fibroblast cultures, with senescence of 5 and 30%. We observed that the presence of high numbers of senescent cells reduced SKPs’ adipogenic differentiation potential. Treatment with baricitinib, a JAK–STAT inhibitor, ameliorated the ability of HGPS SKPs to differentiate into adipocytes. Our findings suggest that the development of lipodystrophy in patients with HGPS may be associated with an increased rate of cellular senescence and chronic inflammation.
Collapse
|
22
|
Ikegami K, Secchia S, Almakki O, Lieb JD, Moskowitz IP. Phosphorylated Lamin A/C in the Nuclear Interior Binds Active Enhancers Associated with Abnormal Transcription in Progeria. Dev Cell 2020; 52:699-713.e11. [PMID: 32208162 DOI: 10.1016/j.devcel.2020.02.011] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 12/13/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023]
Abstract
LMNA encodes nuclear Lamin A/C that tethers lamina-associated domains (LADs) to the nuclear periphery. Mutations in LMNA cause degenerative disorders including the premature aging disorder Hutchinson-Gilford progeria, but the mechanisms are unknown. We report that Ser22-phosphorylated (pS22) Lamin A/C was localized to the nuclear interior in human fibroblasts throughout the cell cycle. pS22-Lamin A/C interacted with a subset of putative active enhancers, not LADs, at locations co-bound by the transcriptional activator c-Jun. In progeria-patient fibroblasts, a subset of pS22-Lamin A/C-binding sites were lost, whereas new pS22-Lamin A/C-binding sites emerged in normally quiescent loci. New pS22-Lamin A/C binding was accompanied by increased histone acetylation, increased c-Jun binding, and upregulation of nearby genes implicated in progeria pathophysiology. These results suggest that Lamin A/C regulates gene expression by enhancer binding. Disruption of the gene regulatory rather than LAD tethering function of Lamin A/C may underlie the pathogenesis of disorders caused by LMNA mutations.
Collapse
Affiliation(s)
- Kohta Ikegami
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA.
| | - Stefano Secchia
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA; Department of Biology, Lunds University, Lund 22362, Sweden
| | - Omar Almakki
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA
| | - Jason D Lieb
- Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA
| | - Ivan P Moskowitz
- Department of Pediatrics, The University of Chicago, Chicago, IL 60637, USA; Department of Human Genetics, The University of Chicago, Chicago, IL 60637, USA; Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
23
|
Pachajoa H, Claros-Hulbert A, García-Quintero X, Perafan L, Ramirez A, Zea-Vera AF. Hutchinson-Gilford Progeria Syndrome: Clinical and Molecular Characterization. Appl Clin Genet 2020; 13:159-164. [PMID: 32943904 PMCID: PMC7481268 DOI: 10.2147/tacg.s238715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 05/13/2020] [Indexed: 11/23/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare congenital disease caused by mutations in the LMNA gene. Children with HGPS are phenotypically characterized by lipodystrophy, short height, low body weight, scleroderma, reduced joint mobility, osteolysis, senile facial features, and cardiovascular compromise that usually lead to death. We aimed to describe the case of a patient who reached above-average age expectancy for children with HGPS in Latin America and describe the clinical and molecular characteristics of the patient. A 14-year-old female patient was presented with progeria-compatible phenotypic characteristics. HGPS was confirmed via LMNA gene sequencing that detected a heterozygous c.1824C>T (p.Gly608Gly) mutation. The primary aim is to describe the HGPS case, the molecular gene mutation finding, and make a short review of the limited available treatment options for children with HGPS. Such as the farnesyl transferase inhibitors in conjunction with other pharmacological therapies that have insinuated improvement in health, and survival rate.
Collapse
Affiliation(s)
- Harry Pachajoa
- Faculty of Health Sciences, Congenital Anomalies and Rare Diseases Investigation Center (CIACER), Universidad Icesi, Cali, Colombia.,Genetic Department, Fundacion Valle del Lili, Cali, Colombia
| | - Angelica Claros-Hulbert
- Pediatric Palliative Care Department, Fundacion Valle del Lili, Cali, Colombia.,Clinical Investigation Center (CIC), Fundacion Valle del Lili, Cali, Colombia
| | - Ximena García-Quintero
- Pediatric Palliative Care Department, Fundacion Valle del Lili, Cali, Colombia.,Clinical Investigation Center (CIC), Fundacion Valle del Lili, Cali, Colombia
| | - Lina Perafan
- Faculty of Health Sciences, Congenital Anomalies and Rare Diseases Investigation Center (CIACER), Universidad Icesi, Cali, Colombia
| | - Andres Ramirez
- Faculty of Health Sciences, Praxis Jessen² + Kollegen, Berlin, Germany
| | | |
Collapse
|
24
|
Evaluation of musculoskeletal phenotype of the G608G progeria mouse model with lonafarnib, pravastatin, and zoledronic acid as treatment groups. Proc Natl Acad Sci U S A 2020; 117:12029-12040. [PMID: 32404427 DOI: 10.1073/pnas.1906713117] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a uniformly fatal condition that is especially prevalent in skin, cardiovascular, and musculoskeletal systems. A wide gap exists between our knowledge of the disease and a promising treatment or cure. The aim of this study was to first characterize the musculoskeletal phenotype of the homozygous G608G BAC-transgenic progeria mouse model, and to determine the phenotype changes of HGPS mice after a five-arm preclinical trial of different treatment combinations with lonafarnib, pravastatin, and zoledronic acid. Microcomputed tomography and CT-based rigidity analyses were performed to assess cortical and trabecular bone structure, density, and rigidity. Bones were loaded to failure with three-point bending to assess strength. Contrast-enhanced µCT imaging of mouse femurs was performed to measure glycosaminoglycan content, thickness, and volume of the femoral head articular cartilage. Advanced glycation end products were assessed with a fluorometric assay. The changes demonstrated in the cortical bone structure, rigidity, stiffness, and modulus of the HGPS G608G mouse model may increase the risk for bending and deformation, which could result in the skeletal dysplasia characteristic of HGPS. Cartilage abnormalities seen in this HGPS model resemble changes observed in the age-matched WT controls, including early loss of glycosaminoglycans, and decreased cartilage thickness and volume. Such changes might mimic prevalent degenerative joint diseases in the elderly. Lonafarnib monotherapy did not improve bone or cartilage parameters, but treatment combinations with pravastatin and zoledronic acid significantly improved bone structure and mechanical properties and cartilage structural parameters, which ameliorate the musculoskeletal phenotype of the disease.
Collapse
|
25
|
Hamczyk MR, Villa-Bellosta R, Quesada V, Gonzalo P, Vidak S, Nevado RM, Andrés-Manzano MJ, Misteli T, López-Otín C, Andrés V. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle cells. EMBO Mol Med 2020; 11:emmm.201809736. [PMID: 30862662 PMCID: PMC6460349 DOI: 10.15252/emmm.201809736] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS) is a rare genetic disorder caused by progerin, a mutant lamin A variant. HGPS patients display accelerated aging and die prematurely, typically from atherosclerosis complications. Recently, we demonstrated that progerin‐driven vascular smooth muscle cell (VSMC) loss accelerates atherosclerosis leading to premature death in apolipoprotein E‐deficient mice. However, the molecular mechanism underlying this process remains unknown. Using a transcriptomic approach, we identify here endoplasmic reticulum stress (ER) and the unfolded protein responses as drivers of VSMC death in two mouse models of HGPS exhibiting ubiquitous and VSMC‐specific progerin expression. This stress pathway was also activated in HGPS patient‐derived cells. Targeting ER stress response with a chemical chaperone delayed medial VSMC loss and inhibited atherosclerosis in both progeria models, and extended lifespan in the VSMC‐specific model. Our results identify a mechanism underlying cardiovascular disease in HGPS that could be targeted in patients. Moreover, these findings may help to understand other vascular diseases associated with VSMC death, and provide insight into aging‐dependent vascular damage related to accumulation of unprocessed toxic forms of lamin A.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain.,Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | - Ricardo Villa-Bellosta
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Fundación Instituto de Investigación Sanitaria Fundación Jiménez Díaz (FIIS-FJD), Madrid, Spain
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Pilar Gonzalo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Sandra Vidak
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - María J Andrés-Manzano
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Tom Misteli
- Cell Biology of Genomes Group, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, Oviedo, Spain .,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain .,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain
| |
Collapse
|
26
|
Saxena S, Kumar S. Pharmacotherapy to gene editing: potential therapeutic approaches for Hutchinson-Gilford progeria syndrome. GeroScience 2020; 42:467-494. [PMID: 32048129 PMCID: PMC7205988 DOI: 10.1007/s11357-020-00167-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS), commonly called progeria, is an extremely rare disorder that affects only one child per four million births. It is characterized by accelerated aging in affected individuals leading to premature death at an average age of 14.5 years due to cardiovascular complications. The main cause of HGPS is a sporadic autosomal dominant point mutation in LMNA gene resulting in differently spliced lamin A protein known as progerin. Accumulation of progerin under nuclear lamina and activation of its downstream effectors cause perturbation in cellular morphology and physiology which leads to a systemic disorder that mainly impairs the cardiovascular system, bones, skin, and overall growth. Till now, no cure has been found for this catastrophic disorder; however, several therapeutic strategies are under development. The current review focuses on the overall progress in the field of therapeutic approaches for the management/cure of HGPS. We have also discussed the new disease models that have been developed for the study of this rare disorder. Moreover, we have highlighted the therapeutic application of extracellular vesicles derived from stem cells against aging and aging-related disorders and, therefore, suggest the same for the treatment of HGPS.
Collapse
Affiliation(s)
- Saurabh Saxena
- Department of Medical Laboratory Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India.
| | - Sanjeev Kumar
- Faculty of Technology and Sciences, Lovely Professional University, Jalandhar - Delhi G.T. Road, Phagwara, Punjab, 144411, India
| |
Collapse
|
27
|
Tsai A, Johnston PR, Gordon LB, Walters M, Kleinman M, Laor T. Skeletal maturation and long-bone growth patterns of patients with progeria: a retrospective study. THE LANCET. CHILD & ADOLESCENT HEALTH 2020; 4:281-289. [PMID: 32119840 PMCID: PMC10640888 DOI: 10.1016/s2352-4642(20)30023-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 01/10/2020] [Accepted: 01/13/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome (termed progeria in this Article) is a rare sporadic genetic disorder. One early clinical manifestation of progeria is abnormal skeletal growth, yet this growth has not been fully characterised. We aimed to characterise the skeletal maturation and long-bone growth patterns of patients with the clinical phenotype of progeria. METHODS For this retrospective study, we reviewed skeletal surveys of patients (aged <20 years) with progeria obtained over a 9·5-year period. Most surveys included radiographs of the hands and long bones (humeri, radii, ulnas, tibias, and fibulas). Bone ages of these patients were estimated by the standards of Greulich and Pyle. Following the established methods for studying long-bone growth, the study cohort was separated into two overlapping age groups: longitudinal bone length measurements were made between physes for the childhood group (aged 12 years or younger) and from the upper margins of the proximal to the lower margin of the distal ossified epiphyses for the adolescent group (aged 10 years or older). Bone age estimates and bone length measurements were plotted against the chronological age of patients and compared with reference standards. Statistical analyses were based on mixed models. FINDINGS 85 patients with progeria and 250 skeletal surveys were included in our study. For both sexes, bone age estimates showed a more advanced skeletal maturation rate throughout all chronological ages than the normal rate of 1 (p<0·0001), with the rate of maturation being 1·09 (SE 0·02) for boys and 1·14 (0·02) for girls. Longitudinal long-bone lengths began to deviate from normal standards by age 1-2 years. Growth curves for these long bones plateaued at about half the normal eventual bone length, and the half-life (the time taken to grow to half the eventual bone length) was also about half the time compared with normal standards. INTERPRETATION Our study established growth curves that might serve as reference standards for skeletal maturation and long-bone growth of patients with the clinical phenotype of progeria. FUNDING The Progeria Research Foundation, the US National Heart, Lung and Blood Institute, the Dana-Farber Cancer Institute Stop&Shop Pediatric Brain Tumor Program, the US National Center for Research Resources, US National Institutes of Health.
Collapse
Affiliation(s)
- Andy Tsai
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA.
| | | | - Leslie B Gordon
- Department of Pediatrics, Hasbro Children's Hospital, Providence, RI, USA; Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Michele Walters
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| | - Monica Kleinman
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Tal Laor
- Department of Radiology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
28
|
Kreienkamp R, Gonzalo S. Metabolic Dysfunction in Hutchinson-Gilford Progeria Syndrome. Cells 2020; 9:cells9020395. [PMID: 32046343 PMCID: PMC7072593 DOI: 10.3390/cells9020395] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/22/2022] Open
Abstract
Hutchinson–Gilford Progeria Syndrome (HGPS) is a segmental premature aging disease causing patient death by early teenage years from cardiovascular dysfunction. Although HGPS does not totally recapitulate normal aging, it does harbor many similarities to the normal aging process, with patients also developing cardiovascular disease, alopecia, bone and joint abnormalities, and adipose changes. It is unsurprising, then, that as physicians and scientists have searched for treatments for HGPS, they have targeted many pathways known to be involved in normal aging, including inflammation, DNA damage, epigenetic changes, and stem cell exhaustion. Although less studied at a mechanistic level, severe metabolic problems are observed in HGPS patients. Interestingly, new research in animal models of HGPS has demonstrated impressive lifespan improvements secondary to metabolic interventions. As such, further understanding metabolism, its contribution to HGPS, and its therapeutic potential has far-reaching ramifications for this disease still lacking a robust treatment strategy.
Collapse
Affiliation(s)
- Ray Kreienkamp
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
- Department of Pediatrics Residency, Washington University Medical School, St. Louis, MO 63105, USA;
| | - Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St Louis, MO 63104, USA
- Correspondence: ; Tel.: +1-314-977-9244
| |
Collapse
|
29
|
Genomic instability and innate immune responses to self-DNA in progeria. GeroScience 2019; 41:255-266. [PMID: 31280482 DOI: 10.1007/s11357-019-00082-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 06/21/2019] [Indexed: 12/15/2022] Open
Abstract
In the last decade, we have seen increasing evidence of the importance of structural nuclear proteins such as lamins in nuclear architecture and compartmentalization of genome function and in the maintenance of mechanical stability and genome integrity. With over 400 mutations identified in the LMNA gene (encoding for A-type lamins) associated with more than ten distinct degenerative disorders, the role of lamins as genome caretakers and the contribution of lamins dysfunction to disease are unarguable. However, the molecular mechanisms whereby lamins mutations cause pathologies remain less understood. Here, we review pathways and mechanisms recently identified as playing a role in the pathophysiology of laminopathies, with special emphasis in Hutchinson Gilford Progeria Syndrome (HGPS). This devastating incurable accelerated aging disease is caused by a silent mutation in the LMNA gene that generates a truncated lamin A protein "progerin" that exerts profound cellular toxicity and organismal decline. Patients usually die in their teens due to cardiovascular complications such as myocardial infarction or stroke. To date, there are no efficient therapies that ameliorate disease progression, stressing the need to understand molecularly disease mechanisms that can be targeted therapeutically. We will summarize data supporting that replication stress is a major cause of genomic instability in laminopathies, which contributes to the activation of innate immune responses to self-DNA that in turn accelerate the aging process.
Collapse
|
30
|
Kreienkamp R, Gonzalo S. Hutchinson-Gilford Progeria Syndrome: Challenges at Bench and Bedside. Subcell Biochem 2019; 91:435-451. [PMID: 30888661 DOI: 10.1007/978-981-13-3681-2_15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The structural nuclear proteins known as "lamins" (A-type and B-type) provide a scaffold for the compartmentalization of genome function that is important to maintain genome stability. Mutations in the LMNA gene -encoding for A-type lamins- are associated with over a dozen of degenerative disorders termed laminopathies, which include muscular dystrophies, lipodystrophies, neuropathies, and premature ageing diseases such as Hutchinson Gilford Progeria Syndrome (HGPS). This devastating disease is caused by the expression of a truncated lamin A protein named "progerin". To date, there is no effective treatment for HGPS patients, who die in their teens from cardiovascular disease. At a cellular level, progerin expression impacts nuclear architecture, chromatin organization, response to mechanical stress, and DNA transactions such as transcription, replication and repair. However, the current view is that key mechanisms behind progerin toxicity still remain to be discovered. Here, we discuss new findings about pathological mechanisms in HGPS, especially the contribution of replication stress to cellular decline, and therapeutic strategies to ameliorate progerin toxicity. In particular, we present evidence for retinoids and calcitriol (hormonal vitamin D metabolite) being among the most potent compounds to ameliorate HGPS cellular phenotypes in vitro, providing the rationale for testing these compounds in preclinical models of the disease in the near term, and in patients in the future.
Collapse
Affiliation(s)
- Ray Kreienkamp
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Doisy Research Center, St Louis University School of Medicine, St. Louis, MO, USA
| | - Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Doisy Research Center, St Louis University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
31
|
Choi JY, Lai JK, Xiong ZM, Ren M, Moorer MC, Stains JP, Cao K. Diminished Canonical β-Catenin Signaling During Osteoblast Differentiation Contributes to Osteopenia in Progeria. J Bone Miner Res 2018; 33:2059-2070. [PMID: 30001457 PMCID: PMC7739562 DOI: 10.1002/jbmr.3549] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 06/25/2018] [Accepted: 06/30/2018] [Indexed: 12/31/2022]
Abstract
Patients with Hutchinson-Gilford progeria syndrome (HGPS) have low bone mass and an atypical skeletal geometry that manifests in a high risk of fractures. Using both in vitro and in vivo models of HGPS, we demonstrate that defects in the canonical WNT/β-catenin pathway, seemingly at the level of the efficiency of nuclear import of β-catenin, impair osteoblast differentiation and that restoring β-catenin activity rescues osteoblast differentiation and significantly improves bone mass. Specifically, we show that HGPS patient-derived iPSCs display defects in osteoblast differentiation, characterized by a decreased alkaline phosphatase activity and mineralizing capacity. We demonstrate that the canonical WNT/β-catenin pathway, a major signaling cascade involved in skeletal homeostasis, is impaired by progerin, causing a reduction in the active β-catenin in the nucleus and thus decreased transcriptional activity, and its reciprocal cytoplasmic accumulation. Blocking farnesylation of progerin restores active β-catenin accumulation in the nucleus, increasing signaling, and ameliorates the defective osteogenesis. Moreover, in vivo analysis of the Zmpste24-/- HGPS mouse model demonstrates that treatment with a sclerostin-neutralizing antibody (SclAb), which targets an antagonist of canonical WNT/β-catenin signaling pathway, fully rescues the low bone mass phenotype to wild-type levels. Together, this study reveals that the β-catenin signaling cascade is a therapeutic target for restoring defective skeletal microarchitecture in HGPS. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ji Young Choi
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD 20742
| | - Jim K Lai
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Zheng-Mei Xiong
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD 20742
| | - Margaret Ren
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD 20742
| | - Megan C Moorer
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Joseph P Stains
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD 21201
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland College Park, MD 20742
| |
Collapse
|
32
|
Choi H, Kim TH, Jeong JK, Strandgren C, Eriksson M, Cho ES. Expression of the Hutchinson-Gilford Progeria Mutation Leads to Aberrant Dentin Formation. Sci Rep 2018; 8:15368. [PMID: 30337599 PMCID: PMC6193977 DOI: 10.1038/s41598-018-33764-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Accepted: 10/05/2018] [Indexed: 12/24/2022] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare accelerated senescence disease, manifesting dental abnormalities and several symptoms suggestive of premature aging. Although irregular secondary dentin formation in HGPS patients has been reported, pathological mechanisms underlying aberrant dentin formation remain undefined. In this study, we analyzed the mandibular molars of a tissue-specific mouse model that overexpresses the most common HGPS mutation (LMNA, c.1824C > T, p.G608G) in odontoblasts. In the molars of HGPS mutant mice at postnatal week 13, targeted expression of the HGPS mutation in odontoblasts results in excessive dentin formation and pulp obliteration. Circumpulpal dentin of HGPS mutants was clearly distinguished from secondary dentin of wild-type (WT) littermates and its mantle dentin by considering the irregular porous structure and loss of dentinal tubules. However, the dentin was significantly thinner in the molars of HGPS mutants at postnatal weeks 3 and 5 than in those of WT mice. In vitro analyses using MDPC-23, a mouse odontoblastic cell line, showed cellular senescence, defects of signaling pathways and consequential downregulation of matrix protein expression in progerin-expressing odontoblasts. These results indicate that expression of the HGPS mutation in odontoblasts disturbs physiological secondary dentin formation. In addition, progerin-expressing odontoblasts secrete paracrine factors that can stimulate odontogenic differentiation of dental pulp cells. Taken together, our results suggest that the aberrant circumpulpal dentin of HGPS mutants results from defects in physiological secondary dentin formation and consequential pathologic response stimulated by paracrine factors from neighboring progerin-expressing odontoblasts.
Collapse
Affiliation(s)
- Hwajung Choi
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, 54896, South Korea
| | - Tak-Heun Kim
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, 54896, South Korea
| | - Ju-Kyeong Jeong
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, 54896, South Korea
| | - Charlotte Strandgren
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, Huddinge, SE-14183, Sweden
| | - Maria Eriksson
- Department of Biosciences and Nutrition, Center for Innovative Medicine, Karolinska Institutet, Huddinge, SE-14183, Sweden
| | - Eui-Sic Cho
- Cluster for Craniofacial Development and Regeneration Research, Institute of Oral Biosciences, Chonbuk National University School of Dentistry, Jeonju, 54896, South Korea.
| |
Collapse
|
33
|
Kim PH, Luu J, Heizer P, Tu Y, Weston TA, Chen N, Lim C, Li RL, Lin PY, Dunn JCY, Hodzic D, Young SG, Fong LG. Disrupting the LINC complex in smooth muscle cells reduces aortic disease in a mouse model of Hutchinson-Gilford progeria syndrome. Sci Transl Med 2018; 10:eaat7163. [PMID: 30257952 PMCID: PMC6166472 DOI: 10.1126/scitranslmed.aat7163] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 09/07/2018] [Indexed: 12/27/2022]
Abstract
Hutchinson-Gilford progeria syndrome is a disorder of premature aging in children caused by de novo mutations in LMNA that lead to the synthesis of an internally truncated form of prelamin A (commonly called progerin). The production of progerin causes multiple disease phenotypes, including an unusual vascular phenotype characterized by the loss of smooth muscle cells in the arterial media and fibrosis of the adventitia. We show that progerin expression, combined with mechanical stress, promotes smooth muscle cell death. Disrupting the linker of the nucleoskeleton and cytoskeleton (LINC) complex in smooth muscle cells ameliorates the toxic effects of progerin on smooth muscle cells and limits the accompanying adventitial fibrosis.
Collapse
Affiliation(s)
- Paul H Kim
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jennings Luu
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Patrick Heizer
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yiping Tu
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Thomas A Weston
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Natalie Chen
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Christopher Lim
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Robert L Li
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Po-Yu Lin
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - James C Y Dunn
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA 94305, USA
| | - Didier Hodzic
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stephen G Young
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Loren G Fong
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
34
|
Greer MM, Kleinman ME, Gordon LB, Massaro J, D'Agostino RB, Baltrusaitis K, Kieran MW, Gordon CM. Pubertal Progression in Female Adolescents with Progeria. J Pediatr Adolesc Gynecol 2018; 31:238-241. [PMID: 29258958 PMCID: PMC6671321 DOI: 10.1016/j.jpag.2017.12.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 12/05/2017] [Accepted: 12/09/2017] [Indexed: 12/25/2022]
Abstract
STUDY OBJECTIVE This study identified the prevalence of menarche and coincident sexual characteristics in female adolescents with Hutchinson-Gilford Progeria Syndrome (HGPS). DESIGN Data were examined to determine the prevalence of menarche in female adolescents older than 12 years; all were participants in clinical trials between 2007 and 2016. SETTING Pediatric hospital in Boston, Massachusetts. PARTICIPANTS Fifteen female adolescents, median age 15 (range, 12.0-20.3) years with a confirmed diagnosis of HGPS. INTERVENTIONS AND MAIN OUTCOME MEASURES Report of menarche, anthropometric and serum hormonal measures, Tanner pubertal staging, and body composition using dual-energy x-ray absorptiometry. RESULTS Nine of 15 (60%) participants reported spontaneous menarche at a median age of 14.4 years (range, 12.0-16.5 years). In those experiencing menarche vs not, median age was older (16.5 vs 13.6 years; P = .02), whereas body mass index did not differ (10.5 vs 10.4; P = .53) nor percentage body fat (19.4% vs. 19.3%; P = .98) or serum leptin levels (0.40 vs 0.40 ng/mL; P = .23). Among those who achieved menarche, 2 of 9 (22%) had Tanner II breast development and 2 of 9 (22%) exhibited Tanner II Pubic hair, all reflecting minimal pubertal development. Only early signs of puberty were similarly seen in the non-menstruating group, including 1 of 6 (17%) with Tanner II breasts and 2 of 6 (33%) with Tanner II pubic hair, and Tanner staging did not differ between those who reported menarche vs those who did not (each P = 1.0). None of the participants achieved Tanner IV or V thelarche over the course of the study. CONCLUSION Menarche was achieved in more than half of adolescent girls with HGPS, in the setting of little to no physical signs of pubertal development and minimal body fat.
Collapse
Affiliation(s)
- Maya Mundkur Greer
- Department of Anesthesia, Boston Children's Hospital, Boston, Massachusetts
| | - Monica E Kleinman
- Department of Anesthesia, Boston Children's Hospital, Boston, Massachusetts
| | - Leslie B Gordon
- Department of Anesthesia, Boston Children's Hospital, Boston, Massachusetts; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Joe Massaro
- Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, Massachusetts
| | - Ralph B D'Agostino
- Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, Massachusetts
| | - Kristin Baltrusaitis
- Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, Massachusetts
| | - Mark W Kieran
- Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Catherine M Gordon
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio.
| |
Collapse
|
35
|
Survey of plasma proteins in children with progeria pre-therapy and on-therapy with lonafarnib. Pediatr Res 2018; 83:982-992. [PMID: 29342131 DOI: 10.1038/pr.2018.9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 10/17/2017] [Indexed: 12/13/2022]
Abstract
BackgroundHutchinson-Gilford progeria syndrome (HGPS) is an ultra-rare, fatal, segmental premature aging syndrome caused by the aberrant lamin A protein, progerin. The protein farnesyltransferase inhibitor, lonafarnib, ameliorates some aspects of cardiovascular and bone disease.MethodsWe performed a prospective longitudinal survey of plasma proteins in 24 children with HGPS (an estimated 10% of the world's population at the time) at baseline and on lonafarnib therapy, compared with age- and gender-matched controls using a multi-analyte, microsphere-based immunofluorescent assay.ResultsThe mean levels for 23/66 (34.8%) proteins were significantly lower and 7/66 (10.6%) were significantly higher in HGPS samples compared with those in controls (P≤0.05). Six proteins whose concentrations were initially lower normalized with lonafarnib therapy: interleukins 1α, 7, and 13, beta-2 microglobulin, C-reactive protein, and myoglobin. Alpha-2 macroglobulin, a protease inhibitor associated with stroke, was elevated at baseline and subsequently normalized with lonafarnib therapy.ConclusionThis is the first study to employ a multi-analyte array platform in HGPS. Novel potential biomarkers identified in this study should be further validated by correlations with clinical disease status, especially proteins associated with cardiovascular disease and those that normalized with lonafarnib therapy.
Collapse
|
36
|
Emerging candidate treatment strategies for Hutchinson-Gilford progeria syndrome. Biochem Soc Trans 2017; 45:1279-1293. [PMID: 29127216 DOI: 10.1042/bst20170141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/25/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS, progeria) is an extremely rare premature aging disorder affecting children, with a disease incidence of ∼1 in 18 million individuals. HGPS is usually caused by a de novo point mutation in exon 11 of the LMNA gene (c.1824C>T, p.G608G), resulting in the increased usage of a cryptic splice site and production of a truncated unprocessed lamin A protein named progerin. Since the genetic cause for HGPS was published in 2003, numerous potential treatment options have rapidly emerged. Strategies to interfere with the post-translational processing of lamin A, to enhance progerin clearance, or directly target the HGPS mutation to reduce the progerin-producing alternative splicing of the LMNA gene have been developed. Here, we give an up-to-date resume of the contributions made by our and other research groups to the growing list of different candidate treatment strategies that have been tested, both in vitro, in vivo in mouse models for HGPS and in clinical trials in HGPS patients.
Collapse
|
37
|
Tariq Z, Zhang H, Chia-Liu A, Shen Y, Gete Y, Xiong ZM, Tocheny C, Campanello L, Wu D, Losert W, Cao K. Lamin A and microtubules collaborate to maintain nuclear morphology. Nucleus 2017; 8:433-446. [PMID: 28557611 DOI: 10.1080/19491034.2017.1320460] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Lamin A (LA) is a critical structural component of the nuclear lamina. Mutations within the LA gene (LMNA) lead to several human disorders, most striking of which is Hutchinson-Gilford Progeria Syndrome (HGPS), a premature aging disorder. HGPS cells are best characterized by an abnormal nuclear morphology known as nuclear blebbing, which arises due to the accumulation of progerin, a dominant mutant form of LA. The microtubule (MT) network is known to mediate changes in nuclear morphology in the context of specific events such as mitosis, cell polarization, nucleus positioning and cellular migration. What is less understood is the role of the microtubule network in determining nuclear morphology during interphase. In this study, we elucidate the role of the cytoskeleton in regulation and misregulation of nuclear morphology through perturbations of both the lamina and the microtubule network. We found that LA knockout cells exhibit a crescent shape morphology associated with the microtubule-organizing center. Furthermore, this crescent shape ameliorates upon treatment with MT drugs, Nocodazole or Taxol. Expression of progerin, in LA knockout cells also rescues the crescent shape, although the response to Nocodazole or Taxol treatment is altered in comparison to cells expressing LA. Together these results describe a collaborative effort between LA and the MT network to maintain nuclear morphology.
Collapse
Affiliation(s)
- Zeshan Tariq
- a Department of Cell Biology and Molecular Genetics , University of Maryland , College Park , MD , USA
| | - Haoyue Zhang
- a Department of Cell Biology and Molecular Genetics , University of Maryland , College Park , MD , USA
| | - Alexander Chia-Liu
- b Department of Physics , University of Maryland , College Park , MD , USA
| | - Yang Shen
- b Department of Physics , University of Maryland , College Park , MD , USA
| | - Yantenew Gete
- a Department of Cell Biology and Molecular Genetics , University of Maryland , College Park , MD , USA
| | - Zheng-Mei Xiong
- a Department of Cell Biology and Molecular Genetics , University of Maryland , College Park , MD , USA
| | - Claire Tocheny
- c Department of Biology , The College of William and Mary , Williamsburg , VA , USA
| | - Leonard Campanello
- b Department of Physics , University of Maryland , College Park , MD , USA
| | - Di Wu
- a Department of Cell Biology and Molecular Genetics , University of Maryland , College Park , MD , USA
| | - Wolfgang Losert
- b Department of Physics , University of Maryland , College Park , MD , USA
| | - Kan Cao
- a Department of Cell Biology and Molecular Genetics , University of Maryland , College Park , MD , USA
| |
Collapse
|
38
|
Bar DZ, Arlt MF, Brazier JF, Norris WE, Campbell SE, Chines P, Larrieu D, Jackson SP, Collins FS, Glover TW, Gordon LB. A novel somatic mutation achieves partial rescue in a child with Hutchinson-Gilford progeria syndrome. J Med Genet 2017; 54:212-216. [PMID: 27920058 PMCID: PMC5384422 DOI: 10.1136/jmedgenet-2016-104295] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 11/03/2016] [Accepted: 11/11/2016] [Indexed: 11/04/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome (HGPS) is a fatal sporadic autosomal dominant premature ageing disease caused by single base mutations that optimise a cryptic splice site within exon 11 of the LMNA gene. The resultant disease-causing protein, progerin, acts as a dominant negative. Disease severity relies partly on progerin levels. METHODS AND RESULTS We report a novel form of somatic mosaicism, where a child possessed two cell populations with different HGPS disease-producing mutations of the same nucleotide-one producing severe HGPS and one mild HGPS. The proband possessed an intermediate phenotype. The mosaicism was initially discovered when Sanger sequencing showed a c.1968+2T>A mutation in blood DNA and a c.1968+2T>C in DNA from cultured fibroblasts. Deep sequencing of DNA from the proband's blood revealed 4.7% c.1968+2T>C mutation, and 41.3% c.1968+2T>A mutation. CONCLUSIONS We hypothesise that the germline mutation was c.1968+2T>A, but a rescue event occurred during early development, where the somatic mutation from A to C at 1968+2 provided a selective advantage. This type of mosaicism where a partial phenotypic rescue event results from a second but milder disease-causing mutation in the same nucleotide has not been previously characterised for any disease.
Collapse
Affiliation(s)
- Daniel Z Bar
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Martin F Arlt
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Joan F Brazier
- Center for Gerontology and Health Care Research, Brown University, Providence, Rhode Island, USA
| | - Wendy E Norris
- Department of Pediatrics, Hasbro Children's Hospital, Providence, Rhode Island, USA
| | - Susan E Campbell
- Center for Gerontology and Health Care Research, Brown University, Providence, Rhode Island, USA
| | - Peter Chines
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Delphine Larrieu
- Department of Biochemistry, The Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Stephen P Jackson
- Department of Biochemistry, The Gurdon Institute, University of Cambridge, Cambridge, UK
| | - Francis S Collins
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Thomas W Glover
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, USA
| | - Leslie B Gordon
- Department of Pediatrics, Hasbro Children's Hospital, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
39
|
Gonzalo S, Kreienkamp R, Askjaer P. Hutchinson-Gilford Progeria Syndrome: A premature aging disease caused by LMNA gene mutations. Ageing Res Rev 2017; 33:18-29. [PMID: 27374873 DOI: 10.1016/j.arr.2016.06.007] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 06/25/2016] [Accepted: 06/28/2016] [Indexed: 01/08/2023]
Abstract
Products of the LMNA gene, primarily lamin A and C, are key components of the nuclear lamina, a proteinaceous meshwork that underlies the inner nuclear membrane and is essential for proper nuclear architecture. Alterations in lamin A and C that disrupt the integrity of the nuclear lamina affect a whole repertoire of nuclear functions, causing cellular decline. In humans, hundreds of mutations in the LMNA gene have been identified and correlated with over a dozen degenerative disorders, referred to as laminopathies. These diseases include neuropathies, muscular dystrophies, lipodystrophies, and premature aging diseases. This review focuses on one of the most severe laminopathies, Hutchinson-Gilford Progeria Syndrome (HGPS), which is caused by aberrant splicing of the LMNA gene and expression of a mutant product called progerin. Here, we discuss current views about the molecular mechanisms that contribute to the pathophysiology of this devastating disease, as well as the strategies being tested in vitro and in vivo to counteract progerin toxicity. In particular, progerin accumulation elicits nuclear morphological abnormalities, misregulated gene expression, defects in DNA repair, telomere shortening, and genomic instability, all of which limit cellular proliferative capacity. In patients harboring this mutation, a severe premature aging disease develops during childhood. Interestingly, progerin is also produced in senescent cells and cells from old individuals, suggesting that progerin accumulation might be a factor in physiological aging. Deciphering the molecular mechanisms whereby progerin expression leads to HGPS is an emergent area of research, which could bring us closer to understanding the pathology of aging.
Collapse
Affiliation(s)
- Susana Gonzalo
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | - Ray Kreienkamp
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA
| | - Peter Askjaer
- Andalusian Center for Developmental Biology (CABD), CSIC/Junta de Andalucia/Universidad Pablo de Olavide, Carretera de Utrera, Km 1, 41013 Seville, Spain
| |
Collapse
|
40
|
Gordon LB, Kleinman ME, Massaro J, D'Agostino RB, Shappell H, Gerhard-Herman M, Smoot LB, Gordon CM, Cleveland RH, Nazarian A, Snyder BD, Ullrich NJ, Silvera VM, Liang MG, Quinn N, Miller DT, Huh SY, Dowton AA, Littlefield K, Greer MM, Kieran MW. Clinical Trial of the Protein Farnesylation Inhibitors Lonafarnib, Pravastatin, and Zoledronic Acid in Children With Hutchinson-Gilford Progeria Syndrome. Circulation 2016; 134:114-25. [PMID: 27400896 DOI: 10.1161/circulationaha.116.022188] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/30/2016] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome is an extremely rare, fatal, segmental premature aging syndrome caused by a mutation in LMNA yielding the farnesylated aberrant protein progerin. Without progerin-specific treatment, death occurs at an average age of 14.6 years from an accelerated atherosclerosis. A previous single-arm clinical trial demonstrated that the protein farnesyltransferase inhibitor lonafarnib ameliorates some aspects of cardiovascular and bone disease. This present trial sought to further improve disease by additionally inhibiting progerin prenylation. METHODS Thirty-seven participants with Hutchinson-Gilford progeria syndrome received pravastatin, zoledronic acid, and lonafarnib. This combination therapy was evaluated, in addition to descriptive comparisons with the prior lonafarnib monotherapy trial. RESULTS No participants withdrew because of side effects. Primary outcome success was predefined by improved per-patient rate of weight gain or carotid artery echodensity; 71.0% of participants succeeded (P<0.0001). Key cardiovascular and skeletal secondary variables were predefined. Secondary improvements included increased areal (P=0.001) and volumetric (P<0.001-0.006) bone mineral density and 1.5- to 1.8-fold increases in radial bone structure (P<0.001). Median carotid artery wall echodensity and carotid-femoral pulse wave velocity demonstrated no significant changes. Percentages of participants with carotid (5% to 50%; P=0.001) and femoral (0% to 12%; P=0.13) artery plaques and extraskeletal calcifications (34.4% to 65.6%; P=0.006) increased. Other than increased bone mineral density, no improvement rates exceeded those of the prior lonafarnib monotherapy treatment trial. CONCLUSIONS Comparisons with lonafarnib monotherapy treatment reveal additional bone mineral density benefit but likely no added cardiovascular benefit with the addition of pravastatin and zoledronic acid. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique identifiers: NCT00879034 and NCT00916747.
Collapse
Affiliation(s)
- Leslie B Gordon
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.).
| | - Monica E Kleinman
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Joe Massaro
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Ralph B D'Agostino
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Heather Shappell
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Marie Gerhard-Herman
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Leslie B Smoot
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Catherine M Gordon
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Robert H Cleveland
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Ara Nazarian
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Brian D Snyder
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Nicole J Ullrich
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - V Michelle Silvera
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Marilyn G Liang
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Nicolle Quinn
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - David T Miller
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Susanna Y Huh
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Anne A Dowton
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Kelly Littlefield
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Maya M Greer
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.)
| | - Mark W Kieran
- From Departments of Anesthesia (L.B.G., M.E.K., A.A.D., K.L., M.M.G.), Cardiology (L.B.S.), Radiology (R.H.C., V.M.S.), Orthopedics (B.D.S.), Neurology (N.J.U.), Dermatology (M.G.L.), Genetics and Genomics (D.T.M.), Gastroenterology and Nutrition (S.Y.H.), and Hematology Oncology (M.W.K.), and Clinical Translational Study Unit (N.Q.), Boston Children's Hospital and Harvard Medical School, MA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Biostatistics, Boston University School of Public Health and Harvard Clinical Research Institute, MA (J.M., R.B.D., H.S.); Division of Cardiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA (M.G.-H.); Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati Children's Hospital Medical Center, OH (C.M.G.); Center for Advanced Orthopaedic Studies, Department of Orthopedic Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (A.N.); and Division of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA (M.W.K.).
| |
Collapse
|
41
|
Zhang H, Sun L, Wang K, Wu D, Trappio M, Witting C, Cao K. Loss of H3K9me3 Correlates with ATM Activation and Histone H2AX Phosphorylation Deficiencies in Hutchinson-Gilford Progeria Syndrome. PLoS One 2016; 11:e0167454. [PMID: 27907109 PMCID: PMC5131972 DOI: 10.1371/journal.pone.0167454] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/14/2016] [Indexed: 12/20/2022] Open
Abstract
Compelling evidence suggests that defective DNA damage response (DDR) plays a key role in the premature aging phenotypes in Hutchinson-Gilford progeria syndrome (HGPS). Studies document widespread alterations in histone modifications in HGPS cells, especially, the global loss of histone H3 trimethylated on lysine 9 (H3K9me3). In this study, we explore the potential connection(s) between H3K9me3 loss and the impaired DDR in HGPS. When cells are exposed to a DNA-damaging agent Doxorubicin (Dox), double strand breaks (DSBs) are generated that result in the phosphorylation of histone H2A variant H2AX (gammaH2AX) within an hour. We find that the intensities of gammaH2AX foci appear significantly weaker in the G0/G1 phase HGPS cells compared to control cells. This reduction is associated with a delay in the recruitment of essential DDR factors. We further demonstrate that ataxia-telangiectasia mutated (ATM) is responsible for the amplification of gammaH2AX signals at DSBs during G0/G1 phase, and its activation is inhibited in the HGPS cells that display significant loss of H3K9me3. Moreover, methylene (MB) blue treatment, which is known to save heterochromatin loss in HGPS, restores H3K9me3, stimulates ATM activity, increases gammaH2AX signals and rescues deficient DDR. In summary, this study demonstrates an early DDR defect of attenuated gammaH2AX signals in G0/G1 phase HGPS cells and provides a plausible connection between H3K9me3 loss and DDR deficiency.
Collapse
Affiliation(s)
- Haoyue Zhang
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Linlin Sun
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Kun Wang
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States of America
| | - Di Wu
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Mason Trappio
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Celeste Witting
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD, United States of America
- * E-mail:
| |
Collapse
|
42
|
Xiong Z, Choi JY, Wang K, Zhang H, Tariq Z, Wu D, Ko E, LaDana C, Sesaki H, Cao K. Methylene blue alleviates nuclear and mitochondrial abnormalities in progeria. Aging Cell 2016; 15:279-90. [PMID: 26663466 PMCID: PMC4783354 DOI: 10.1111/acel.12434] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2015] [Indexed: 12/17/2022] Open
Abstract
Hutchinson–Gilford progeria syndrome (HGPS), a fatal premature aging disease, is caused by a single‐nucleotide mutation in the LMNA gene. Previous reports have focused on nuclear phenotypes in HGPS cells, yet the potential contribution of the mitochondria, a key player in normal aging, remains unclear. Using high‐resolution microscopy analysis, we demonstrated a significantly increased fraction of swollen and fragmented mitochondria and a marked reduction in mitochondrial mobility in HGPS fibroblast cells. Notably, the expression of PGC‐1α, a central regulator of mitochondrial biogenesis, was inhibited by progerin. To rescue mitochondrial defects, we treated HGPS cells with a mitochondrial‐targeting antioxidant methylene blue (MB). Our analysis indicated that MB treatment not only alleviated the mitochondrial defects but also rescued the hallmark nuclear abnormalities in HGPS cells. Additional analysis suggested that MB treatment released progerin from the nuclear membrane, rescued perinuclear heterochromatin loss and corrected misregulated gene expression in HGPS cells. Together, these results demonstrate a role of mitochondrial dysfunction in developing the premature aging phenotypes in HGPS cells and suggest MB as a promising therapeutic approach for HGPS.
Collapse
Affiliation(s)
- Zheng‐Mei Xiong
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Ji Young Choi
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Kun Wang
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
- Center for Bioinformatics and Computational Biology University of Maryland College Park MD 20742 USA
| | - Haoyue Zhang
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Zeshan Tariq
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Di Wu
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Eunae Ko
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Christina LaDana
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| | - Hiromi Sesaki
- Department of Cell Biology Johns Hopkins University School of Medicine Baltimore MD 21205 USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics University of Maryland College Park MD 20742 USA
| |
Collapse
|
43
|
Swahari V, Nakamura A. Speeding up the clock: The past, present and future of progeria. Dev Growth Differ 2015; 58:116-30. [DOI: 10.1111/dgd.12251] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 10/15/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Vijay Swahari
- Neuroscience Center; University of North Carolina; Chapel Hill North Carolina USA
| | - Ayumi Nakamura
- Neuroscience Center; University of North Carolina; Chapel Hill North Carolina USA
| |
Collapse
|
44
|
Strandgren C, Nasser HA, McKenna T, Koskela A, Tuukkanen J, Ohlsson C, Rozell B, Eriksson M. Transgene silencing of the Hutchinson-Gilford progeria syndrome mutation results in a reversible bone phenotype, whereas resveratrol treatment does not show overall beneficial effects. FASEB J 2015; 29:3193-205. [PMID: 25877214 DOI: 10.1096/fj.14-269217] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/31/2015] [Indexed: 11/11/2022]
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder that is most commonly caused by a de novo point mutation in exon 11 of the LMNA gene, c.1824C>T, which results in an increased production of a truncated form of lamin A known as progerin. In this study, we used a mouse model to study the possibility of recovering from HGPS bone disease upon silencing of the HGPS mutation, and the potential benefits from treatment with resveratrol. We show that complete silencing of the transgenic expression of progerin normalized bone morphology and mineralization already after 7 weeks. The improvements included lower frequencies of rib fractures and callus formation, an increased number of osteocytes in remodeled bone, and normalized dentinogenesis. The beneficial effects from resveratrol treatment were less significant and to a large extent similar to mice treated with sucrose alone. However, the reversal of the dental phenotype of overgrown and laterally displaced lower incisors in HGPS mice could be attributed to resveratrol. Our results indicate that the HGPS bone defects were reversible upon suppressed transgenic expression and suggest that treatments targeting aberrant progerin splicing give hope to patients who are affected by HGPS.
Collapse
Affiliation(s)
- Charlotte Strandgren
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hasina Abdul Nasser
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Tomás McKenna
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Antti Koskela
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juha Tuukkanen
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Claes Ohlsson
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Björn Rozell
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Eriksson
- *Department of Biosciences and Nutrition, Center for Innovative Medicine, and Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden; Department of Anatomy and Cell Biology, University of Oulu, Oulu, Finland; Centre for Bone and Arthritis Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; and Department of Experimental Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
45
|
Chu Y, Xu ZG, Xu Z, Ma L. Hutchinson-Gilford progeria syndrome caused by an LMNA mutation: a case report. Pediatr Dermatol 2015; 32:271-5. [PMID: 25556323 DOI: 10.1111/pde.12406] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Hutchinson-Gilford progeria syndrome is a rare genetic disorder characterized by premature aging of the skin, bones, heart, and blood vessels. We report a 6-year-old boy who was born at full term but presented with scleroderma-like appearance at 1 month of age and gradually developed clinical manifestations of progeria. He had characteristic facial features of prominent eyes, scalp, and leg veins; loss of scalp hair, eyebrows, and eyelashes; stunted growth; scleroderma-like changes of the skin; and a premature aged appearance. Metabolic investigations showed transient methylmalonic aciduria, and genetic testing of the peripheral blood identified the c.1824C>T heterozygous LMNA mutation. The present case is reported because of its rarity.
Collapse
Affiliation(s)
- Yan Chu
- Department of Dermatology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | | | | | | |
Collapse
|
46
|
Yang SH, Procaccia S, Jung HJ, Nobumori C, Tatar A, Tu Y, Bayguinov YR, Hwang SJ, Tran D, Ward SM, Fong LG, Young SG. Mice that express farnesylated versions of prelamin A in neurons develop achalasia. Hum Mol Genet 2015; 24:2826-40. [PMID: 25652409 DOI: 10.1093/hmg/ddv043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Accepted: 02/02/2015] [Indexed: 12/15/2022] Open
Abstract
Neurons in the brain produce lamin C but almost no lamin A, a consequence of the removal of prelamin A transcripts by miR-9, a brain-specific microRNA. We have proposed that miR-9-mediated regulation of prelamin A in the brain could explain the absence of primary neurological disease in Hutchinson-Gilford progeria syndrome, a genetic disease caused by the synthesis of an internally truncated form of farnesyl-prelamin A (progerin). This explanation makes sense, but it is not entirely satisfying because it is unclear whether progerin-even if were expressed in neurons-would be capable of eliciting neuropathology. To address that issue, we created a new Lmna knock-in allele, Lmna(HG-C), which produces progerin transcripts lacking an miR-9 binding site. Mice harboring the Lmna(HG-C) allele produced progerin in neurons, but they had no pathology in the central nervous system. However, these mice invariably developed esophageal achalasia, and the enteric neurons and nerve fibers in gastrointestinal tract were markedly abnormal. The same disorder, achalasia, was observed in genetically modified mice that express full-length farnesyl-prelamin A in neurons (Zmpste24-deficient mice carrying two copies of a Lmna knock-in allele yielding full-length prelamin A transcripts lacking a miR-9 binding site). Our findings indicate that progerin and full-length farnesyl-prelamin A are toxic to neurons of the enteric nervous system.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yulia R Bayguinov
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | - Stephen G Young
- Department of Medicine, Molecular Biology Institute and Department of Human Genetics, University of California, Los Angeles, CA 90095, USA and
| |
Collapse
|
47
|
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare, uniformly fatal, segmental "premature aging" disease in which children exhibit phenotypes that may give us insights into the aging process at both the cellular and organismal levels. Initial presentation in early childhood is primarily based on growth and dermatologic findings. Primary morbidity and mortality for children with HGPS is from atherosclerotic cardiovascular disease and strokes with death occurring at an average age of 14.6 years. There is increasing data to support a unique phenotype of the craniofacial and cerebrovascular anatomy that accompanies the premature aging process. Strokes in HGPS can occur downstream of carotid artery and/or vertebral artery occlusion, stenosis, and calcification, with prominent collateral vessel formation. Both large and small vessel disease are present, and strokes are often clinically silent. Despite the presence of multisystem premature aging, children with HGPS do not appear to have cognitive deterioration, suggesting that some aspects of brain function may be protected from the deleterious effects of progerin, the disease-causing protein. Based on limited autopsy material, there is no pathologic evidence of dementia or Alzheimer-type changes. In a transgenic mouse model of progeria with expression of the most common HGPS mutation in brain, skin, bone, and heart, there are distortions of neuronal nuclei at the ultrastructural level with irregular shape and severe invaginations, but no evidence of inclusions or aberrant tau in brain sections. Importantly, the nuclear distortions did not result in significant changes in gene expression in hippocampal neurons. This chapter will discuss both preclinical and clinical aspects of the genetics, pathobiology, clinical phenotype, clinical care, and treatment of HGPS, with special attention toward neurologic and cutaneous findings.
Collapse
Affiliation(s)
- Nicole J Ullrich
- Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Leslie B Gordon
- Department of Anesthesia, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI, USA; Progeria Research Foundation, Peabody, MA, USA.
| |
Collapse
|
48
|
Gordon A, Gordon L. The Progeria Research Foundation: its remarkable journey from obscurity to treatment. Expert Opin Orphan Drugs 2014. [DOI: 10.1517/21678707.2014.970172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
49
|
Reichert C, Gölz L, Götz W, Wolf M, Deschner J, Jäger A. Dental and craniofacial characteristics in a patient with Hutchinson-Gilford progeria syndrome. J Orofac Orthop 2014; 75:251-63. [PMID: 25001855 DOI: 10.1007/s00056-014-0216-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2012] [Accepted: 01/31/2013] [Indexed: 11/28/2022]
Abstract
The Hutchinson-Gilford progeria syndrome (HGPS) is an exceptionally rare medical disorder caused by mutations in the lamin A/C gene. Affected patients display typical features of premature aging. Beside general medical disorders, these patients have several specific features related to the craniofacial phenotype and the oral cavity. In this article, the dental and craniofacial characteristics of a 9-year-old girl with HGPS are presented. It is the first report addressing orthodontic tooth movement and microbiological features in a HGPS patient. We describe and discuss pathologic findings and provide a detailed histology of the teeth which had to be extracted during initial treatment.
Collapse
Affiliation(s)
- Christoph Reichert
- Department of Orthodontics, Dental Hospital, University of Bonn, Welschnonnenstr. 17, 53111, Bonn, Germany,
| | | | | | | | | | | |
Collapse
|
50
|
Gordon LB, Massaro J, D'Agostino RB, Campbell SE, Brazier J, Brown WT, Kleinman ME, Kieran MW. Impact of farnesylation inhibitors on survival in Hutchinson-Gilford progeria syndrome. Circulation 2014; 130:27-34. [PMID: 24795390 PMCID: PMC4082404 DOI: 10.1161/circulationaha.113.008285] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 04/15/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Hutchinson-Gilford progeria syndrome is an ultrarare segmental premature aging disease resulting in early death from heart attack or stroke. There is no approved treatment, but starting in 2007, several recent single-arm clinical trials administered inhibitors of protein farnesylation aimed at reducing toxicity of the disease-producing protein progerin. No study assessed whether treatments influence patient survival. The key elements necessary for this analysis are a robust natural history of survival and comparison with a sufficiently large patient population that has been treated for a sufficient time period with disease-targeting medications. METHODS AND RESULTS We generated Kaplan-Meier survival analyses for the largest untreated Hutchinson-Gilford progeria syndrome cohort to date. Mean survival was 14.6 years. Comparing survival for treated versus age- and sex-matched untreated cohorts, hazard ratio was 0.13 (95% confidence interval, 0.04-0.37; P<0.001) with median follow-up of 5.3 years from time of treatment initiation. There were 21 of 43 deaths in untreated versus 5 of 43 deaths among treated subjects. Treatment increased mean survival by 1.6 years. CONCLUSIONS This study provides a robust untreated disease survival profile that can be used for comparisons now and in the future to assess changes in survival with treatments for Hutchinson-Gilford progeria syndrome. The current comparisons estimating increased survival with protein farnesylation inhibitors provide the first evidence of treatments influencing survival for this fatal disease. CLINICAL TRIAL REGISTRATION URL http://www.clinicaltrials.gov. Unique Indentifiers: NCT00425607, NCT00879034, and NCT00916747.
Collapse
Affiliation(s)
- Leslie B Gordon
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.).
| | - Joe Massaro
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| | - Ralph B D'Agostino
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| | - Susan E Campbell
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| | - Joan Brazier
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| | - W Ted Brown
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| | - Monica E Kleinman
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| | - Mark W Kieran
- From the Department of Pediatrics, Hasbro Children's Hospital and Warren Alpert Medical School of Brown University, Providence, RI (L.B.G.); Department of Anesthesia, Division of Critical Care Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA (L.B.G., M.E.K.); Department of Mathematics and Statistics, Boston University, Harvard Clinical Research Institute, Boston, MA (J.M., R.B.D.); Center for Gerontology and Health Care Research, Brown University, Providence, RI (S.E.C., J.B.); Department of Genetics, New York State Institute for Basic Research, Staten Island, NY (W.T.B.); Hematology-Oncology, Boston Children's Hospital, Division of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA (M.W.K.)
| |
Collapse
|