Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Mech Ageing Dev ; 216: 111879, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37832833

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.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Cardiovascular System , Progeria , Humans , Adolescent , Progeria/genetics , Progeria/therapy , Progeria/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/therapy , Aging/metabolism , Atherosclerosis/pathology , Cardiovascular System/metabolism
2.
Arch Med Res ; 54(5): 102837, 2023 07.
Article in English | MEDLINE | ID: mdl-37390702

ABSTRACT

In humans, aging is characterized by a gradual decline of physical and psychological functions, with the concomitant onset of chronic-degenerative diseases, which ultimately lead to death. The study of Hutchinson-Gilford progeria syndrome (HGPS), a premature aging disorder that recapitulates several features of natural aging, has provided important insights into deciphering the aging process. The genetic origin of HGPS is a de novo point mutation in the LMNA gene that drives the synthesis of progerin, mutant version of lamin A. Progerin is aberrantly anchored to the nuclear envelope disrupting a plethora of molecular processes; nonetheless, how progerin exerts a cascade of deleterious alterations at the cellular and systemic levels is not fully understood. Over the past decade, the use of different cellular and animal models for HGPS has allowed the identification of the molecular mechanisms underlying HGPS, paving the way towards the development of therapeutic treatments against the disease. In this review, we present an updated overview of the biology of HGPS, including its clinical features, description of key cellular processes affected by progerin (nuclear morphology and function, nucleolar activity, mitochondrial function, protein nucleocytoplasmic trafficking and telomere homeostasis), as well as discussion of the therapeutic strategies under development.


Subject(s)
Progeria , Animals , Humans , Progeria/therapy , Progeria/drug therapy , Aging , Mitochondria/metabolism
3.
Cell Reprogram ; 25(4): 136-138, 2023 08.
Article in English | MEDLINE | ID: mdl-37327373

ABSTRACT

A recent study in Aging Cell showed that transcriptional activation of endogenous Oct4 using the CRISPR/dCas9 activator system is sufficient for cellular rejuvenation and extending the lifespan of a progeria mouse model. Although transient expression of reprogramming factors Oct4, Sox2, Klf4, and c-Myc (OSKM) has been shown to ameliorate age-related phenotypes in vivo, oncogenic risk, for example, from c-Myc, has raised safety concerns for its use in therapeutics. The authors demonstrated that transient activation of endogenous Oct4 expression restored age-related epigenetic patterns, suppressed expression of mutant progerin, and reduced vascular pathological features associated with the disease. At the same time, the transient Oct4 overexpression resulted in lower incidence of cancer transformation compared with constituent OSKM overexpression. Successful activation of endogenous Oct4 by CRISPR/dCas9 paves the way for novel therapeutic approaches for the treatment of progeria and age-related diseases, with potential implications for the broader field of cellular reprogramming-based rejuvenation.


Subject(s)
Progeria , Mice , Animals , Progeria/genetics , Progeria/therapy , Progeria/metabolism , Rejuvenation , Clustered Regularly Interspaced Short Palindromic Repeats , Cellular Reprogramming , Disease Models, Animal
4.
Genes (Basel) ; 14(3)2023 02 27.
Article in English | MEDLINE | ID: mdl-36980874

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare, autosomal-dominant, and fatal premature aging syndrome. HGPS is most often derived from a de novo point mutation in the LMNA gene, which results in an alternative splicing defect and the generation of the mutant protein, progerin. Progerin behaves in a dominant-negative fashion, leading to a variety of cellular and molecular changes, including nuclear abnormalities, defective DNA damage response (DDR) and DNA repair, and accelerated telomere attrition. Intriguingly, many of the manifestations of the HGPS cells are shared with normal aging cells. However, at a clinical level, HGPS does not fully match normal aging because of the accelerated nature of the phenotypes and its primary effects on connective tissues. Furthermore, the epigenetic changes in HGPS patients are of great interest and may play a crucial role in the pathogenesis of HGPS. Finally, various treatments for the HGPS patients have been developed in recent years with important effects at a cellular level, which translate to symptomatic improvement and increased lifespan.


Subject(s)
Progeria , Humans , Progeria/genetics , Progeria/therapy , Progeria/metabolism , Cellular Senescence/genetics , Cell Nucleus/genetics , Epigenesis, Genetic
5.
Nat Commun ; 13(1): 3068, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35654881

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature ageing disorder caused by a point mutation in the LMNA gene (LMNA c.1824 C > T), resulting in the production of a detrimental protein called progerin. Adenine base editors recently emerged with a promising potential for HGPS gene therapy. However adeno-associated viral vector systems currently used in gene editing raise concerns, and the long-term effects of heterogeneous mutation correction in highly proliferative tissues like the skin are unknown. Here we use a non-integrative transient lentiviral vector system, expressing an adenine base editor to correct the HGPS mutation in the skin of HGPS mice. Transient adenine base editor expression corrected the mutation in 20.8-24.1% of the skin cells. Four weeks post delivery, the HGPS skin phenotype was improved and clusters of progerin-negative keratinocytes were detected, indicating that the mutation was corrected in both progenitor and differentiated skin cells. These results demonstrate that transient non-integrative viral vector mediated adenine base editor expression is a plausible approach for future gene-editing therapies.


Subject(s)
Progeria , Adenine , Animals , Lamin Type A/genetics , Lamin Type A/metabolism , Mice , Mutation , Phenotype , Progeria/genetics , Progeria/metabolism , Progeria/therapy
6.
J Atheroscler Thromb ; 29(4): 439-447, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-34511576

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) and Werner syndrome (WS) are two of the representative genetic progeroid syndromes and have been widely studied in the field of aging research. HGPS is a pediatric disease in which premature aging symptoms appear in early childhood, and death occurs at an average age of 14.5 years, mainly due to cardiovascular disease (CVD). Conversely, WS patients exhibit accelerated aging phenotypes after puberty and die in their 50s due to CVD and malignant tumors. Both diseases are models of human aging, leading to a better understanding of the aging-associated development of CVD. In this review, we discuss the pathogenesis and treatment of atherosclerotic diseases presented by both progeroid syndromes with the latest findings.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Progeria , Werner Syndrome , Aging , Atherosclerosis/genetics , Cardiovascular Diseases/genetics , Child, Preschool , Humans , Progeria/genetics , Progeria/therapy , Werner Syndrome/complications , Werner Syndrome/genetics , Werner Syndrome/therapy
7.
Int J Mol Sci ; 22(22)2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34830197

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.


Subject(s)
Atherosclerosis/complications , Atherosclerosis/therapy , Cord Blood Stem Cell Transplantation/adverse effects , Dyslipidemias/complications , Dyslipidemias/therapy , Progeria/complications , Progeria/therapy , Adolescent , Body Height , Follow-Up Studies , Humans , Male , Pulse Wave Analysis , Range of Motion, Articular , Transplantation, Homologous/adverse effects , Treatment Outcome , Weight Gain
8.
Eur Heart J ; 42(42): 4352-4369, 2021 11 07.
Article in English | MEDLINE | ID: mdl-34389865

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.


Subject(s)
Progeria , Telomerase , Animals , Cellular Senescence/genetics , Endothelial Cells/metabolism , Humans , Longevity , Mice , Progeria/genetics , Progeria/therapy , Proteomics , Telomerase/genetics
9.
Int J Mol Sci ; 22(13)2021 Jul 03.
Article in English | MEDLINE | ID: mdl-34281245

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS), or progeria, is an extremely rare disorder that belongs to the class of laminopathies, diseases characterized by alterations in the genes that encode for the lamin proteins or for their associated interacting proteins. In particular, progeria is caused by a point mutation in the gene that codifies for the lamin A gene. This mutation ultimately leads to the biosynthesis of a mutated version of lamin A called progerin, which accumulates abnormally in the nuclear lamina. This accumulation elicits several alterations at the nuclear, cellular, and tissue levels that are phenotypically reflected in a systemic disorder with important alterations, mainly in the cardiovascular system, bones, skin, and overall growth, which results in premature death at an average age of 14.5 years. In 2020, lonafarnib became the first (and only) FDA approved drug for treating progeria. In this context, the present review focuses on the different therapeutic strategies currently under development, with special attention to the new small molecules described in recent years, which may represent the upcoming first-in-class drugs with new mechanisms of action endowed with effectiveness not only to treat but also to cure progeria.


Subject(s)
Piperidines/therapeutic use , Progeria/therapy , Pyridines/therapeutic use , Aging/genetics , Aging, Premature/genetics , Cell Nucleus/metabolism , Cellular Senescence/genetics , Fibroblasts/metabolism , Humans , Lamin Type A/genetics , Laminopathies/therapy , Mutation , Nuclear Lamina/genetics , Nuclear Lamina/physiology , Phenotype , Progeria/genetics , Progeria/metabolism , Skin/metabolism , Small Molecule Libraries/pharmacology
11.
Med Sci (Paris) ; 37(4): 413-416, 2021 Apr.
Article in French | MEDLINE | ID: mdl-33908864

ABSTRACT

Hutchinson-Gilford Progeria (acute premature aging) is caused by a de novo point mutation in the lamin A gene. Recently, this mutation has been accurately corrected by base editing in patient cell lines and in a mouse model, resulting in nearly complete reversal to a normal phenotype. This success opens the perspective for clinical applications in Progeria and other diseases.


Subject(s)
Lamin Type A/genetics , Point Mutation , Progeria/therapy , Targeted Gene Repair , Animals , Disease Models, Animal , Fluorescent Antibody Technique , Gene Editing/methods , Humans , Lamin Type A/metabolism , Mice , Progeria/genetics
13.
Curr Gene Ther ; 21(3): 216-229, 2021.
Article in English | MEDLINE | ID: mdl-33655857

ABSTRACT

Lamin A/C encoded by the LMNA gene is an essential component for maintaining the nuclear structure. Mutation in the lamin A/C leads to a group of inherited disorders is known as laminopathies. In the human body, there are several mutations in the LMNA gene that have been identified. It can affect diverse organs or tissues or can be systemic, causing different diseases. In this review, we mainly focused on one of the most severe laminopathies, Hutchinson-Gilford progeria syndrome (HGPS). HGPS is an immensely uncommon, deadly, metameric ill-timed laminopathies caused by the abnormal splicing of the LMNA gene and production of an aberrant protein known as progerin. Here, we also presented the currently available data on the molecular mechanism, pathophysiology, available treatment, and future approaches to this deadly disease. Due to the production of progerin, an abnormal protein leads to an abnormality in nuclear structure, defects in DNA repair, shortening of telomere, and impairment in gene regulation which ultimately results in aging in the early stage of life. Now some treatment options are available for this disease, but a proper understanding of the molecular mechanism of this disease will help to develop a more appropriate treatment which makes it an emerging area of research.


Subject(s)
Gene Expression Regulation , Lamin Type A/physiology , Progeria/physiopathology , Progeria/therapy , Cell Nucleus/metabolism , DNA Repair , Gene Silencing , Humans , Mutation , Phenotype , Telomere/physiology
15.
Nature ; 589(7843): 608-614, 2021 01.
Article in English | MEDLINE | ID: mdl-33408413

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS or progeria) is typically caused by a dominant-negative C•G-to-T•A mutation (c.1824 C>T; p.G608G) in LMNA, the gene that encodes nuclear lamin A. This mutation causes RNA mis-splicing that produces progerin, a toxic protein that induces rapid ageing and shortens the lifespan of children with progeria to approximately 14 years1-4. Adenine base editors (ABEs) convert targeted A•T base pairs to G•C base pairs with minimal by-products and without requiring double-strand DNA breaks or donor DNA templates5,6. Here we describe the use of an ABE to directly correct the pathogenic HGPS mutation in cultured fibroblasts derived from children with progeria and in a mouse model of HGPS. Lentiviral delivery of the ABE to fibroblasts from children with HGPS resulted in 87-91% correction of the pathogenic allele, mitigation of RNA mis-splicing, reduced levels of progerin and correction of nuclear abnormalities. Unbiased off-target DNA and RNA editing analysis did not detect off-target editing in treated patient-derived fibroblasts. In transgenic mice that are homozygous for the human LMNA c.1824 C>T allele, a single retro-orbital injection of adeno-associated virus 9 (AAV9) encoding the ABE resulted in substantial, durable correction of the pathogenic mutation (around 20-60% across various organs six months after injection), restoration of normal RNA splicing and reduction of progerin protein levels. In vivo base editing rescued the vascular pathology of the mice, preserving vascular smooth muscle cell counts and preventing adventitial fibrosis. A single injection of ABE-expressing AAV9 at postnatal day 14 improved vitality and greatly extended the median lifespan of the mice from 215 to 510 days. These findings demonstrate the potential of in vivo base editing as a possible treatment for HGPS and other genetic diseases by directly correcting their root cause.


Subject(s)
Adenine/metabolism , Gene Editing/methods , Mutation , Progeria/genetics , Progeria/therapy , Alleles , Alternative Splicing , Animals , Aorta/pathology , Base Pairing , Child , DNA/genetics , Disease Models, Animal , Female , Fibroblasts/metabolism , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type A/metabolism , Longevity , Male , Mice , Mice, Transgenic , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Progeria/pathology , RNA/genetics
16.
Pak J Pharm Sci ; 33(3): 1179-1182, 2020 May.
Article in English | MEDLINE | ID: mdl-33191246

ABSTRACT

Hutchinson-Gilford Progeria syndrome (or Progeria) is an exceptionally rare genetic disorder in children. It is caused by a rare point mutation in the lamin gene. It encodes lamin A protein, resulting in the de-shaping of nuclear membrane. This altered structure of the nuclear membrane renders the nucleus unstable. The shortened lifespan of the nucleus makes the cell liable for rapid ageing. Children are healthy by appearance when they are born but the signs appear after 12-24 months of age. Cardiovascular system is greatly affected which became a reason for the death of most of the patients of progeria. Stiffened joints disturb the bone movements; and alopecia affects the appearance of the patient. Rate of occurrence of the disease is one per four hundred thousand of people, though both sexes are equally affected.


Subject(s)
Aging, Premature , Lamin Type A/genetics , Point Mutation , Progeria/therapy , Female , Genetic Predisposition to Disease , Humans , Male , Phenotype , Progeria/genetics , Progeria/pathology , Progeria/physiopathology , Prognosis
17.
Aging Cell ; 19(7): e13175, 2020 07.
Article in English | MEDLINE | ID: mdl-32596971

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is an autosomal-dominant genetic disease that leads to accelerated aging and often premature death caused by cardiovascular complications. Till now clinical management of HGPS has largely relied on the treatment of manifestations and on the prevention of secondary complications, cure for the disease has not yet been established. Addressing this need cannot only benefit progeria patients but may also provide insights into intervention design for combating physiological aging. By using the systematic review approach, this article revisits the overall progress in the development of strategies for HGPS treatment over the last ten years, from 2010 to 2019. In total, 1,906 articles have been retrieved, of which 56 studies have been included for further analysis. Based on the articles analyzed, the trends in the use of different HGPS models, along with the prevalence, efficiency, and limitations of different reported treatment strategies, have been examined. Emerging strategies for preclinical studies, and possible targets for intervention development, have also been presented as avenues for future research.


Subject(s)
Progeria/therapy , Disease Progression , Humans , Syndrome
18.
Cell ; 181(1): 19, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32243789

ABSTRACT

We asked three researchers how their personal connection to disease has affected them and what lessons it has taught them along the way.


Subject(s)
Castleman Disease/drug therapy , Celiac Disease/drug therapy , Progeria/therapy , Drug Development , Drug Repositioning , Humans
19.
Sci Adv ; 6(8): eaay5556, 2020 02.
Article in English | MEDLINE | ID: mdl-32128409

ABSTRACT

Vascular dysfunction is a typical characteristic of aging, but its contributing roles to systemic aging and the therapeutic potential are lacking experimental evidence. Here, we generated a knock-in mouse model with the causative Hutchinson-Gilford progeria syndrome (HGPS) LmnaG609G mutation, called progerin. The Lmnaf/f ;TC mice with progerin expression induced by Tie2-Cre exhibit defective microvasculature and neovascularization, accelerated aging, and shortened life span. Single-cell transcriptomic analysis of murine lung endothelial cells revealed a substantial up-regulation of inflammatory response. Molecularly, progerin interacts and destabilizes deacylase Sirt7; ectopic expression of Sirt7 alleviates the inflammatory response caused by progerin in endothelial cells. Vascular endothelium-targeted Sirt7 gene therapy, driven by an ICAM2 promoter, improves neovascularization, ameliorates aging features, and extends life span in Lmnaf/f ;TC mice. These data support endothelial dysfunction as a primary trigger of systemic aging and highlight gene therapy as a potential strategy for the clinical treatment of HGPS and age-related vascular dysfunction.


Subject(s)
Endothelium, Vascular/metabolism , Genetic Therapy , Longevity , Progeria/genetics , Progeria/metabolism , Sirtuins/genetics , Animals , Cellular Senescence , Disease Models, Animal , Endothelial Cells , Gene Expression Profiling , Genetic Therapy/methods , Humans , Longevity/genetics , Mice , Mice, Knockout , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Progeria/therapy , Single-Cell Analysis , Vasodilation
SELECTION OF CITATIONS
SEARCH DETAIL
...