Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.692
Filter
Add more filters

Publication year range
1.
Cell ; 184(2): 293, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33482093

ABSTRACT

The ultra-rare, pediatric premature aging disorder Hutchinson-Gilford progeria syndrome (HGPS) is caused by mutation of LMNA, encoding the nuclear architectural protein lamin A. Patients develop atherosclerosis and typically die of heart failure in their teens. FDA-approved Zokinvy prevents farnesylation of lamin A, reduces vascular stiffness, and extends survival in HGPS patients. To view this Bench to Bedside, open or download the PDF.


Subject(s)
Enzyme Inhibitors/therapeutic use , Farnesyltranstransferase/antagonists & inhibitors , Progeria/drug therapy , Progeria/enzymology , Enzyme Inhibitors/pharmacology , Farnesyltranstransferase/metabolism , Humans , Molecular Targeted Therapy
2.
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
3.
Cell ; 165(6): 1312-1313, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259142

ABSTRACT

Progerin, a mutated lamin A, causes the severe premature-aging syndrome Hutchinson-Gilford progeria (HGPS). Kubben et al. present a driving mechanism for HGPS involving trapping of NRF2 at the nuclear periphery by progerin. This local restriction results in impaired NRF2 signaling and chronic oxidative stress.


Subject(s)
Lamin Type A/metabolism , Progeria/metabolism , Cell Nucleus/metabolism , Humans , Nuclear Proteins/metabolism , Oxidative Stress , Protein Precursors/metabolism
4.
Cell ; 165(6): 1361-1374, 2016 Jun 02.
Article in English | MEDLINE | ID: mdl-27259148

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare, invariably fatal premature aging disorder. The disease is caused by constitutive production of progerin, a mutant form of the nuclear architectural protein lamin A, leading, through unknown mechanisms, to diverse morphological, epigenetic, and genomic damage and to mesenchymal stem cell (MSC) attrition in vivo. Using a high-throughput siRNA screen, we identify the NRF2 antioxidant pathway as a driver mechanism in HGPS. Progerin sequesters NRF2 and thereby causes its subnuclear mislocalization, resulting in impaired NRF2 transcriptional activity and consequently increased chronic oxidative stress. Suppressed NRF2 activity or increased oxidative stress is sufficient to recapitulate HGPS aging defects, whereas reactivation of NRF2 activity in HGPS patient cells reverses progerin-associated nuclear aging defects and restores in vivo viability of MSCs in an animal model. These findings identify repression of the NRF2-mediated antioxidative response as a key contributor to the premature aging phenotype.


Subject(s)
Aging, Premature/metabolism , Antioxidants/metabolism , NF-E2-Related Factor 2/metabolism , Progeria/metabolism , Aging, Premature/genetics , Cell Line , Cell Survival , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , NF-E2-Related Factor 2/genetics , Progeria/genetics , RNA, Small Interfering , Transcription Factors/metabolism , Transcription, Genetic
5.
Cell ; 165(3): 566-79, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27087445

ABSTRACT

Hepatic glucose release into the circulation is vital for brain function and survival during periods of fasting and is modulated by an array of hormones that precisely regulate plasma glucose levels. We have identified a fasting-induced protein hormone that modulates hepatic glucose release. It is the C-terminal cleavage product of profibrillin, and we name it Asprosin. Asprosin is secreted by white adipose, circulates at nanomolar levels, and is recruited to the liver, where it activates the G protein-cAMP-PKA pathway, resulting in rapid glucose release into the circulation. Humans and mice with insulin resistance show pathologically elevated plasma asprosin, and its loss of function via immunologic or genetic means has a profound glucose- and insulin-lowering effect secondary to reduced hepatic glucose release. Asprosin represents a glucogenic protein hormone, and therapeutically targeting it may be beneficial in type II diabetes and metabolic syndrome.


Subject(s)
Fasting/metabolism , Microfilament Proteins/metabolism , Peptide Fragments/metabolism , Peptide Hormones/metabolism , Adipose Tissue, White/metabolism , Amino Acid Sequence , Animals , Antibodies/administration & dosage , Circadian Rhythm , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Fasting/blood , Female , Fetal Growth Retardation/metabolism , Fibrillin-1 , Glucose/metabolism , Humans , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microfilament Proteins/blood , Microfilament Proteins/chemistry , Microfilament Proteins/genetics , Molecular Sequence Data , Peptide Fragments/blood , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Hormones/blood , Peptide Hormones/chemistry , Peptide Hormones/genetics , Progeria/metabolism , Recombinant Proteins/administration & dosage , Sequence Alignment
6.
Annu Rev Biochem ; 84: 131-64, 2015.
Article in English | MEDLINE | ID: mdl-25747401

ABSTRACT

Lamins are intermediate filament proteins that form a scaffold, termed nuclear lamina, at the nuclear periphery. A small fraction of lamins also localize throughout the nucleoplasm. Lamins bind to a growing number of nuclear protein complexes and are implicated in both nuclear and cytoskeletal organization, mechanical stability, chromatin organization, gene regulation, genome stability, differentiation, and tissue-specific functions. The lamin-based complexes and their specific functions also provide insights into possible disease mechanisms for human laminopathies, ranging from muscular dystrophy to accelerated aging, as observed in Hutchinson-Gilford progeria and atypical Werner syndromes.


Subject(s)
Cell Nucleus/metabolism , Lamins/metabolism , Animals , Cell Nucleus/chemistry , Cell Nucleus/genetics , Chromatin/chemistry , Chromatin/metabolism , Gene Expression Regulation , Humans , Lamins/chemistry , Lamins/genetics , Progeria/pathology
7.
Nat Rev Mol Cell Biol ; 18(10): 595-609, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28792007

ABSTRACT

Ageing is the predominant risk factor for many common diseases. Human premature ageing diseases are powerful model systems to identify and characterize cellular mechanisms that underpin physiological ageing. Their study also leads to a better understanding of the causes, drivers and potential therapeutic strategies of common diseases associated with ageing, including neurological disorders, diabetes, cardiovascular diseases and cancer. Using the rare premature ageing disorder Hutchinson-Gilford progeria syndrome as a paradigm, we discuss here the shared mechanisms between premature ageing and ageing-associated diseases, including defects in genetic, epigenetic and metabolic pathways; mitochondrial and protein homeostasis; cell cycle; and stem cell-regenerative capacity.


Subject(s)
Aging, Premature/metabolism , Aging, Premature/pathology , Aging/metabolism , Aging/pathology , Aging/genetics , Aging, Premature/genetics , Animals , DNA Repair , Epigenesis, Genetic , Genomic Instability , Humans , Progeria/genetics , Progeria/metabolism , Progeria/pathology
8.
Cell ; 156(3): 400-7, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24485450

ABSTRACT

Rare diseases are powerful windows into biological processes and can serve as models for the development of therapeutic strategies. The progress made on the premature aging disorder Progeria is a shining example of the impact that studies of rare diseases can have.


Subject(s)
Progeria/drug therapy , Progeria/physiopathology , Translational Research, Biomedical , Aging/genetics , Aging/pathology , Child , Farnesyltranstransferase/antagonists & inhibitors , Humans , Lamin Type A , Nuclear Proteins/metabolism , Progeria/genetics , Progeria/pathology , Protein Precursors/metabolism
9.
EMBO J ; 42(1): e110937, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36382717

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a lethal premature aging disorder without an effective therapeutic regimen. Because of their targetability and influence on gene expression, microRNAs (miRNAs) are attractive therapeutic tools to treat diseases. Here we identified that hsa-miR-59 (miR-59) was markedly upregulated in HGPS patient cells and in multiple tissues of an HGPS mouse model (LmnaG609G/G609G ), which disturbed the interaction between RNAPII and TFIIH, resulting in abnormal expression of cell cycle genes by targeting high-mobility group A family HMGA1 and HMGA2. Functional inhibition of miR-59 alleviated the cellular senescence phenotype of HGPS cells. Treatment with AAV9-mediated anti-miR-59 reduced fibrosis in the quadriceps muscle, heart, and aorta, suppressed epidermal thinning and dermal fat loss, and yielded a 25.5% increase in longevity of LmnaG609G/G609G mice. These results identify a new strategy for the treatment of HGPS and provide insight into the etiology of HGPS disease.


Subject(s)
MicroRNAs , Progeria , Mice , Animals , Progeria/genetics , Antagomirs/therapeutic use , Cellular Senescence/genetics , MicroRNAs/genetics , Phenotype
10.
Cell ; 148(6): 1085-8, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424220

ABSTRACT

Dysregulation of splicing and alternative splicing underlies many genetic and acquired diseases. We present an overview of recent strategies and successes in modulating splicing therapeutically in clinical and preclinical contexts. Effective approaches include restoring open reading frames, influencing alternative splicing, and inducing exon inclusion to generate beneficial proteins and remove deleterious ones.


Subject(s)
Disease/genetics , Genetic Therapy , RNA Splicing/drug effects , Alternative Splicing , Animals , Humans , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Mutation , Neoplasms/genetics , Neoplasms/therapy , Oligonucleotides, Antisense/therapeutic use , Progeria/genetics , Progeria/therapy
11.
Cell ; 149(3): 565-77, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541428

ABSTRACT

Human LMNA gene mutations result in laminopathies that include Emery-Dreifuss muscular dystrophy (AD-EDMD) and Hutchinson-Gilford progeria, the premature aging syndrome (HGPS). The Lmna null (Lmna(-/-)) and progeroid LmnaΔ9 mutant mice are models for AD-EDMD and HGPS, respectively. Both animals develop severe tissue pathologies with abbreviated life spans. Like HGPS cells, Lmna(-/-) and LmnaΔ9 fibroblasts have typically misshapen nuclei. Unexpectedly, Lmna(-/-) or LmnaΔ9 mice that are also deficient for the inner nuclear membrane protein Sun1 show markedly reduced tissue pathologies and enhanced longevity. Concordantly, reduction of SUN1 overaccumulation in LMNA mutant fibroblasts and in cells derived from HGPS patients corrected nuclear defects and cellular senescence. Collectively, these findings implicate Sun1 protein accumulation as a common pathogenic event in Lmna(-/-), LmnaΔ9, and HGPS disorders.


Subject(s)
Lamin Type A/metabolism , Membrane Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Muscular Dystrophy, Emery-Dreifuss/metabolism , Muscular Dystrophy, Emery-Dreifuss/pathology , Nuclear Proteins/metabolism , Progeria/metabolism , Animals , Cell Line , Cellular Senescence , Disease Models, Animal , Fibroblasts/metabolism , Humans , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Progeria/pathology
12.
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
13.
Proc Natl Acad Sci U S A ; 121(18): e2400752121, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38648484

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a rare disease caused by the expression of progerin, a mutant protein that accelerates aging and precipitates death. Given that atherosclerosis complications are the main cause of death in progeria, here, we investigated whether progerin-induced atherosclerosis is prevented in HGPSrev-Cdh5-CreERT2 and HGPSrev-SM22α-Cre mice with progerin suppression in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), respectively. HGPSrev-Cdh5-CreERT2 mice were undistinguishable from HGPSrev mice with ubiquitous progerin expression, in contrast with the ameliorated progeroid phenotype of HGPSrev-SM22α-Cre mice. To study atherosclerosis, we generated atheroprone mouse models by overexpressing a PCSK9 gain-of-function mutant. While HGPSrev-Cdh5-CreERT2 and HGPSrev mice developed a similar level of excessive atherosclerosis, plaque development in HGPSrev-SM22α-Cre mice was reduced to wild-type levels. Our studies demonstrate that progerin suppression in VSMCs, but not in ECs, prevents exacerbated atherosclerosis in progeroid mice.


Subject(s)
Atherosclerosis , Endothelial Cells , Lamin Type A , Muscle, Smooth, Vascular , Progeria , Animals , Mice , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Lamin Type A/metabolism , Lamin Type A/genetics , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Progeria/metabolism , Progeria/genetics , Progeria/pathology , Proprotein Convertase 9/metabolism , Proprotein Convertase 9/genetics
14.
Proc Natl Acad Sci U S A ; 121(27): e2406946121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917015

ABSTRACT

Progerin, the protein that causes Hutchinson-Gilford progeria syndrome, triggers nuclear membrane (NM) ruptures and blebs, but the mechanisms are unclear. We suspected that the expression of progerin changes the overall structure of the nuclear lamina. High-resolution microscopy of smooth muscle cells (SMCs) revealed that lamin A and lamin B1 form independent meshworks with uniformly spaced openings (~0.085 µm2). The expression of progerin in SMCs resulted in the formation of an irregular meshwork with clusters of large openings (up to 1.4 µm2). The expression of progerin acted in a dominant-negative fashion to disrupt the morphology of the endogenous lamin B1 meshwork, triggering irregularities and large openings that closely resembled the irregularities and openings in the progerin meshwork. These abnormal meshworks were strongly associated with NM ruptures and blebs. Of note, the progerin meshwork was markedly abnormal in nuclear blebs that were deficient in lamin B1 (~50% of all blebs). That observation suggested that higher levels of lamin B1 expression might normalize the progerin meshwork and prevent NM ruptures and blebs. Indeed, increased lamin B1 expression reversed the morphological abnormalities in the progerin meshwork and markedly reduced the frequency of NM ruptures and blebs. Thus, progerin expression disrupts the overall structure of the nuclear lamina, but that effect-along with NM ruptures and blebs-can be abrogated by increased lamin B1 expression.


Subject(s)
Lamin Type A , Lamin Type B , Nuclear Lamina , Nuclear Lamina/metabolism , Lamin Type A/metabolism , Lamin Type A/genetics , Lamin Type B/metabolism , Lamin Type B/genetics , Humans , Progeria/metabolism , Progeria/genetics , Progeria/pathology , Animals , Protein Precursors/metabolism , Protein Precursors/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Mice
15.
Proc Natl Acad Sci U S A ; 121(41): e2321378121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39352925

ABSTRACT

Progerin causes Hutchinson-Gilford progeria syndrome (HGPS), but how progerin accelerates aging is still an interesting question. Here, we provide evidence linking nuclear envelope (NE) budding and accelerated aging. Mechanistically, progerin disrupts nuclear lamina to induce NE budding in concert with lamin A/C, resulting in transport of chromatin into the cytoplasm where it is removed via autophagy, whereas emerin antagonizes this process. Primary cells from both HGPS patients and mouse models express progerin and display NE budding and chromatin loss, and ectopically expressing progerin in cells can mimic this process. More excitingly, we screen a NE budding inhibitor chaetocin by high-throughput screening, which can dramatically sequester progerin from the NE and prevent this NE budding through sustaining ERK1/2 activation. Chaetocin alleviates NE budding-induced chromatin loss and ameliorates HGPS defects in cells and mice and significantly extends lifespan of HGPS mice. Collectively, we propose that progerin-induced NE budding participates in the induction of progeria, highlight the roles of chaetocin and sustained ERK1/2 activation in anti-aging, and provide a distinct avenue for treating HGPS.


Subject(s)
Lamin Type A , Nuclear Envelope , Nuclear Proteins , Progeria , Progeria/metabolism , Progeria/drug therapy , Progeria/pathology , Progeria/genetics , Animals , Lamin Type A/metabolism , Lamin Type A/genetics , Mice , Humans , Nuclear Envelope/metabolism , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Aging/metabolism , Aging/drug effects , Chromatin/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Disease Models, Animal , Autophagy/drug effects
16.
Proc Natl Acad Sci U S A ; 120(47): e2309200120, 2023 Nov 21.
Article in English | MEDLINE | ID: mdl-37967221

ABSTRACT

Patients with Hutchinson-Gilford progeria syndrome (HGPS) present with a number of premature aging phenotypes, including DNA damage accumulation, and many of them die of cardiovascular complications. Although vascular pathologies have been reported, whether HGPS patients exhibit cardiac dysfunction and its underlying mechanism is unclear, rendering limited options for treating HGPS-related cardiomyopathy. In this study, we reported a cardiac atrophy phenotype in the LmnaG609G/G609G mice (hereafter, HGPS mice). Using a GFP-based reporter system, we demonstrated that the efficiency of nonhomologous end joining (NHEJ) declined by 50% in HGPS cardiomyocytes in vivo, due to the attenuated interaction between γH2AX and Progerin, the causative factor of HGPS. As a result, genomic instability in cardiomyocytes led to an increase of CHK2 protein level, promoting the LKB1-AMPKα interaction and AMPKα phosphorylation, which further led to the activation of FOXO3A-mediated transcription of atrophy-related genes. Moreover, inhibiting AMPK enlarged cardiomyocyte sizes both in vitro and in vivo. Most importantly, our proof-of-concept study indicated that isoproterenol treatment significantly reduced AMPKα and FOXO3A phosphorylation in the heart, attenuated the atrophy phenotype, and extended the mean lifespan of HGPS mice by ~21%, implying that targeting cardiac atrophy may be an approach to HGPS treatment.


Subject(s)
Aging, Premature , Progeria , Humans , Mice , Animals , Progeria/metabolism , Heart , DNA Damage , Genomic Instability , AMP-Activated Protein Kinases/genetics , Lamin Type A/genetics , Lamin Type A/metabolism
17.
EMBO J ; 40(6): e107165, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33619770

ABSTRACT

Mitochondria contain an autonomous and spatially segregated genome. The organizational unit of their genome is the nucleoid, which consists of mitochondrial DNA (mtDNA) and associated architectural proteins. Here, we show that phase separation is the primary physical mechanism for assembly and size control of the mitochondrial nucleoid (mt-nucleoid). The major mtDNA-binding protein TFAM spontaneously phase separates in vitro via weak, multivalent interactions into droplets with slow internal dynamics. TFAM and mtDNA form heterogenous, viscoelastic structures in vitro, which recapitulate the dynamics and behavior of mt-nucleoids in vivo. Mt-nucleoids coalesce into larger droplets in response to various forms of cellular stress, as evidenced by the enlarged and transcriptionally active nucleoids in mitochondria from patients with the premature aging disorder Hutchinson-Gilford Progeria Syndrome (HGPS). Our results point to phase separation as an evolutionarily conserved mechanism of genome organization.


Subject(s)
DNA, Mitochondrial/genetics , Genome, Mitochondrial/genetics , Mitochondria/genetics , Progeria/pathology , Cell Line , Child , Child, Preschool , DNA-Binding Proteins/metabolism , HEK293 Cells , Humans , Mitochondrial Proteins/metabolism , Progeria/genetics , Transcription Factors/metabolism
18.
Nat Rev Genet ; 20(5): 299-309, 2019 05.
Article in English | MEDLINE | ID: mdl-30760854

ABSTRACT

Many recent advances have emerged in the telomere and telomerase fields. This Timeline article highlights the key advances that have expanded our views on the mechanistic underpinnings of telomeres and telomerase and their roles in ageing and disease. Three decades ago, the classic view was that telomeres protected the natural ends of linear chromosomes and that telomerase was a specific telomere-terminal transferase necessary for the replication of chromosome ends in single-celled organisms. While this concept is still correct, many diverse fields associated with telomeres and telomerase have substantially matured. These areas include the discovery of most of the key molecular components of telomerase, implications for limits to cellular replication, identification and characterization of human genetic disorders that result in premature telomere shortening, the concept that inhibiting telomerase might be a successful therapeutic strategy and roles for telomeres in regulating gene expression. We discuss progress in these areas and conclude with challenges and unanswered questions in the field.


Subject(s)
Aging/genetics , Genomics/history , Neoplasms/genetics , Telomerase/genetics , Telomere/chemistry , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/pathology , Aging/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA/chemistry , DNA/genetics , DNA/metabolism , Gene Expression Regulation , Genomics/methods , History, 20th Century , History, 21st Century , Humans , Molecular Chaperones , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Progeria/genetics , Progeria/metabolism , Progeria/pathology , Ribonucleoproteins, Small Nuclear/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Shelterin Complex , Telomerase/metabolism , Telomere/metabolism , Telomere Homeostasis , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism
19.
Mol Ther ; 32(9): 3101-3113, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39095992

ABSTRACT

Osteoarthritis (OA) pain is often associated with the expression of tumor necrosis factor alpha (TNF-α), suggesting that TNF-α is one of the main contributing factors that cause inflammation, pain, and OA pathology. Thus, inhibition of TNF-α could potentially improve OA symptoms and slow disease progression. Anti-TNF-α treatments with antibodies, however, require multiple treatments and cannot entirely block TNF-α. TNF-α-induced protein 8-like 2 (TIPE2) was found to regulate the immune system's homeostasis and inflammation through different mechanisms from anti-TNF-α therapies. With a single treatment of adeno-associated virus (AAV)-TIPE2 gene delivery in the accelerated aging Zmpste24-/- (Z24-/-) mouse model, we found differences in Safranin O staining intensity within the articular cartilage (AC) region of the knee between TIPE2-treated mice and control mice. The glycosaminoglycan content (orange-red) was degraded in the Z24-/- cartilage while shown to be restored in the TIPE2-treated Z24-/- cartilage. We also observed that chondrocytes in Z24-/- mice exhibited a variety of senescent-associated phenotypes. Treatment with TIPE2 decreased TNF-α-positive cells, ß-galactosidase (ß-gal) activity, and p16 expression seen in Z24-/- mice. Our study demonstrated that AAV-TIPE2 gene delivery effectively blocked TNF-α-induced inflammation and senescence, resulting in the prevention or delay of knee OA in our accelerated aging Z24-/- mouse model.


Subject(s)
Cellular Senescence , Dependovirus , Disease Models, Animal , Genetic Therapy , Inflammation , Intracellular Signaling Peptides and Proteins , Osteoarthritis , Progeria , Animals , Mice , Osteoarthritis/therapy , Osteoarthritis/genetics , Osteoarthritis/metabolism , Osteoarthritis/etiology , Osteoarthritis/pathology , Cellular Senescence/genetics , Inflammation/genetics , Inflammation/metabolism , Inflammation/therapy , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Genetic Therapy/methods , Progeria/genetics , Progeria/therapy , Progeria/metabolism , Dependovirus/genetics , Aging , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Chondrocytes/metabolism , Mice, Knockout , Tumor Necrosis Factor-alpha/metabolism , Humans
20.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35197292

ABSTRACT

Prelamin A is a farnesylated precursor of lamin A, a nuclear lamina protein. Accumulation of the farnesylated prelamin A variant progerin, with an internal deletion including its processing site, causes Hutchinson-Gilford progeria syndrome. Loss-of-function mutations in ZMPSTE24, which encodes the prelamin A processing enzyme, lead to accumulation of full-length farnesylated prelamin A and cause related progeroid disorders. Some data suggest that prelamin A also accumulates with physiological aging. Zmpste24-/- mice die young, at ∼20 wk. Because ZMPSTE24 has functions in addition to prelamin A processing, we generated a mouse model to examine effects solely due to the presence of permanently farnesylated prelamin A. These mice have an L648R amino acid substitution in prelamin A that blocks ZMPSTE24-catalyzed processing to lamin A. The LmnaL648R/L648R mice express only prelamin and no mature protein. Notably, nearly all survive to 65 to 70 wk, with ∼40% of male and 75% of female LmnaL648R/L648R mice having near-normal lifespans of 90 wk (almost 2 y). Starting at ∼10 wk of age, LmnaL648R/L648R mice of both sexes have lower body masses than controls. By ∼20 to 30 wk of age, they exhibit detectable cranial, mandibular, and dental defects similar to those observed in Zmpste24-/- mice and have decreased vertebral bone density compared to age- and sex-matched controls. Cultured embryonic fibroblasts from LmnaL648R/L648R mice have aberrant nuclear morphology that is reversible by treatment with a protein farnesyltransferase inhibitor. These novel mice provide a model to study the effects of farnesylated prelamin A during physiological aging.


Subject(s)
Lamin Type A/metabolism , Longevity , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Progeria/genetics , Animals , Binding Sites , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mice , Mutation , Phenotype , Prenylation
SELECTION OF CITATIONS
SEARCH DETAIL