Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Adv Sci (Weinh) ; 11(30): e2307751, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38894550

RESUMEN

Genomic instability is not only a hallmark of senescent cells but also a key factor driving cellular senescence, and replication stress is the main source of genomic instability. Defective prelamin A processing caused by lamin A/C (LMNA) or zinc metallopeptidase STE24 (ZMPSTE24) gene mutations results in premature aging. Although previous studies have shown that dysregulated lamin A interferes with DNA replication and causes replication stress, the relationship between lamin A dysfunction and replication stress remains largely unknown. Here, an increase in baseline replication stress and genomic instability is found in prelamin A-expressing cells. Moreover, prelamin A confers hypersensitivity of cells to exogenous replication stress, resulting in decreased cell survival and exacerbated genomic instability. These effects occur because prelamin A promotes MRE11-mediated resection of stalled replication forks. Fanconi anemia (FA) proteins, which play important roles in replication fork maintenance, are downregulated by prelamin A in a retinoblastoma (RB)/E2F-dependent manner. Additionally, prelamin A inhibits the activation of the FA pathway upon replication stress. More importantly, FA pathway downregulation is an upstream event of p53-p21 axis activation during the induction of prelamin A expression. Overall, these findings highlight the critical role of FA pathway dysfunction in driving replication stress-induced genomic instability and cellular senescence in prelamin A-expressing cells.


Asunto(s)
Replicación del ADN , Inestabilidad Genómica , Lamina Tipo A , Inestabilidad Genómica/genética , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Humanos , Replicación del ADN/genética , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Senescencia Celular/genética
2.
Int J Mol Sci ; 25(2)2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38279282

RESUMEN

The accumulation of farnesylated prelamin A has been suggested as one of the mechanisms responsible for the loss of fat in type 2 familial partial lipodystrophy due to variants in the LMNA gene. In this rare disease, fat loss appears in women after puberty, affecting sex-hormone-dependent anatomical areas. This study investigated the impact of 17-ß-estradiol on adipogenesis in murine preadipocytes subjected to a pharmacologically induced accumulation of farnesylated and non-farnesylated prelamin A. To induce the accumulation of non-farnesylated or farnesylated prelamin A, 3T3-L1 cells were treated with the farnesyltransferase inhibitor 277 or the methyltransferase inhibitor N-acetyl-S-farnesyl-l-cysteine methylester. Subsequently, the cells were induced to undergo adipocyte differentiation in the presence or absence of 17-ß-estradiol. Prelamin A accumulation was assessed through immunofluorescence, while real-time PCR and Western blot techniques were used to quantify several adipogenic genes and evaluate protein levels, respectively. The results showed that 17-ß-estradiol increased adipogenesis, although the combination of this hormone plus farnesylated prelamin A led to a reduction in the number of mature adipocytes and the expression of the different genes involved in adipogenesis. In conclusion, the influence of farnesylated prelamin A accumulation on adipogenesis manifested only in the presence of estradiol. These in vitro findings suggest a potential mechanism that could explain the characteristic phenotype in women suffering type 2 familial partial lipodystrophy.


Asunto(s)
Lamina Tipo A , Lipodistrofia Parcial Familiar , Humanos , Femenino , Ratones , Animales , Lipodistrofia Parcial Familiar/genética , Lipodistrofia Parcial Familiar/metabolismo , Adipogénesis , Células 3T3-L1 , Proteínas Nucleares/genética , Estradiol/farmacología
3.
Nucleus ; 14(1): 2288476, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38050983

RESUMEN

Several related progeroid disorders are caused by defective post-translational processing of prelamin A, the precursor of the nuclear scaffold protein lamin A, encoded by LMNA. Prelamin A undergoes farnesylation and additional modifications at its C-terminus. Subsequently, the farnesylated C-terminal segment is cleaved off by the zinc metalloprotease ZMPSTE24. The premature aging disorder Hutchinson Gilford progeria syndrome (HGPS) and a related progeroid disease, mandibuloacral dysplasia (MAD-B), are caused by mutations in LMNA and ZMPSTE24, respectively, that result in failure to process the lamin A precursor and accumulate permanently farnesylated forms of prelamin A. The farnesyl transferase inhibitor (FTI) lonafarnib is known to correct the aberrant nuclear morphology of HGPS patient cells and improves lifespan in children with HGPS. Importantly, and in contrast to a previous report, we show here that FTI treatment also improves the aberrant nuclear phenotypes in MAD-B patient cells with mutations in ZMPSTE24 (P248L or L425P). As expected, lonafarnib does not correct nuclear defects for cells with lamin A processing-proficient mutations. We also examine prelamin A processing in fibroblasts from two individuals with a prevalent laminopathy mutation LMNA-R644C. Despite the proximity of residue R644 to the prelamin A cleavage site, neither R644C patient cell line shows a prelamin A processing defect, and both have normal nuclear morphology. This work clarifies the prelamin A processing status and role of FTIs in a variety of laminopathy patient cells and supports the FDA-approved indication for the FTI Zokinvy for patients with processing-deficient progeroid laminopathies, but not for patients with processing-proficient laminopathies.


Asunto(s)
Lipodistrofia , Progeria , Niño , Humanos , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Progeria/tratamiento farmacológico , Progeria/genética , Progeria/metabolismo , Inhibidores Enzimáticos/farmacología , Mutación , Lipodistrofia/metabolismo , Fibroblastos/metabolismo , Transferasas/genética , Transferasas/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Proteínas de la Membrana/metabolismo
4.
Nucleus ; 14(1): 2270345, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37885131

RESUMEN

As human longevity increases, understanding the molecular mechanisms that drive aging becomes ever more critical to promote health and prevent age-related disorders. Premature aging disorders or progeroid syndromes can provide critical insights into aspects of physiological aging. A major cause of progeroid syndromes which result from mutations in the genes LMNA and ZMPSTE24 is disruption of the final posttranslational processing step in the production of the nuclear scaffold protein lamin A. LMNA encodes the lamin A precursor, prelamin A and ZMPSTE24 encodes the prelamin A processing enzyme, the zinc metalloprotease ZMPSTE24. Progeroid syndromes resulting from mutations in these genes include the clinically related disorders Hutchinson-Gilford progeria syndrome (HGPS), mandibuloacral dysplasia-type B, and restrictive dermopathy. These diseases have features that overlap with one another and with some aspects of physiological aging, including bone defects resembling osteoporosis and atherosclerosis (the latter primarily in HGPS). The progeroid syndromes have ignited keen interest in the relationship between defective prelamin A processing and its accumulation in normal physiological aging. In this review, we examine the hypothesis that diminished processing of prelamin A by ZMPSTE24 is a driver of physiological aging. We review features a new mouse (LmnaL648R/L648R) that produces solely unprocessed prelamin A and provides an ideal model for examining the effects of its accumulation during aging. We also discuss existing data on the accumulation of prelamin A or its variants in human physiological aging, which call out for further validation and more rigorous experimental approaches to determine if prelamin A contributes to normal aging.


Asunto(s)
Lamina Tipo A , Progeria , Humanos , Animales , Ratones , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Promoción de la Salud , Progeria/genética , Progeria/metabolismo , Envejecimiento/genética , Proteínas de la Membrana/metabolismo
5.
Life Sci ; 301: 120567, 2022 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35461839

RESUMEN

AIMS: Nuclear prelamin A recognition factor-like (NARFL) is involved in cytosolic iron­sulfur (FeS) protein biogenesis and cellular defense against oxidative stress. Previous study reported that increased oxidative stress and subintestinal vessel (SIV) malformation in narfl knockout zebrafish. However, the underlying mechanism of oxidative stress caused by NARFL deficiency remains unclear. The present study was sought to investigate the function of NARFL in endothelial cells. METHODS: NARFL knockdown assay was performed in two cell lines and NADPH oxidase (Nox) were measured using Western blotting. Nox inhibitors were selected for assessing the potential sources of reactive oxygen species (ROS) generation. Cell migration was detected using wound healing assay and transwell assay. Cell cycle was analyzed using flow cytometry. Promoter activity assay and Chromatin immunoprecipitation (ChIP) assay were chosen for investigating the molecular mechanism of Nox transcription. RESULTS: NARFL deficiency resulted in upregulated expressions of Nox2, Nox4, and p47phox and increased ROS levels in endothelial cells. Nox2 knockdown reversed the effects and improved endothelial dysfunctions caused by NARFL deficiency. ChIP experiments revealed that NARFL knockdown increased the recruitment of RNA polymerase II and modification of histones at the promoter sites of Nox2 and Nox4. CONCLUSION: NARFL knockdown induced the transcriptional activation of Nox2 and Nox4, which resulted in increased ROS levels and impaired endothelial functions.


Asunto(s)
Células Endoteliales , NADPH Oxidasas , Animales , Células Endoteliales/metabolismo , NADPH Oxidasa 2/genética , NADPH Oxidasa 2/metabolismo , NADPH Oxidasa 4/metabolismo , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Pez Cebra
6.
Aging Cell ; 19(8): e13200, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32910507

RESUMEN

Several progeroid disorders are caused by deficiency in the endoprotease ZMPSTE24 which leads to accumulation of prelamin A at the nuclear envelope. ZMPSTE24 cleaves prelamin A twice: at the third carboxyl-terminal amino acid following farnesylation of a -CSIM motif; and 15 residues upstream to produce mature lamin A. The carboxyl-terminal cleavage can also be performed by RAS-converting enzyme 1 (RCE1) but little is known about the importance of this cleavage for the ability of prelamin A to cause disease. Here, we found that knockout of RCE1 delayed senescence and increased proliferation of ZMPSTE24-deficient fibroblasts from a patient with non-classical Hutchinson-Gilford progeria syndrome (HGPS), but did not influence proliferation of classical LMNA-mutant HGPS cells. Knockout of Rce1 in Zmpste24-deficient mice at postnatal week 4-5 increased body weight and doubled the median survival time. The absence of Rce1 in Zmpste24-deficient fibroblasts did not influence nuclear shape but reduced an interaction between prelamin A and AKT which activated AKT-mTOR signaling and was required for the increased proliferation. Prelamin A levels increased in Rce1-deficient cells due to a slower turnover rate but its localization at the nuclear rim was unaffected. These results strengthen the idea that the presence of misshapen nuclei does not prevent phenotype improvement and suggest that targeting RCE1 might be useful for treating the rare progeroid disorders associated with ZMPSTE24 deficiency.


Asunto(s)
Genes ras/genética , Proteínas de la Membrana/deficiencia , Metaloendopeptidasas/deficiencia , Progeria/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Noqueados , Fenotipo
7.
Ageing Res Rev ; 62: 101073, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32446955

RESUMEN

Lamin A, a main constituent of the nuclear lamina, is the major splicing product of the LMNA gene, which also encodes lamin C, lamin A delta 10 and lamin C2. Involvement of lamin A in the ageing process became clear after the discovery that a group of progeroid syndromes, currently referred to as progeroid laminopathies, are caused by mutations in LMNA gene. Progeroid laminopathies include Hutchinson-Gilford Progeria, Mandibuloacral Dysplasia, Atypical Progeria and atypical-Werner syndrome, disabling and life-threatening diseases with accelerated ageing, bone resorption, lipodystrophy, skin abnormalities and cardiovascular disorders. Defects in lamin A post-translational maturation occur in progeroid syndromes and accumulated prelamin A affects ageing-related processes, such as mTOR signaling, epigenetic modifications, stress response, inflammation, microRNA activation and mechanosignaling. In this review, we briefly describe the role of these pathways in physiological ageing and go in deep into lamin A-dependent mechanisms that accelerate the ageing process. Finally, we propose that lamin A acts as a sensor of cell intrinsic and environmental stress through transient prelamin A accumulation, which triggers stress response mechanisms. Exacerbation of lamin A sensor activity due to stably elevated prelamin A levels contributes to the onset of a permanent stress response condition, which triggers accelerated ageing.


Asunto(s)
Envejecimiento , Envejecimiento/genética , Humanos , Lamina Tipo A/genética , MicroARNs , Mutación , Proteínas Nucleares , Progeria/genética , Precursores de Proteínas/genética
8.
FASEB J ; 34(6): 7905-7914, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32282093

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) arises when a truncated form of farnesylated prelamin A accumulates at the nuclear envelope, leading to misshapen nuclei. Previous studies of adult Zmpste24-deficient mice, a mouse model of progeria, have reported a metabolic response involving inhibition of the mTOR (mammalian target of rapamycin) kinase and activation of autophagy. However, exactly how mTOR or autophagy is involved in progeria remains unclear. Here, we investigate this question by crossing Zmpste24+/- mice with mice hypomorphic in mTOR (mTOR△/+ ), or mice heterozygous in autophagy-related gene 7 (Atg7+/- ). We find that accumulation of prelamin A induces premature aging through mTOR overactivation and impaired autophagy in newborn Zmpste24-/- mice. Zmpste24-/- mice with genetically reduced mTOR activity, but not heterozygosity in Atg7, show extended lifespan. Moreover, mTOR inhibition partially restores autophagy and S6K1 activity. We also show that progerin interacts with the Akt phosphatase to promote full activation of the Akt/mTOR signaling pathway. Finally, although we find that genetic reduction of mTOR postpones premature aging in Zmpste24 KO mice, frequent embryonic lethality occurs. Together, our findings show that over-activated mTOR contributes to premature aging in Zmpste24-/- mice, and suggest a potential strategy in treating HGPS patients with mTOR inhibitors.


Asunto(s)
Envejecimiento Prematuro/metabolismo , Lamina Tipo A/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Autofagia/fisiología , Proteína 7 Relacionada con la Autofagia/metabolismo , Línea Celular , Línea Celular Tumoral , Núcleo Celular/metabolismo , Femenino , Fibroblastos/metabolismo , Células HEK293 , Humanos , Células MCF-7 , Masculino , Metaloendopeptidasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Progeria/metabolismo , Transducción de Señal/fisiología
9.
Acta Myol ; 39(4): 320-335, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33458588

RESUMEN

LMNA gene encodes for lamin A/C, attractive proteins linked to nuclear structure and functions. When mutated, it causes different rare diseases called laminopathies. In particular, an Arginine change in Histidine in position 527 (p.Arg527His) falling in the C-terminal domain of lamin A precursor form (prelamin A) causes mandibuloacral dysplasia Type A (MADA), a segmental progeroid syndrome characterized by skin, bone and metabolic anomalies. The well-characterized cellular models made difficult to assess the tissue-specific functions of 527His prelamin A. Here, we describe the generation and characterization of a MADA transgenic mouse overexpressing 527His LMNA gene, encoding mutated prelamin A. Bodyweight is slightly affected, while no difference in lifespan was observed in transgenic animals. Mild metabolic anomalies and thinning and loss of hairs from the back were the other observed phenotypic MADA manifestations. Histological analysis of tissues relevant for MADA syndrome revealed slight increase in adipose tissue inflammatory cells and a reduction of hypodermis due to a loss of subcutaneous adipose tissue. At cellular levels, transgenic cutaneous fibroblasts displayed nuclear envelope aberrations, presence of prelamin A, proliferation, and senescence rate defects. Gene transcriptional pattern was found differentially modulated between transgenic and wildtype animals, too. In conclusion, the presence of 527His Prelamin A accumulation is further linked to the appearance of mild progeroid features and metabolic disorder without lifespan reduction.


Asunto(s)
Acroosteólisis/etiología , Acroosteólisis/metabolismo , Modelos Animales de Enfermedad , Lamina Tipo A/genética , Lipodistrofia/etiología , Lipodistrofia/metabolismo , Mandíbula/anomalías , Mutación/genética , Piel/patología , Acroosteólisis/patología , Animales , Femenino , Lipodistrofia/patología , Masculino , Mandíbula/metabolismo , Mandíbula/patología , Ratones , Ratones Transgénicos
10.
J Cell Physiol ; 234(10): 17800-17811, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30895608

RESUMEN

Urinary incontinence (UI) is known as a distressing condition particularly among older adults, and negatively associated with health-related quality of life in both males and females. Prelamin A accumulation has been found in all progeroid laminopathies and is obviously linked to cell and organism aging. Therefore, this study was expected to investigate the effect of prelamin A on detrusor on UI. Prelamin A expression in clinical and animal samples was detected. To investigate the degree of prelamin A accumulation and detrusor calcification/aging, the detrusor cells were subcultured separately into low and high passage. The low-passage subculture cells were treated with transfection of overexpressed prelamin A plasmid, and transfection of overexpressed prelamin A plasmid and application of farnesyl transferase inhibitor (FTIs) H-9279, respectively. Zmpste24, Icmt and lamin A/C expression were detected to explore how prelamin A affected detrusor calcification/aging. Prelamin A was overexpressed in aged detrusor cells, indicating prelamin A expression was positively related to the age of subjects. The degree of prelamin A accumulation and detrusor calcification/aging was higher in aged rats and high passage subculture cells. Zmpste24, Icmt and lamin A/C were poorly expressed in cells transfected with overexpressed prelamin A, as well as cell proliferation activity decreased and calcium deposition and apoptotic rate increased. Furthermore, we also found that the effect of overexpressed prelamin A was lost when cells were treated with H-9279. These findings provide evidence that prelamin A overexpression impairs degradation of its farnesylated form, thus causing prelamin A accumulation which induces detrusor calcification/aging in UI.


Asunto(s)
Envejecimiento/metabolismo , Calcinosis/metabolismo , Lamina Tipo A/metabolismo , Incontinencia Urinaria/metabolismo , Adulto , Anciano , Animales , Células Cultivadas , Femenino , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Proteínas Nucleares/metabolismo , Calidad de Vida , Ratas , Ratas Sprague-Dawley
11.
Bioorg Med Chem ; 26(20): 5547-5554, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30309670

RESUMEN

Lamin A contributes to the structure of nuclei in all mammalian cells and plays an important role in cell division and migration. Mature lamin A is derived from a farnesylated precursor protein, known as prelamin A, which undergoes post-translational cleavage catalyzed by the zinc metalloprotease STE24 (ZPMSTE24). Accumulation of farnesylated prelamin A in the nuclear envelope compromises cell division, impairs mitosis and induces an increased expression of inflammatory gene products. ZMPSTE24 has been proposed as a potential therapeutic target in oncology. A library of peptidomimetic compounds were synthesized and screened for their ability to induce accumulation of prelamin A in cancer cells and block cell migration in pancreatic ductal adenocarcinoma cells. The results of this study suggest that inhibitors of lamin A maturation may interfere with cell migration, the biological process required for cancer metastasis.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Lamina Tipo A/metabolismo , Peptidomiméticos/química , Peptidomiméticos/farmacología , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antineoplásicos/síntesis química , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Humanos , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/metabolismo , Invasividad Neoplásica/patología , Invasividad Neoplásica/prevención & control , Peptidomiméticos/síntesis química , Ácidos Fosfínicos/síntesis química , Ácidos Fosfínicos/química , Ácidos Fosfínicos/farmacología
12.
Cells ; 7(4)2018 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-29690642

RESUMEN

The mammalian nuclear lamina proteins—prelamin A- and B-type lamins—are post-translationally modified by farnesylation, endoproteolysis, and carboxymethylation at a carboxy-terminal CAAX (C, cysteine; a, aliphatic amino acid; X, any amino acid) motif. However, prelamin A processing into mature lamin A is a unique process because it results in the production of farnesylated and carboxymethylated peptides. In cells from patients with Hutchinson⁻Gilford progeria syndrome, the mutant prelamin A protein, progerin, cannot release its prenylated carboxyl-terminal moiety and therefore remains permanently associated with the nuclear envelope (NE), causing severe nuclear alterations and a dysmorphic morphology. To obtain a better understanding of the abnormal interaction and retention of progerin in the NE, we analyzed the spatiotemporal distribution of the EGFP fusion proteins with or without a nuclear localization signal (NLS) and a functional CAAX motif in HeLa cells transfected with a series of plasmids that encode the carboxy-terminal ends of progerin and prelamin A. The farnesylated carboxy-terminal fusion peptides bind to the NE and induce the formation of abnormally shaped nuclei. In contrast, the unfarnesylated counterparts exhibit a diffuse localization in the nucleoplasm, without obvious NE deformation. High levels of farnesylated prelamin A and progerin carboxy-terminal peptides induce nucleophagic degradation of the toxic protein, including several nuclear components and chromatin. However, SUN1, a constituent of the linker of nucleoskeleton and cytoskeleton (LINC) complex, is excluded from these autophagic NE protrusions. Thus, nucleophagy requires NE flexibility, as indicated by SUN1 delocalization from the elongated NE⁻autophagosome complex.

13.
Ageing Res Rev ; 42: 1-13, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29208544

RESUMEN

Mandibuloacral dysplasia (MAD) is a rare genetic condition characterized by bone abnormalities including localized osteolysis and generalized osteoporosis, skin pigmentation, lipodystrophic signs and mildly accelerated ageing. The molecular defects associated with MAD are mutations in LMNA or ZMPSTE24 (FACE1) gene, causing type A or type B MAD, respectively. Downstream of LMNA or ZMPSTE24 mutations, the lamin A precursor, prelamin A, is accumulated in cells and affects chromatin dynamics and stress response. A new form of mandibuloacral dysplasia has been recently associated with mutations in POLD1 gene, encoding DNA polymerase delta, a major player in DNA replication. Of note, involvement of prelamin A in chromatin dynamics and recruitment of DNA repair factors has been also determined under physiological conditions, at the border between stress response and cellular senescence. Here, we review current knowledge on MAD clinical and pathogenetic aspects and highlight aspects typical of physiological ageing.


Asunto(s)
Acroosteólisis/diagnóstico por imagen , Acroosteólisis/metabolismo , Envejecimiento Prematuro/diagnóstico por imagen , Envejecimiento Prematuro/metabolismo , Envejecimiento/metabolismo , Lipodistrofia/diagnóstico por imagen , Lipodistrofia/metabolismo , Mandíbula/anomalías , Acroosteólisis/genética , Envejecimiento/genética , Envejecimiento/patología , Envejecimiento Prematuro/genética , Animales , Humanos , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Lipodistrofia/genética , Mandíbula/diagnóstico por imagen , Mandíbula/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mutación/fisiología
14.
Oncotarget ; 8(2): 2708-2718, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27835862

RESUMEN

Two daughters in a Chinese consanguineous family were diagnosed as diffuse pulmonary arteriovenous malformations (PAVMs) and screened using whole exome sequencing (WES) and copy number variations (CNVs) chips. Though no mutation was found in the established causative genes of capillary malformation-AVMs (CM-AVMs) or PAVMs, Ser161Ile (hg19 NM_022493 c.482G>T) mutation in nuclear prelamin A recognition factor-like (NARFL) was identified. Ser161Ile mutation in NARFL conservation region was predicted to be deleterious and absent in 500 population controls and Exome Aggregation Consortium (ExAC) Database. And there was a dosage effect of the mutation on mRNA levels among family members and population controls, consistent with the instability of mutant mRNA in vitro. Accordingly, in lung tissue of the proband, NARFL protein expression was reduced but Fe3+ was overloaded with vascular endothelial growth factor (VEGF) overexpression. Furthermore, NARFL-knockdown cell lines demonstrated decreased activity of cytosolic aconitase, while NARFL-knockout zebrafish presented ectopic subintestinal vessels sprouts and upregulated VEGF. So we concluded that the Ser161Ile mutant induced NARFL deficiency and eventually diffuse PAVMs probably through VEGF pathway. In a word, we detected a functional mutation in NARFL, which might be the pathogenic gene in this pedigree.


Asunto(s)
Malformaciones Arteriovenosas/diagnóstico , Malformaciones Arteriovenosas/genética , Estudios de Asociación Genética , Hidrogenasas/genética , Mutación , Arteria Pulmonar/anomalías , Animales , Biopsia , Línea Celular , Hibridación Genómica Comparativa , Consanguinidad , Análisis Mutacional de ADN , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Hidrogenasas/química , Hidrogenasas/metabolismo , Inmunohistoquímica , Proteínas Hierro-Azufre , Modelos Moleculares , Neovascularización Patológica/genética , Linaje , Fenotipo , Conformación Proteica , Estabilidad del ARN , Radiografía Torácica , Tomografía Computarizada por Rayos X , Secuenciación del Exoma , Adulto Joven , Pez Cebra
15.
Nucleus ; 7(5): 498-511, 2016 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-27676213

RESUMEN

The accumulation of prelamin A is linked to disruption of cellular homeostasis, tissue degeneration and aging. Its expression is implicated in compromised genome stability and increased levels of DNA damage, but to date there is no complete explanation for how prelamin A exerts its toxic effects. As the nuclear lamina is important for DNA replication we wanted to investigate the relationship between prelamin A expression and DNA replication fork stability. In this study we report that the expression of prelamin A in U2OS cells induced both mono-ubiquitination of proliferating cell nuclear antigen (PCNA) and subsequent induction of Pol η, two hallmarks of DNA replication fork stalling. Immunofluorescence microscopy revealed that cells expressing prelamin A presented with high levels of colocalisation between PCNA and γH2AX, indicating collapse of stalled DNA replication forks into DNA double-strand breaks. Subsequent protein-protein interaction assays showed prelamin A interacted with PCNA and that its presence mitigated interactions between PCNA and the mature nuclear lamina. Thus, we propose that the cytotoxicity of prelamin A arises in part, from it actively competing against mature lamin A to bind PCNA and that this destabilises DNA replication to induce fork stalling which in turn contributes to genomic instability.


Asunto(s)
Replicación del ADN , Lamina Tipo A/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Línea Celular Tumoral , Daño del ADN , Dimerización , Histonas/metabolismo , Humanos , Unión Proteica , Transporte de Proteínas , Timina/metabolismo , Ubiquitinación
16.
Cells ; 5(3)2016 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-27409638

RESUMEN

Progeroid laminopathies, including Hutchinson-Gilford Progeria Syndrome (HGPS, OMIM #176670), are premature and accelerated aging diseases caused by defects in nuclear A-type Lamins. Most HGPS patients carry a de novo point mutation within exon 11 of the LMNA gene encoding A-type Lamins. This mutation activates a cryptic splice site leading to the deletion of 50 amino acids at its carboxy-terminal domain, resulting in a truncated and permanently farnesylated Prelamin A called Prelamin A Δ50 or Progerin. Some patients carry other LMNA mutations affecting exon 11 splicing and are named "HGPS-like" patients. They also produce Progerin and/or other truncated Prelamin A isoforms (Δ35 and Δ90) at the transcriptional and/or protein level. The results we present show that morpholino antisense oligonucleotides (AON) prevent pathogenic LMNA splicing, markedly reducing the accumulation of Progerin and/or other truncated Prelamin A isoforms (Prelamin A Δ35, Prelamin A Δ90) in HGPS-like patients' cells. Finally, a patient affected with Mandibuloacral Dysplasia type B (MAD-B, carrying a homozygous mutation in ZMPSTE24, encoding an enzyme involved in Prelamin A maturation, leading to accumulation of wild type farnesylated Prelamin A), was also included in this study. These results provide preclinical proof of principle for the use of a personalized antisense approach in HGPS-like and MAD-B patients, who may therefore be eligible for inclusion in a therapeutic trial based on this approach, together with classical HGPS patients.

17.
Atherosclerosis ; 245: 200-11, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26724531

RESUMEN

BACKGROUND: Some LMNA mutations responsible for lipodystrophies, and some HIV-protease inhibitors (PIs) induce accumulation of farnesylated prelamin A and premature senescence in some cell types. Patients with LMNA mutations or under PI-based therapy suffer from early atherosclerosis. The metalloprotease ZMPSTE24 is the key enzyme in prelamin A maturation. AIM: We studied whether altered expression of ZMPSTE24 could contribute to vascular cell dysfunction in response to LMNA mutations or PI treatments. METHODS: Protein expression of prelamin A and ZMPSTE24 were evaluated in patients' cells and in human cultured VSMCs. Oxidative stress, inflammation, senescence and transdifferentiation/calcification were evaluated in VSMCs. RESULTS: Fibroblasts from LMNA-mutated lipodystrophic patients (mutations R482W, D47Y or R133L) and peripheral blood mononuclear cells from PI-treated-HIV-infected patients expressed increased prelamin A and decreased ZMPSTE24, which was also observed in VSMCs overexpressing mutant LMNA or treated with PIs. These alterations correlated with oxidative stress, inflammation, senescence and calcification (all p < 0.05). ZMPSTE24 silencing in native VSMCs recapitulated the mutant LMNA- and PI-induced accumulation of farnesylated prelamin A, oxidative stress, inflammation, senescence and calcification. A negative regulator of ZMPSTE24, miRNA-141-3p, was enhanced in LMNA-mutated or PI-treated VSMCs. The farnesylation inhibitors pravastatin and FTI-277, or the antioxidant N-acetyl cysteine, partly restored ZMPSTE24 expression, and concomitantly decreased oxidative stress, inflammation, senescence, and calcification of PI-treated VSCMs. CONCLUSIONS: ZMPSTE24 downregulation is a major contributor in VSMC dysfunctions resulting from LMNA mutations or PI treatments that could translate in early atherosclerosis at the clinical level. These novel pathophysiological mechanisms could open new therapeutic perspectives for cardiovascular aging.


Asunto(s)
Regulación hacia Abajo , Inhibidores de la Proteasa del VIH/farmacología , Lamina Tipo A/genética , Lipodistrofia/genética , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Mutación , Calcificación Vascular/genética , Células Cultivadas , Senescencia Celular/efectos de los fármacos , ADN/genética , Análisis Mutacional de ADN , Humanos , Lamina Tipo A/metabolismo , Lipodistrofia/tratamiento farmacológico , Lipodistrofia/metabolismo , Proteínas de la Membrana/biosíntesis , Metaloendopeptidasas/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Calcificación Vascular/tratamiento farmacológico , Calcificación Vascular/metabolismo
18.
Aging (Albany NY) ; 7(7): 486-99, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26232943

RESUMEN

Glia Maturation Factor-ß (GMF), a brain specific protein, is induced by proteinuria in renal tubules. Ectopic GMF overexpression causes apoptosisin vitro via cellular vulnerability to oxidative stress. In order to examine the roles of GMF in non-brain tissue, we constructed transgenic mice overexpressing GMF (GMF-TG). The GMF-TG mice exhibited appearance phenotypes associated with premature aging. The GMF-TG mice also demonstrated short lifespans and reduced hair regrowth, suggesting an accelerated aging process. The production of an abnormal lamin A, a nuclear envelope protein, plays a causal role in both normal aging and accelerated aging diseases, known as laminopathies. Importantly, we identified the abnormal lamin A (prelamin A), accompanied by a down-regulation of a lamin A processing enzyme (Zmpste24) in the kidney of the GMF-TG mice. The GMF-TG mice showed accelerated aging in the kidney, compared with wild-type mice, showing increased TGF-ß1, CTGF gene and serum creatinine. The gene expression of p21/waf1 was increased at an earlier stage of life, at 10 weeks, which was in turn down-regulated at a later stage, at 60 weeks. In conclusion, we propose that GMF-TG mice might be a novel mouse model of accelerated aging, due to the abnormal lamin A.


Asunto(s)
Envejecimiento Prematuro/genética , Factor de Maduración de la Glia/genética , Proteínas de la Membrana/biosíntesis , Metaloendopeptidasas/biosíntesis , Estrés Oxidativo/genética , Envejecimiento/genética , Animales , Creatinina/sangre , Regulación hacia Abajo , Expresión Génica/efectos de los fármacos , Factor de Maduración de la Glia/biosíntesis , Cabello/crecimiento & desarrollo , Estimación de Kaplan-Meier , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas p21(ras)/metabolismo
19.
Proteomics Clin Appl ; 8(3-4): 258-68, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24415579

RESUMEN

PURPOSE: Malignant mesothelioma is a neoplastic disease linked to asbestos exposure whose diagnosis is limited, so detection methods for an early diagnosis and treatment result essential. Here, we compared proteomic profiles of malignant pleural mesothelioma (MPM) and benign biopsies to search potential biomarkers useful in differential diagnosis. EXPERIMENTAL DESIGN: Tissue biopsies were obtained from 53 patients who were subjected to a diagnostic thoracoscopy. 2DE/MS based approach was used for proteomic analysis and protein validation was carried out by Western blot analysis versus benign and lung carcinoma samples. RESULTS: Among the proteins identified we confirmed known MPM biomarkers such as calretinin and suggested the new ones as prelamin A/C, desmin, vimentin, calretinin, fructose-bisphosphate aldolase A, myosin regulatory light chain 2, ventricular/cardiac muscle isoform, myosin light chain 3 and myosin light chain 6B. Ingenuity software was used to identify the biological processes to which these proteins belong and to construct a potential network. CONCLUSIONS AND CLINICAL RELEVANCE: Overall, our results suggest potential biomarkers that can be useful in occupational medicine for the early identification of the onset of disease in health surveillance of past asbestos-exposed workers, for monitoring the progress of disease and for assessing the response to treatment.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Neoplasias Pulmonares/genética , Mesotelioma/genética , Neoplasias Pleurales/genética , Proteómica , Adulto , Anciano , Anciano de 80 o más Años , Amianto/toxicidad , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Lamina Tipo B/biosíntesis , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/patología , Masculino , Mesotelioma/inducido químicamente , Mesotelioma/patología , Mesotelioma Maligno , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Neoplasias Pleurales/inducido químicamente , Neoplasias Pleurales/patología
20.
J Cell Sci ; 127(Pt 1): 147-57, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-24155329

RESUMEN

The dynamic organisation of the cell nucleus is profoundly modified during growth, development and senescence as a result of changes in chromatin arrangement and gene transcription. A plethora of data suggests that the nuclear lamina is a key player in chromatin dynamics and argues in favour of a major involvement of prelamin A in fundamental mechanisms regulating cellular senescence and organism ageing. As the best model to analyse the role of prelamin A in normal ageing, we used cells from centenarian subjects. We show that prelamin A is accumulated in fibroblasts from centenarians owing to downregulation of its specific endoprotease ZMPSTE24, whereas other nuclear envelope constituents are mostly unaffected and cells do not enter senescence. Accumulation of prelamin A in nuclei of cells from centenarians elicits loss of heterochromatin, as well as recruitment of the inactive form of 53BP1, associated with rapid response to oxidative stress. These effects, including the prelamin-A-mediated increase of nuclear 53BP1, can be reproduced by rapamycin treatment of cells from younger individuals. These data identify prelamin A and 53BP1 as new targets of rapamycin that are associated with human longevity. We propose that the reported mechanisms safeguard healthy ageing in humans through adaptation of the nuclear environment to stress stimuli.


Asunto(s)
Envejecimiento/genética , Antibióticos Antineoplásicos/farmacología , Fibroblastos/efectos de los fármacos , Longevidad/genética , Proteínas Nucleares/genética , Precursores de Proteínas/genética , Sirolimus/farmacología , Anciano de 80 o más Años , Envejecimiento/metabolismo , Núcleo Celular/efectos de los fármacos , Núcleo Celular/genética , Núcleo Celular/metabolismo , Senescencia Celular/efectos de los fármacos , Senescencia Celular/genética , Cromatina/efectos de los fármacos , Cromatina/genética , Cromatina/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/agonistas , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lamina Tipo A , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Metaloendopeptidasas/antagonistas & inhibidores , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Proteínas Nucleares/agonistas , Proteínas Nucleares/metabolismo , Estrés Oxidativo , Precursores de Proteínas/agonistas , Precursores de Proteínas/metabolismo , Transducción de Señal , Proteína 1 de Unión al Supresor Tumoral P53
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA