Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
1.
Life Sci ; 341: 122489, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38340979

ABSTRACT

Lamins are inner nuclear membrane proteins that belong to the intermediate filament family. Lamin A/C lie adjacent to the heterochromatin structure in polymer form, providing skeletal to the nucleus. Based on the localization, lamin A/C provides nuclear stability and cytoskeleton to the nucleus and modulates chromatin organization and gene expression. Besides being the structural protein making the inner nuclear membrane in polymer form, lamin A/C functions as a signalling molecule involved in gene expression as an enhancer inside the nucleus. Lamin A/C regulates various cellular pathways like autophagy and energy balance in the cytoplasm. Its expression is highly variable in differentiated tissues, higher in hard tissues like bone and muscle cells, and lower in soft tissues like the liver and brain. In muscle cells, including the heart, lamin A/C must be expressed in a balanced state. Lamin A/C mutation is linked with various diseases, such as muscular dystrophy, lipodystrophy, and cardiomyopathies. It has been observed that a good number of mutations in the LMNA gene impact cardiac activity and its function. Although several works have been published, there are still several unexplored areas left regarding the lamin A/C function and structure in the cardiovascular system and its pathological state. In this review, we focus on the structural organization, expression pattern, and function of lamin A/C, its interacting partners, and the pathophysiology associated with mutations in the lamin A/C gene, with special emphasis on cardiovascular diseases. With the recent finding on lamin A/C, we have summarized the possible therapeutic interventions to treat cardiovascular symptoms and reverse the molecular changes.


Subject(s)
Cardiomyopathies , Muscular Dystrophies , Humans , Lamin Type A/genetics , Lamin Type A/chemistry , Lamin Type A/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Mutation , Polymers
2.
Biol Cell ; 116(3): e2300094, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38404031

ABSTRACT

BACKGROUND INFORMATION: Lamins are type V intermediate filament proteins underlying the inner nuclear membrane which provide structural rigidity to the nucleus, tether the chromosomes, maintain nuclear homeostasis, and remain dynamically associated with developmentally regulated regions of the genome. A large number of mutations particularly in the LMNA gene encoding lamin A/C results in a wide array of human diseases, collectively termed as laminopathies. Dilated Cardiomyopathy (DCM) is one such laminopathic cardiovascular disease which is associated with systolic dysfunction of left or both ventricles leading to cardiac arrhythmia which ultimately culminates into myocardial infarction. RESULTS: In this work, we have unraveled the epigenetic landscape to address the regulation of gene expression in mouse myoblast cell line in the context of the missense mutation LMNA 289A

Subject(s)
Cardiomyopathy, Dilated , Animals , Humans , Mice , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/metabolism , Lamin Type A/genetics , Lamin Type A/chemistry , Lamin Type A/metabolism , Mutation , NF-kappa B/genetics , NF-kappa B/metabolism , Nuclear Lamina
3.
Biochem Biophys Res Commun ; 637: 210-217, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36403485

ABSTRACT

Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disorder caused by C-terminally truncated lamin A, termed as the pre-progerin product. Progerin is a C-terminally farnesylated protein derived from pre-progerin, which causes nuclear deformation at the inner-nuclear membrane. As an alternative or additional mechanism, a farnesylation-independent abnormal interaction between the C-terminus of progerin and Ig-like domain has been proposed. However, the molecular mechanism underlying the role of unfarnesylated C-terminus of pre-progerin in HGPS remains largely unknown. In this study, we determined the crystal structures of C-terminal peptide of progerin and Ig-like domain of lamin A/C. Results showed that the C-terminal cysteine residue of progerin forms a disulfide bond with the only cysteine residue of the Ig-like domain. This finding suggested that unfarnesylated progerin can form a disulfide bond with the Ig-like domain in the lamin meshwork. The Alphafold2-assisted docking structure showed that disulfide bond formation was promoted by a weak interaction between the groove of Ig-like domain and the unfarnesylated C-terminal tail region of progerin. Our results provide molecular insights into the normal aging process as well as premature aging of humans.


Subject(s)
Aging, Premature , Lamin Type A , Progeria , Humans , Aging, Premature/genetics , Cysteine , Disulfides , Immunoglobulin Domains , Lamin Type A/chemistry , Progeria/genetics
4.
J Biol Chem ; 298(9): 102256, 2022 09.
Article in English | MEDLINE | ID: mdl-35839855

ABSTRACT

Nuclear lamins maintain the nuclear envelope structure by forming long linear filaments via two alternating molecular arrangements of coiled-coil dimers, known as A11 and A22 binding modes. The A11 binding mode is characterized by the antiparallel interactions between coil 1b domains, whereas the A22 binding mode is facilitated by interactions between the coil 2 domains of lamin. The junction between A11- and A22-interacting dimers in the lamin tetramer produces another parallel head-tail interaction between coil 1a and the C-terminal region of coil 2, called the ACN interaction. During mitosis, phosphorylation in the lamin N-terminal head region by the cyclin-dependent kinase (CDK) complex triggers depolymerization of lamin filaments, but the associated mechanisms remain unknown at the molecular level. In this study, we revealed using the purified proteins that phosphorylation by the CDK1 complex promotes disassembly of lamin filaments by directly abolishing the ACN interaction between coil 1a and the C-terminal portion of coil 2. We further observed that this interaction was disrupted as a result of alteration of the ionic interactions between coil 1a and coil 2. Combined with molecular modeling, we propose a mechanism for CDK1-dependent disassembly of the lamin filaments. Our results will help to elucidate the cell cycle-dependent regulation of nuclear morphology at the molecular level.


Subject(s)
CDC2 Protein Kinase , Intermediate Filaments , Lamin Type A , CDC2 Protein Kinase/chemistry , Humans , Intermediate Filaments/chemistry , Lamin Type A/chemistry , Polymerization , Protein Domains
5.
Cell Death Dis ; 12(8): 786, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34381017

ABSTRACT

Ovarian cancer is the most malignant gynecologic cancer. Previous studies found that lamin-A was associated with DNA damage repair proteins but the underlying mechanism remains unclear. We speculate that this may be related to its interacting proteins, such as Hsp90. The aim of this study is to investigate the effects of Hsp90 on DNA damage repair and chemoresistance of ovarian cancer cells. In our research, co-immunoprecipitation (co-IP) and mass spectrometry (MS) were used to identify proteins interacting with lamin-A and the interaction domain. Next, the relationship between lamin-A and Hsp90 was explored by Western blotting (WB) and immunofluorescence staining. Then, effect of Hsp90 inhibition on DNA damage repair was assessed through detecting Rad50 and Ku80 by WB. Furthermore, to test the roles of 17-AAG on cell chemosensitivity, CCK-8 and colony formation assay were carried out. Meanwhile, IC50 of cells were calculated, followed by immunofluorescence to detect DNA damage. At last, the mouse xenograft model was used in determining the capacity of 17-AAG and DDP to suppress tumor growth and metastatic potential. The results showed that lamin-A could interact with Hsp90 via the domain of lamin-A1-430. Besides, the distribution of Hsp90 could be affected by lamin-A. After lamin-A knockdown, Hsp90 decreased in the cytoplasm and increased in the nucleus, suggesting that the interaction between lamin-A and Hsp90 may be related to the nucleocytoplasmic transport of Hsp90. Moreover, inhibition of Hsp90 led to an obvious decrease in the expression of DSBs (DNA double-strand break) repair proteins, as well as cell proliferation ability upon DDP treatment and IC50 of DDP, causing more serious DNA damage. In addition, the combination of 17-AAG and DDP restrained the growth of ovarian cancer efficiently in vivo and prolonged the survival time of tumor-bearing mice.


Subject(s)
DNA Damage , DNA Repair , Drug Resistance, Neoplasm , HSP90 Heat-Shock Proteins/metabolism , Lamin Type A/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Animals , Benzoquinones/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/drug effects , DNA Repair/drug effects , Drug Resistance, Neoplasm/drug effects , Female , Gene Knockdown Techniques , Humans , Lactams, Macrocyclic/pharmacology , Lamin Type A/chemistry , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Protein Binding/drug effects , Protein Domains , Survival Analysis
6.
Biochem Biophys Res Commun ; 550: 191-196, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33706103

ABSTRACT

Lamins are nuclear intermediate filament proteins that play an essential role in maintaining the nuclear structure by forming a 3-D meshwork. Lamins consist of the N-terminal unstructured head, the coiled-coil rod domain, and the C-terminal tail, which is mostly unstructured except for the Ig-like domain. To date, the Ig-like domain has been characterized as a monomeric structure. Here, we determined the crystal structures of human lamin A/C, including the Ig-like domain and its N- and C-terminal flanking sequences. Interestingly, the structures showed a homodimer formed by beta-strand interactions between the N- and C-terminal flanking sequences. This interaction also provides a molecular implication for the creation of a 3-D meshwork between the 3.5-nm-thick filaments. Furthermore, we determined the crystal structure of the corresponding region of lamin B1. The structure showed a similar dimeric assembly, also formed by beta-strand interactions, albeit the intersubunit distance was much shorter. Since the Ig-like domain contains many genetic hotspots causing lamin-related diseases in lamin A/C, our findings will help understand the detailed assembly of lamins in a 3-D meshwork structure and lamin-related diseases at the molecular level.


Subject(s)
Immunoglobulin Domains , Lamin Type A/chemistry , Lamin Type A/metabolism , Lamin Type B/chemistry , Lamin Type B/metabolism , Protein Multimerization , Crystallography, X-Ray , Humans , Models, Molecular , Protein Stability
7.
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
8.
Eur J Med Genet ; 64(2): 104138, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33422685

ABSTRACT

Mandibuloacral dysplasia with type A lipodystrophy is a rare autosomal recessive disorder characterized by craniofacial dysmorphism, type A lipodystrophy, clavicular dysplasia, and acroostelolysis. It is caused by homozygous or compound heterozygous missense mutations in LMNA gene. We report five Tunisian patients harboring the same homozygous c.1580G > A; p. (Arg527His) mutation in LMNA gene. The patients presented with typical features of mandibuloacral dysplasia including, prominent eyes, thin or beaked nose, dental overcrowding, mandibular hypoplasia, short and broad finger's distal phalanges with round tips and lipodystrophy type A. Newly recognized signs are growth hormone deficiency and dilated cardiomyopathy. Genotype-phenotype correlation found that at least one of the disease's LMNA mutant alleles involve one of the highly conserved aminoacids, residing in a key site domain for protein function within the C-terminal globular domain of A-type lamins. Also, the severity of the disease depends on the position in the protein's domain and on the type of substitution of the concerned aminoacid.


Subject(s)
Acro-Osteolysis/genetics , Lamin Type A/genetics , Lipodystrophy/genetics , Mandible/abnormalities , Acro-Osteolysis/pathology , Adolescent , Child , Female , Humans , Lamin Type A/chemistry , Lipodystrophy/pathology , Male , Mandible/pathology , Mutation, Missense , Pedigree , Phenotype , Protein Domains
9.
J Biol Chem ; 296: 100165, 2021.
Article in English | MEDLINE | ID: mdl-33293369

ABSTRACT

The integral membrane zinc metalloprotease ZMPSTE24 is important for human health and longevity. ZMPSTE24 performs a key proteolytic step in maturation of prelamin A, the farnesylated precursor of the nuclear scaffold protein lamin A. Mutations in the genes encoding either prelamin A or ZMPSTE24 that prevent cleavage cause the premature aging disease Hutchinson-Gilford progeria syndrome (HGPS) and related progeroid disorders. ZMPSTE24 has a novel structure, with seven transmembrane spans that form a large water-filled membrane chamber whose catalytic site faces the chamber interior. Prelamin A is the only known mammalian substrate for ZMPSTE24; however, the basis of this specificity remains unclear. To define the sequence requirements for ZMPSTE24 cleavage, we mutagenized the eight residues flanking the prelamin A scissile bond (TRSY↓LLGN) to all other 19 amino acids, creating a library of 152 variants. We also replaced these eight residues with sequences derived from putative ZMPSTE24 cleavage sites from amphibian, bird, and fish prelamin A. Cleavage of prelamin A variants was assessed using an in vivo yeast assay that provides a sensitive measure of ZMPSTE24 processing efficiency. We found that residues on the C-terminal side of the cleavage site are most sensitive to changes. Consistent with other zinc metalloproteases, including thermolysin, ZMPSTE24 preferred hydrophobic residues at the P1' position (Leu647), but in addition, showed a similar, albeit muted, pattern at P2'. Our findings begin to define a consensus sequence for ZMPSTE24 that helps to clarify how this physiologically important protease functions and may ultimately lead to identifying additional substrates.


Subject(s)
Lamin Type A/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Metalloendopeptidases/chemistry , Metalloendopeptidases/metabolism , Zinc/metabolism , Amino Acid Sequence , Catalytic Domain , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Membrane Proteins/genetics , Metalloendopeptidases/genetics , Mutation , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity
10.
Cells ; 9(7)2020 07 07.
Article in English | MEDLINE | ID: mdl-32645958

ABSTRACT

The molecular architecture and assembly mechanism of intermediate filaments have been enigmatic for decades. Among those, lamin filaments are of particular interest due to their universal role in cell nucleus and numerous disease-related mutations. Filament assembly is driven by specific interactions of the elementary dimers, which consist of the central coiled-coil rod domain flanked by non-helical head and tail domains. We aimed to investigate the longitudinal 'head-to-tail' interaction of lamin dimers (the so-called ACN interaction), which is crucial for filament assembly. To this end, we prepared a series of recombinant fragments of human lamin A centred around the N- and C-termini of the rod. The fragments were stabilized by fusions to heterologous capping motifs which provide for a correct formation of parallel, in-register coiled-coil dimers. As a result, we established crystal structures of two N-terminal fragments one of which highlights the propensity of the coiled-coil to open up, and one C-terminal rod fragment. Additional studies highlighted the capacity of such N- and C-terminal fragments to form specific complexes in solution, which were further characterized using chemical cross-linking. These data yielded a molecular model of the ACN complex which features a 6.5 nm overlap of the rod ends.


Subject(s)
Lamin Type A/chemistry , Crystallography, X-Ray , Humans , Mass Spectrometry
11.
Cells ; 9(5)2020 05 24.
Article in English | MEDLINE | ID: mdl-32456328

ABSTRACT

Mechanotransduction translates forces into biological responses and regulates cell functionalities. It is implicated in several diseases, including laminopathies which are pathologies associated with mutations in lamins and lamin-associated proteins. These pathologies affect muscle, adipose, bone, nerve, and skin cells and range from muscular dystrophies to accelerated aging. Although the exact mechanisms governing laminopathies and gene expression are still not clear, a strong correlation has been found between cell functionality and nuclear behavior. New theories base on the direct effect of external force on the genome, which is indeed sensitive to the force transduced by the nuclear lamina. Nuclear lamina performs two essential functions in mechanotransduction pathway modulating the nuclear stiffness and governing the chromatin remodeling. Indeed, A-type lamin mutation and deregulation has been found to affect the nuclear response, altering several downstream cellular processes such as mitosis, chromatin organization, DNA replication-transcription, and nuclear structural integrity. In this review, we summarize the recent findings on the molecular composition and architecture of the nuclear lamina, its role in healthy cells and disease regulation. We focus on A-type lamins since this protein family is the most involved in mechanotransduction and laminopathies.


Subject(s)
Lamin Type A/metabolism , Laminopathies/metabolism , Laminopathies/pathology , Mechanotransduction, Cellular , Animals , Humans , Lamin Type A/chemistry , Lamin Type A/genetics , Laminopathies/genetics , Mutation/genetics , Protein Binding
12.
Mol Med Rep ; 21(6): 2459-2465, 2020 06.
Article in English | MEDLINE | ID: mdl-32323820

ABSTRACT

Progressive cardiac conduction defect (PCCD) is an inherited autosomal dominant cardiac disorder characterized by an age­dependent cardiac electrical conduction block. Several genes have been associated with the genetic pathogenesis of PCCD. The present study aimed to identify the causal mutation of PCCD and to investigate the association between genotype and phenotype in a Chinese family with PCCD. A total of 39 family members were included in the present study. All subjects participated in physical, biochemical, electrocardiography and echocardiography examinations. Whole­exome sequencing was performed for four individuals from the same generation, including three patients with PCCD and one normal control with no cardiovascular disease. Sanger sequencing and in silico analysis were used to identify the causal mutation. Whole­exome sequencing and variant identification revealed a candidate nonsense mutation (c.1443C>A, p.Tyr481*) in lamin A/C (LMNA). The mutation was identified in seven patients (including the proband) and two asymptomatic mutation carriers, but it was not detected in 100 control subjects of matched ancestry. Clinical examinations identified typical symptoms in patients with PCCD, including bradycardia and various types of conduction defect, and excluded other phenotypes related to the LMNA mutation. The genotype and phenotype were co­associated among all participants. In the present study, the c.1443C>A mutation in the LMNA gene was identified as a potential cause of PCCD. In silico analysis predicted that the identified mutation was damaging through its effect on the lamin tail domain of LMNA. From the present study, it could be suggested that genetic screening and family counseling, early pacemaker implantation or a sudden death in the family may be essential for risk stratification and treatment of patients with PCCD.


Subject(s)
Cardiac Conduction System Disease/diagnosis , Heart Block/diagnosis , Lamin Type A/genetics , Adolescent , Adult , Aged , Base Sequence , Cardiac Conduction System Disease/genetics , Codon, Nonsense , Electrocardiography , Female , Genotype , Heart Block/genetics , Humans , Lamin Type A/chemistry , Middle Aged , Pedigree , Protein Structure, Tertiary , Exome Sequencing , Young Adult
13.
Dev Cell ; 52(6): 699-713.e11, 2020 03 23.
Article in English | MEDLINE | ID: mdl-32208162

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.


Subject(s)
Cell Nucleus/metabolism , Enhancer Elements, Genetic , Lamin Type A/genetics , Mutation , Progeria/genetics , Active Transport, Cell Nucleus , Adolescent , Binding Sites , Cell Line , Cells, Cultured , Child , Fibroblasts/metabolism , Humans , Lamin Type A/chemistry , Lamin Type A/metabolism , Male , Protein Binding
14.
Cells ; 10(1)2020 12 31.
Article in English | MEDLINE | ID: mdl-33396475

ABSTRACT

Intermediate filaments (IFs) commonly have structural elements of a central α-helical coiled-coil domain consisting of coil 1a, coil 1b, coil 2, and their flanking linkers. Recently, the crystal structure of a long lamin A/C fragment was determined and showed detailed features of a tetrameric unit. The structure further suggested a new binding mode between tetramers, designated eA22, where a parallel overlap of coil 1a and coil 2 is the critical interaction. This study investigated the biochemical effects of genetic mutations causing human diseases, focusing on the eA22 interaction. The mutant proteins exhibited either weakened or augmented interactions between coil 1a and coil 2. The ensuing biochemical results indicated that the interaction requires the separation of the coiled-coils in the N-terminal of coil 1a and the C-terminal of coil 2, coupled with the structural transition in the central α-helical rod domain. This study provides insight into the role of coil 1a as a molecular regulator in the elongation of IF proteins.


Subject(s)
Intermediate Filaments/metabolism , Lamin Type A/metabolism , Lamins/metabolism , Amino Acid Sequence , Cell Line, Tumor , Circular Dichroism , Fluorescent Antibody Technique , Gene Expression , Humans , Intermediate Filaments/chemistry , Lamin Type A/chemistry , Lamin Type A/genetics , Lamins/chemistry , Lamins/genetics , Mutation , Protein Binding , Protein Conformation , Protein Conformation, alpha-Helical , Protein Domains , Protein Multimerization , Recombinant Proteins
16.
Nat Cell Biol ; 21(10): 1248-1260, 2019 10.
Article in English | MEDLINE | ID: mdl-31576060

ABSTRACT

While nuclear lamina abnormalities are hallmarks of human diseases, their interplay with epigenetic regulators and precise epigenetic landscape remain poorly understood. Here, we show that loss of the lysine acetyltransferase MOF or its associated NSL-complex members KANSL2 or KANSL3 leads to a stochastic accumulation of nuclear abnormalities with genomic instability patterns including chromothripsis. SILAC-based MOF and KANSL2 acetylomes identified lamin A/C as an acetylation target of MOF. HDAC inhibition or acetylation-mimicking lamin A derivatives rescue nuclear abnormalities observed in MOF-deficient cells. Mechanistically, loss of lamin A/C acetylation resulted in its increased solubility, defective phosphorylation dynamics and impaired nuclear mechanostability. We found that nuclear abnormalities include EZH2-dependent histone H3 Lys 27 trimethylation and loss of nascent transcription. We term this altered epigenetic landscape "heterochromatin enrichment in nuclear abnormalities" (HENA). Collectively, the NSL-complex-dependent lamin A/C acetylation provides a mechanism that maintains nuclear architecture and genome integrity.


Subject(s)
Cell Nucleus/ultrastructure , Histone Acetyltransferases/metabolism , Lamin Type A/metabolism , Nuclear Proteins/metabolism , Acetylation , Animals , Cell Nucleus/metabolism , Cells, Cultured , Epigenesis, Genetic , Fibroblasts , Heterochromatin , Histone Acetyltransferases/genetics , Histones/genetics , Histones/metabolism , Lamin Type A/chemistry , Lamin Type A/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics
17.
Anal Chem ; 91(17): 10937-10942, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31393705

ABSTRACT

Proteoforms, the primary effectors of biological processes, are the different forms of proteins that arise from molecular processing events such as alternative splicing and post-translational modifications. Heart diseases exhibit changes in proteoform levels, motivating the development of a deeper understanding of the heart proteoform landscape. Our recently developed two-dimensional top-down proteomics platform coupling serial size exclusion chromatography (sSEC) to reversed-phase chromatography (RPC) expanded coverage of the human heart proteome and allowed observation of high-molecular weight proteoforms. However, most of these observed proteoforms were not identified due to the difficulty in obtaining quality tandem mass spectrometry (MS2) fragmentation data for large proteoforms from complex biological mixtures on a chromatographic time scale. Herein, we sought to identify human heart proteoforms in this data set using an enhanced version of Proteoform Suite, which identifies proteoforms by intact mass alone. Specifically, we added a new feature to Proteoform Suite to determine candidate identifications for isotopically unresolved proteoforms larger than 50 kDa, enabling subsequent MS2 identification of important high-molecular weight human heart proteoforms such as lamin A (72 kDa) and trifunctional enzyme subunit α (79 kDa). With this new workflow for large proteoform identification, endogenous human cardiac myosin binding protein C (140 kDa) was identified for the first time. This study demonstrates the integration of our sSEC-RPC-MS proteomics platform with intact-mass analysis through Proteoform Suite to create a catalog of human heart proteoforms and facilitate the identification of large proteoforms in complex systems.


Subject(s)
Carrier Proteins/isolation & purification , Lamin Type A/isolation & purification , Mitochondrial Trifunctional Protein, alpha Subunit/isolation & purification , Myocardium/chemistry , Protein Processing, Post-Translational , Proteome/isolation & purification , Software , Alternative Splicing , Amino Acid Sequence , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Chromatography, Gel , Chromatography, Reverse-Phase , Humans , Lamin Type A/chemistry , Lamin Type A/metabolism , Mitochondrial Trifunctional Protein, alpha Subunit/chemistry , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Myocardium/metabolism , Proteome/chemistry , Proteome/metabolism , Proteomics/methods , Tandem Mass Spectrometry
18.
Nat Commun ; 10(1): 3056, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31296869

ABSTRACT

Lamin A is a nuclear intermediate filament protein critical for nuclear architecture and mechanics and mutated in a wide range of human diseases. Yet little is known about the molecular architecture of lamins and mechanisms of their assembly. Here we use SILAC cross-linking mass spectrometry to determine interactions within lamin dimers and between dimers in higher-order polymers. We find evidence for a compression mechanism where coiled coils in the lamin A rod can slide onto each other to contract rod length, likely driven by a wide range of electrostatic interactions with the flexible linkers between coiled coils. Similar interactions occur with unstructured regions flanking the rod domain during oligomeric assembly. Mutations linked to human disease block these interactions, suggesting that this spring-like contraction can explain in part the dynamic mechanical stretch and flexibility properties of the lamin polymer and other intermediate filament networks.


Subject(s)
Intermediate Filament Proteins/metabolism , Lamin Type A/metabolism , Nuclear Matrix/metabolism , Protein Multimerization/physiology , Amino Acid Sequence/physiology , Animals , Cardiomyopathy, Dilated/genetics , Cross-Linking Reagents/chemistry , Elasticity , Humans , Intermediate Filament Proteins/chemistry , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/isolation & purification , Lamin Type A/chemistry , Lamin Type A/genetics , Lamin Type A/isolation & purification , Mass Spectrometry/methods , Muscular Dystrophies/genetics , Mutation , Nuclear Envelope/metabolism , Protein Domains/genetics , Protein Structure, Secondary/physiology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
19.
Mol Biol Cell ; 30(15): 1786-1790, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31306095

ABSTRACT

Laminopathies are a diverse group of rare diseases with various pathologies in different tissues, which are linked to mutations in the LMNA gene. Historically, the structural disease model proposed mechanical defects of the lamina and nuclear fragility, the gene expression model impairment of spatial chromatin organization and signaling pathways as underlying mechanisms leading to the pathologies. Exciting findings in the past few years showing that mechanical forces are directly transmitted into the nucleus, where they affect chromatin organization and mechanoresponsive signaling molecules, have led to a revised concept of an integrative unified disease model, in which lamin-mediated pathways in mechanotransduction and chromatin regulation are highly interconnected and mutually dependent. In this Perspective we highlight breakthrough findings providing new insight into lamin-linked mechanisms of mechanotransduction and chromatin regulation and discuss how a combined and interrelated impairment of these functions by LMNA mutations may impair the complex mechanosignaling network and cause tissue-specific pathologies in laminopathies.


Subject(s)
Gene Expression Regulation , Lamin Type A/chemistry , Lamin Type A/genetics , Models, Biological , Muscular Diseases/genetics , Animals , Humans , Mechanotransduction, Cellular
20.
Cell Death Dis ; 10(6): 451, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31171766

ABSTRACT

Stress-induced premature senescence (SIPS), a state of cell growth arrest due to various stimuli, is implicated in the pathogeneses of hepatic fibrogenesis. Progerin, a permanently farnesylated mutant lamin A protein, likely leads to premature senescence to influent liver diseases. The previous reports showed that activation of insulin-like growth factor-1 (IGF-1) signaling could enhance cell longevity and attenuate liver fibrosis. However, the underlying mechanisms about hepatocyte premature senility in liver fibrosis, and how IGF-1 regulates cell premature aging and fibrogenesis, remain poorly understood. In the present study, we found the augment of hepatocyte oxidation and premature aging, along with the decrease of plasm IGF-1 level in patients with liver fibrosis and CCl4-induced liver injury rat models. Nevertheless, IGF-1 gene transfer to CCl4 rats to overexpress intrahepatic IGF-1 relieved hepatocyte oxidative stress and premature senescence, which was likely mediated by the p53/progerin pathway, to improve hepatic steatosis and fibrogenesis. In vitro, H2O2 caused abnormal accumulation of progerin in nuclear and activation of nuclear p53-progerin interaction to trigger primary rat hepatocyte premature senescence through the p21-independent pathway; while these effects were rescued by prolonged exogenous IGF-1 or the IGF-1 adenovirus vector. Furthermore, the IGF-1 adenovirus vector, transfected to H2O2-treated hepatocytes, reversed oxidative stress-induced premature senescence via enhancing cytoplasmic AKT1-p53 interaction and subsequently inhibiting nuclear p53-progerin interaction. Consequently, our data illuminate a novel role of IGF-1 in regulating stress-induced hepatocyte premature senescence in liver fibrosis: prolonged IGF-1 relieves oxidative stress-initiated hepatocyte premature senescence via inhibition of nuclear p53-progerin interaction to ameliorate hepatic steatosis and fibrogenesis.


Subject(s)
Cellular Senescence/genetics , Hepatocytes/metabolism , Insulin-Like Growth Factor I/metabolism , Lamin Type A/metabolism , Liver Cirrhosis/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Carbon Tetrachloride/toxicity , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cellular Senescence/drug effects , Disease Models, Animal , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/physiology , Humans , Hydrogen Peroxide/metabolism , Insulin-Like Growth Factor I/genetics , Lamin Type A/chemistry , Lamin Type A/genetics , Liver/metabolism , Liver/pathology , Liver Cirrhosis/chemically induced , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Oxidative Stress , Protein Prenylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction/genetics , Tumor Suppressor Protein p53/genetics , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL