Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
Exp Dermatol ; 31(11): 1748-1760, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36320153

RESUMEN

Inflammaging is a theory of ageing which purports that low-level chronic inflammation leads to cellular dysfunction and premature ageing of surrounding tissue. Skin is susceptible to inflammaging because it is the first line of defence from the environment, particularly solar radiation. To better understand the impact of ageing and photoexposure on epidermal biology, we performed a system biology-based analysis of photoexposed face and arm, and photoprotected buttock sites, from women between the ages of 20s to 70s. Biopsies were analysed by histology, transcriptomics, and proteomics and skin surface biomarkers collected from tape strips. We identified morphological changes with age of epidermal thinning, rete ridge pathlength loss and stratum corneum thickening. The SASP biomarkers IL-8 and IL-1RA/IL1-α were consistently elevated in face across age and cis/trans-urocanic acid were elevated in arms and face with age. In older arms, the DNA damage response biomarker 53BP1 showed higher puncti numbers in basal layers and epigenetic ageing were accelerated. Genes associated with differentiation and senescence showed increasing expression in the 30s whereas genes associated with hypoxia and glycolysis increased in the 50's. Proteomics comparing 60's vs 20's confirmed elevated levels of differentiation and glycolytic-related proteins. Representative immunostaining for proteins of differentiation, senescence and oxygen sensing/hypoxia showed similar relationships. This system biology-based analysis provides a body of evidence that young photoexposed skin is undergoing inflammaging. We propose the presence of chronic inflammation in young skin contributes to an imbalance of epidermal homeostasis that leads to a prematurely aged appearance during later life.


Asunto(s)
Epidermis , Piel , Humanos , Femenino , Anciano , Adulto Joven , Adulto , Piel/metabolismo , Homeostasis , Inflamación/metabolismo , Hipoxia/metabolismo , Senescencia Celular
2.
Biochem Soc Trans ; 48(3): 981-991, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32539085

RESUMEN

The metazoan nucleus is equipped with a meshwork of intermediate filament proteins called the A- and B-type lamins. Lamins lie beneath the inner nuclear membrane and serve as a nexus to maintain the architectural integrity of the nucleus, chromatin organization, DNA repair and replication and to regulate nucleocytoplasmic transport. Perturbations or mutations in various components of the nuclear lamina result in a large spectrum of human diseases collectively called laminopathies. One of the most well-characterized laminopathies is Hutchinson-Gilford progeria (HGPS), a rare segmental premature aging syndrome that resembles many features of normal human aging. HGPS patients exhibit alopecia, skin abnormalities, osteoporosis and succumb to cardiovascular complications in their teens. HGPS is caused by a mutation in LMNA, resulting in a mutated form of lamin A, termed progerin. Progerin expression results in a myriad of cellular phenotypes including abnormal nuclear morphology, loss of peripheral heterochromatin, transcriptional changes, DNA replication defects, DNA damage and premature cellular senescence. A key challenge is to elucidate how these different phenotypes are causally and mechanistically linked. In this mini-review, we highlight some key findings and present a model on how progerin-induced phenotypes may be temporally and mechanistically linked.


Asunto(s)
Envejecimiento Prematuro/genética , Senescencia Celular , Progeria/genética , Animales , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Daño del ADN , Reparación del ADN , Replicación del ADN , Perfilación de la Expresión Génica , Humanos , Proteínas de Filamentos Intermediarios/metabolismo , Lamina Tipo A/genética , Ratones , Mutación , Lámina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fenotipo , Dominios Proteicos
3.
Cell Tissue Res ; 360(3): 621-31, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25532872

RESUMEN

The intermediate filament A- and B-type lamins are key architectural components of the nuclear lamina, a proteinaceous meshwork that lies underneath the inner nuclear membrane. In the past decade, many different monogenic human diseases have been linked to mutations in various components of the nuclear lamina. Mutations in LMNA (encoding lamin A and C) cause a variety of human diseases, collectively called laminopathies. These include cardiomyopathies, muscular dystrophies, lipodystrophies and progeroid syndromes. In addition, elevated levels of lamin B1, attributable to genomic duplications of the LMNB1 locus, cause adult-onset autosomal dominant leukodystrophy. The molecular mechanism(s) enabling the mutations and perturbations of the nuclear lamina to give rise to such a wide variety of diseases that affect various tissues remains unclear. The composition of the nuclear lamina changes dynamically during development, between cell types and even within the same cell during differentiation and ageing. Here, we discuss the functional and cellular aspects of lamina remodelling and their implications for the tissue-specific nature of laminopathies.


Asunto(s)
Enfermedad , Lámina Nuclear/patología , Animales , Senescencia Celular , Enfermedad/genética , Predisposición Genética a la Enfermedad , Humanos , Laminas/genética , Mutación/genética
4.
Nature ; 459(7244): 278-81, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19369939

RESUMEN

Trypanosoma brucei is the causative agent of African sleeping sickness in humans and one of the causes of nagana in cattle. This protozoan parasite evades the host immune system by antigenic variation, a periodic switching of its variant surface glycoprotein (VSG) coat. VSG switching is spontaneous and occurs at a rate of about 10(-2)-10(-3) per population doubling in recent isolates from nature, but at a markedly reduced rate (10(-5)-10(-6)) in laboratory-adapted strains. VSG switching is thought to occur predominantly through gene conversion, a form of homologous recombination initiated by a DNA lesion that is used by other pathogens (for example, Candida albicans, Borrelia sp. and Neisseria gonorrhoeae) to generate surface protein diversity, and by B lymphocytes of the vertebrate immune system to generate antibody diversity. Very little is known about the molecular mechanism of VSG switching in T. brucei. Here we demonstrate that the introduction of a DNA double-stranded break (DSB) adjacent to the approximately 70-base-pair (bp) repeats upstream of the transcribed VSG gene increases switching in vitro approximately 250-fold, producing switched clones with a frequency and features similar to those generated early in an infection. We were also able to detect spontaneous DSBs within the 70-bp repeats upstream of the actively transcribed VSG gene, indicating that a DSB is a natural intermediate of VSG gene conversion and that VSG switching is the result of the resolution of this DSB by break-induced replication.


Asunto(s)
Variación Antigénica/genética , Roturas del ADN de Doble Cadena , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Modelos Genéticos , Proteínas de Saccharomyces cerevisiae/metabolismo , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/inmunología , Animales , Reparación del ADN/genética , Replicación del ADN , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Conversión Génica/genética , Reacción en Cadena de la Polimerasa , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Análisis de Secuencia , Glicoproteínas Variantes de Superficie de Trypanosoma/genética , Glicoproteínas Variantes de Superficie de Trypanosoma/inmunología
5.
PLoS Pathog ; 8(8): e1002900, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22952449

RESUMEN

Trypanosoma brucei is a master of antigenic variation and immune response evasion. Utilizing a genomic repertoire of more than 1000 Variant Surface Glycoprotein-encoding genes (VSGs), T. brucei can change its protein coat by "switching" from the expression of one VSG to another. Each active VSG is monoallelically expressed from only one of approximately 15 subtelomeric sites. Switching VSG expression occurs by three predominant mechanisms, arguably the most significant of which is the non-reciprocal exchange of VSG containing DNA by duplicative gene conversion (GC). How T. brucei orchestrates its complex switching mechanisms remains to be elucidated. Recent work has demonstrated that an exogenous DNA break in the active site could initiate a GC based switch, yet the source of the switch-initiating DNA lesion under natural conditions is still unknown. Here we investigated the hypothesis that telomere length directly affects VSG switching. We demonstrate that telomerase deficient strains with short telomeres switch more frequently than genetically identical strains with long telomeres and that, when the telomere is short, switching preferentially occurs by GC. Our data supports the hypothesis that a short telomere at the active VSG expression site results in an increase in subtelomeric DNA breaks, which can initiate GC based switching. In addition to their significance for T. brucei and telomere biology, the findings presented here have implications for the many diverse pathogens that organize their antigenic genes in subtelomeric regions.


Asunto(s)
Variación Antigénica/genética , Variación Genética , Telómero/genética , Trypanosoma brucei brucei/genética , Glicoproteínas Variantes de Superficie de Trypanosoma/genética , ADN Protozoario/genética , Conversión Génica , Duplicación de Gen , Humanos , Fenotipo , Telomerasa/genética , Telomerasa/metabolismo , Telómero/metabolismo , Homeostasis del Telómero/genética , Trypanosoma brucei brucei/inmunología , Trypanosoma brucei brucei/metabolismo , Glicoproteínas Variantes de Superficie de Trypanosoma/inmunología , Glicoproteínas Variantes de Superficie de Trypanosoma/metabolismo
6.
Aging Cell ; 23(5): e14105, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38504487

RESUMEN

Hutchinson-Gilford Progeria syndrome (HGPS) is a severe premature ageing disorder caused by a 50 amino acid truncated (Δ50AA) and permanently farnesylated lamin A (LA) mutant called progerin. On a cellular level, progerin expression leads to heterochromatin loss, impaired nucleocytoplasmic transport, telomeric DNA damage and a permanent growth arrest called cellular senescence. Although the genetic basis for HGPS has been elucidated 20 years ago, the question whether the Δ50AA or the permanent farnesylation causes cellular defects has not been addressed. Moreover, we currently lack mechanistic insight into how the only FDA-approved progeria drug Lonafarnib, a farnesyltransferase inhibitor (FTI), ameliorates HGPS phenotypes. By expressing a variety of LA mutants using a doxycycline-inducible system, and in conjunction with FTI, we demonstrate that the permanent farnesylation, and not the Δ50AA, is solely responsible for progerin-induced cellular defects, as well as its rapid accumulation and slow clearance. Importantly, FTI does not affect clearance of progerin post-farnesylation and we demonstrate that early, but not late FTI treatment prevents HGPS phenotypes. Collectively, our study unravels the precise contributions of progerin's permanent farnesylation to its turnover and HGPS cellular phenotypes, and how FTI treatment ameliorates these. These findings are applicable to other diseases associated with permanently farnesylated proteins, such as adult-onset autosomal dominant leukodystrophy.


Asunto(s)
Lamina Tipo A , Progeria , Lamina Tipo A/metabolismo , Lamina Tipo A/genética , Humanos , Progeria/metabolismo , Progeria/genética , Progeria/patología , Progeria/tratamiento farmacológico , Farnesiltransferasa/metabolismo , Farnesiltransferasa/antagonistas & inhibidores , Farnesiltransferasa/genética , Prenilación de Proteína , Dibenzocicloheptenos , Piperidinas , Piridinas
7.
Front Physiol ; 14: 1297637, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38074322

RESUMEN

Aging is the result of a gradual functional decline at the cellular, and ultimately, organismal level, resulting in an increased risk of developing a variety of chronic illnesses, such as cardiovascular disease, stroke, cancer and diabetes. The skin is the largest organ of the human body, and the site where signs of aging are most visible. These signs include thin and dry skin, sagging, loss of elasticity, wrinkles, as well as aberrant pigmentation. The appearance of these features is accelerated by exposure to extrinsic factors such as ultraviolet (UV) radiation or pollution, as well as intrinsic factors including time, genetics, and hormonal changes. At the cellular level, aging is associated with impaired proteostasis and an accumulation of macromolecular damage, genomic instability, chromatin reorganization, telomere shortening, remodelling of the nuclear lamina, proliferation defects and premature senescence. Cellular senescence is a state of permanent growth arrest and a key hallmark of aging in many tissues. Due to their inability to proliferate, senescent cells no longer contribute to tissue repair or regeneration. Moreover, senescent cells impair tissue homeostasis, promote inflammation and extracellular matrix (ECM) degradation by secreting molecules collectively known as the "senescence-associated secretory phenotype" (SASP). Senescence can be triggered by a number of different stimuli such as telomere shortening, oncogene expression, or persistent activation of DNA damage checkpoints. As a result, these cells accumulate in aging tissues, including human skin. In this review, we focus on the role of cellular senescence during skin aging and the development of age-related skin pathologies, and discuss potential strategies to rejuvenate aged skin.

8.
Nat Commun ; 14(1): 8252, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-38086788

RESUMEN

Telomeres are nucleoprotein structures at the ends of linear chromosomes. In humans, they consist of TTAGGG repeats, which are bound by dedicated proteins such as the shelterin complex. This complex blocks unwanted DNA damage repair at telomeres, e.g. by suppressing nonhomologous end joining (NHEJ) through its subunit TRF2. Here, we describe ZNF524, a zinc finger protein that directly binds telomeric repeats with nanomolar affinity, and reveal base-specific sequence recognition by cocrystallization with telomeric DNA. ZNF524 localizes to telomeres and specifically maintains the presence of the TRF2/RAP1 subcomplex at telomeres without affecting other shelterin members. Loss of ZNF524 concomitantly results in an increase in DNA damage signaling and recombination events. Overall, ZNF524 is a direct telomere-binding protein involved in the maintenance of telomere integrity.


Asunto(s)
Telómero , Proteína 2 de Unión a Repeticiones Teloméricas , Humanos , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Telómero/genética , Telómero/metabolismo , Complejo Shelterina , Proteínas de Unión a Telómeros/metabolismo , ADN/genética , ADN/metabolismo
9.
Front Immunol ; 13: 825032, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35273606

RESUMEN

The epidermis, outermost layer of the skin, forms a barrier and is involved in innate and adaptive immunity in an organism. Keratinocytes participate in all these three protective processes. However, a regulator of keratinocyte protective responses against external dangers and stresses remains elusive. We found that upregulation of the orphan gene 2610528A11Rik was a common factor in the skin of mice with several types of inflammation. In the human epidermis, peptide expression of G protein-coupled receptor 15 ligand (GPR15L), encoded by the human ortholog C10orf99, was highly induced in the lesional skin of patients with atopic dermatitis or psoriasis. C10orf99 gene transfection into normal human epidermal keratinocytes (NHEKs) induced the expression of inflammatory mediators and reduced the expression of barrier-related genes. Gene ontology analyses showed its association with translation, mitogen-activated protein kinase (MAPK), mitochondria, and lipid metabolism. Treatment with GPR15L reduced the expression levels of filaggrin and loricrin in human keratinocyte 3D cultures. Instead, their expression levels in mouse primary cultured keratinocytes did not show significant differences between the wild-type and 2610528A11Rik deficient keratinocytes. Lipopolysaccharide-induced expression of Il1b and Il6 was less in 2610528A11Rik deficient mouse keratinocytes than in wild-type, and imiquimod-induced psoriatic dermatitis was blunted in 2610528A11Rik deficient mice. Furthermore, repetitive subcutaneous injection of GPR15L in mouse ears induced skin inflammation in a dose-dependent manner. These results suggest that C10orf99/GPR15L is a primary inducible regulator that reduces the barrier formation and induces the inflammatory response of keratinocytes.


Asunto(s)
Dermatitis Atópica , Queratinocitos , Animales , Péptidos Catiónicos Antimicrobianos/metabolismo , Proteínas de Unión al ADN/metabolismo , Dermatitis Atópica/metabolismo , Humanos , Inflamación/genética , Inflamación/metabolismo , Queratinocitos/metabolismo , Ligandos , Ratones , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
10.
J Invest Dermatol ; 142(6): 1670-1681.e12, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-34740582

RESUMEN

Nicotinamide (NAM), a NAM adenine dinucleotide precursor, is known for its benefits to skin health. Under standard culture conditions, NAM delays the differentiation and enhances the proliferation of human primary keratinocytes, leading to the maintenance of stem cells. In this study, we investigated the effects of NAM on photoaging in two-dimensional human primary keratinocyte cultures and three-dimensional organotypic epidermal models. In both models, we found that UVB irradiation and hydrogen peroxide induced human primary keratinocyte premature terminal differentiation and senescence. In three-dimensional organotypics, the phenotype was characterized by a thickening of the granular layer expressing filaggrin and loricrin, but thinning of the epidermis overall. NAM limited premature differentiation and ameliorated senescence, as evidenced by the maintenance of lamin B1 levels in both models, with decreased lipofuscin staining and reduced IL-6/IL-8 secretion in three-dimensional models, compared to those in UVB-only controls. In addition, DNA damage observed after irradiation was accompanied by a decline in energy metabolism, whereas both effects were partially prevented by NAM. Our data thus highlight the protective effects of NAM against photoaging and oxidative stress in the human epidermis and pinpoint DNA repair and energy metabolism as crucial underlying mechanisms.


Asunto(s)
Envejecimiento de la Piel , Humanos , Queratinocitos/metabolismo , Niacinamida/farmacología , Estrés Oxidativo , Rayos Ultravioleta/efectos adversos
11.
EMBO Rep ; 10(7): 714-21, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19543232

RESUMEN

For much of the last century, the differentiated state that characterizes the many cell types of an adult organism was thought to be stable and abrogated only in rare instances by transdifferentiation, metaplasia or cancer. This stability was thought to reside in the autoregulatory molecular circuitry that exists between the cytoplasm and the nucleus, a status quo that could be disrupted during somatic cell nuclear transfer, to reprogramme cells to a pluripotent state. Pioneering work in the 1980s showed that transdifferentiation of cell lineages could be induced by the addition of transcription factors. However, these conversions were usually confined to cell types from the same germ layer, and proof of conversion was difficult to obtain. This deficiency has now been overturned by demonstrations that exogenously added transcription factors can convert differentiated cell types into embryonic-like induced pluripotent stem cells. Here, we highlight the recent progress, and the implications of this work for our understanding of the relationship between the pluripotent and more differentiated cell states.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes/citología , Animales , Reprogramación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Humanos , Cinética , Células Madre Pluripotentes/metabolismo
12.
Med ; 2(4): 353-354, 2021 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-35590156

RESUMEN

Hutchinson-Gilford Progeria is an accelerated aging syndrome caused by permanently farnesylated mutant lamin A, termed progerin. Recently, the FDA approved Lonafarnib, a farnesyltransferase inhibitor, to treat progeria, while Koblan and colleagues used novel gene editing methods to target the root cause of this disease by correcting the LMNA mutation.


Asunto(s)
Progeria , Envejecimiento , Inhibidores Enzimáticos/uso terapéutico , Humanos , Lamina Tipo A/genética , Mutación , Progeria/tratamiento farmacológico
13.
Mech Ageing Dev ; 198: 111525, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34166688

RESUMEN

The skin is comprised of different cell types with different proliferative capacities. Skin aging occurs with chronological age and upon exposure to extrinsic factors such as photodamage. During aging, senescent cells accumulate in different compartments of the human skin, leading to impaired skin physiology. Diverse skin cell types may respond differently to senescence-inducing stimuli and it is not clear how this results in aging-associated skin phenotypes and pathologies. This review aims to examine and provide an overview of current evidence of cellular senescence in the skin. We will focus on cellular characteristics and behaviour of different skin cell types undergoing senescence in the epidermis and dermis, with a particular focus on the complex interplay between mitochondrial dysfunction, autophagy and DNA damage pathways. We will also examine how the dermis and epidermis cope with the accumulation of DNA damage during aging.


Asunto(s)
Envejecimiento , Daño del ADN/fisiología , Envejecimiento de la Piel/patología , Piel , Envejecimiento/patología , Envejecimiento/fisiología , Autofagia , Senescencia Celular/fisiología , Humanos , Mitocondrias/fisiología , Piel/metabolismo , Piel/patología
14.
Cell Cycle ; 19(24): 3508-3520, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33305692

RESUMEN

Aneuploidy is the condition of having an imbalanced karyotype, which is associated with tumor initiation, evolution, and acquisition of drug-resistant features, possibly by generating heterogeneous populations of cells with distinct genotypes and phenotypes. Multicellular eukaryotes have therefore evolved a range of extrinsic and cell-autonomous mechanisms for restraining proliferation of aneuploid cells, including activation of the tumor suppressor protein p53. However, accumulating evidence indicates that a subset of aneuploid cells can escape p53-mediated growth restriction and continue proliferating in vitro. Here we show that such aneuploid cell lines display a robust modal karyotype and low frequency of chromosomal aberrations despite ongoing chromosome instability. Indeed, while these aneuploid cells are able to survive for extended periods in vitro, their chromosomally unstable progeny remain subject to p53-induced senescence and growth restriction, leading to subsequent elimination from the aneuploid pool. This mechanism helps maintain low levels of heterogeneity in aneuploid populations and may prevent detrimental evolutionary processes such as cancer progression and development of drug resistance.


Asunto(s)
Aneuploidia , Senescencia Celular/genética , Células Epiteliales/metabolismo , Epitelio Pigmentado de la Retina/citología , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control del Ciclo Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Inestabilidad Cromosómica/genética , Segregación Cromosómica/genética , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Cariotipo , Proteína p53 Supresora de Tumor/genética
15.
Aging Cell ; 19(3): e13108, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32087607

RESUMEN

Hutchinson-Gilford progeria is a premature aging syndrome caused by a truncated form of lamin A called progerin. Progerin expression results in a variety of cellular defects including heterochromatin loss, DNA damage, impaired proliferation and premature senescence. It remains unclear how these different progerin-induced phenotypes are temporally and mechanistically linked. To address these questions, we use a doxycycline-inducible system to restrict progerin expression to different stages of the cell cycle. We find that progerin expression leads to rapid and widespread loss of heterochromatin in G1-arrested cells, without causing DNA damage. In contrast, progerin triggers DNA damage exclusively during late stages of DNA replication, when heterochromatin is normally replicated, and preferentially in cells that have lost heterochromatin. Importantly, removal of progerin from G1-arrested cells restores heterochromatin levels and results in no permanent proliferative impediment. Taken together, these results delineate the chain of events that starts with progerin expression and ultimately results in premature senescence. Moreover, they provide a proof of principle that removal of progerin from quiescent cells restores heterochromatin levels and their proliferative capacity to normal levels.


Asunto(s)
Daño del ADN/genética , Heterocromatina/metabolismo , Lamina Tipo A/metabolismo , Progeria/metabolismo , Transducción de Señal/genética , Envejecimiento Prematuro/genética , Envejecimiento Prematuro/metabolismo , Proliferación Celular/genética , Células Cultivadas , Senescencia Celular/genética , Daño del ADN/efectos de los fármacos , Replicación del ADN/genética , Doxorrubicina/farmacología , Fibroblastos/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Expresión Génica , Humanos , Lamina Tipo A/genética , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Progeria/genética
16.
Oncogene ; 39(9): 2030, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31754212

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

17.
Mol Cell Biol ; 26(13): 4911-9, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16782879

RESUMEN

In cancer cells and germ cells, shortening of chromosome ends is prevented by telomerase. Telomerase-deficient cells have a replicative life span, after which they enter senescence. Senescent cells can give rise to survivors that maintain chromosome ends through recombination-based amplification of telomeric or subtelomeric repeats. We found that in Trypanosoma brucei, critically short telomeres are stable in the absence of telomerase. Telomere stabilization ensured genomic integrity and could have implications for telomere maintenance in human telomerase-deficient cells. Cloning and sequencing revealed 7 to 27 TTAGGG repeats on stabilized telomeres and no changes in the subtelomeric region. Clones with short telomeres were used to study telomere elongation dynamics, which differed dramatically at transcriptionally active and silent telomeres, after restoration of telomerase. We propose that transcription makes the termini of short telomeres accessible for rapid elongation by telomerase and that telomere elongation in T. brucei is not regulated by a protein-counting mechanism. Many minichromosomes were lost after long-term culture in the absence of telomerase, which may reflect their different mitotic segregation properties.


Asunto(s)
ADN Protozoario/metabolismo , Telomerasa/genética , Telómero/genética , Telómero/metabolismo , Trypanosoma brucei brucei/enzimología , Trypanosoma brucei brucei/genética , Animales , Cromosomas/metabolismo , Secuencias Repetitivas de Ácidos Nucleicos/genética , Eliminación de Secuencia , Telomerasa/metabolismo , Telómero/enzimología , Transcripción Genética
18.
J Dermatol Sci ; 96(2): 58-65, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31727429

RESUMEN

Aging is an inevitable consequence of human life resulting in a gradual deterioration of cell, tissue and organismal function and an increased risk to develop chronic ailments. Premature aging syndromes, also known as progeroid syndromes, recapitulate many clinical features of normal aging and offer a unique opportunity to elucidate fundamental mechanisms that contribute to human aging. Progeroid syndromes can be broadly classified into those caused by perturbations of the nuclear lamina, a meshwork of proteins located underneath the inner nuclear membrane (laminopathies); and a second group that is caused by mutations that directly impair DNA replication and repair. We will focus mainly on laminopathies caused by incorrect processing of lamin A, an intermediate filament protein that resides at the nuclear periphery. Hutchinson-Gilford Progeria (HGPS) is an accelerated aging syndrome caused by a mutation in lamin A and one of the best studied laminopathies. HGPS patients exhibit clinical characteristics of premature aging, including alopecia, aberrant pigmentation, loss of subcutaneous fat and die in their teens as a result of atherosclerosis and cardiovascular complications. Here we summarize how cell- and mouse-based disease models provided mechanistic insights into human aging and discuss experimental strategies under consideration for the treatment of these rare genetic disorders.


Asunto(s)
Envejecimiento Prematuro/diagnóstico , Envejecimiento Prematuro/genética , Lamina Tipo A/genética , Lámina Nuclear/metabolismo , Envejecimiento , Animales , Núcleo Celular/metabolismo , Senescencia Celular , Cromatina/metabolismo , Contractura/congénito , Contractura/diagnóstico , Contractura/genética , Daño del ADN , Reparación del ADN , Replicación del ADN , Heterocromatina , Humanos , Ratones , Mutación , Proteínas Nucleares/metabolismo , Progeria/diagnóstico , Progeria/genética , Precursores de Proteínas/genética , Anomalías Cutáneas/diagnóstico , Anomalías Cutáneas/genética , Telómero/metabolismo , Síndrome de Werner/diagnóstico , Síndrome de Werner/genética
19.
Oncogene ; 38(7): 998-1018, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30190546

RESUMEN

Cell cycle regulation, especially faithful DNA replication and mitosis, are crucial to maintain genome stability. Cyclin-dependent kinase (CDK)/cyclin complexes drive most processes in cellular proliferation. In response to DNA damage, cell cycle surveillance mechanisms enable normal cells to arrest and undergo repair processes. Perturbations in genomic stability can lead to tumor development and suggest that cell cycle regulators could be effective targets in anticancer therapy. However, many clinical trials ended in failure due to off-target effects of the inhibitors used. Here, we investigate in vivo the importance of WEE1- and MYT1-dependent inhibitory phosphorylation of mammalian CDK1. We generated Cdk1AF knockin mice, in which two inhibitory phosphorylation sites are replaced by the non-phosphorylatable amino acids T14A/Y15F. We uncovered that monoallelic expression of CDK1AF is early embryonic lethal in mice and induces S phase arrest accompanied by γH2AX and DNA damage checkpoint activation in mouse embryonic fibroblasts (MEFs). The chromosomal fragmentation in Cdk1AF MEFs does not rely on CDK2 and is partly caused by premature activation of MUS81-SLX4 structure-specific endonuclease complexes, as well as untimely onset of chromosome condensation followed by nuclear lamina disassembly. We provide evidence that tumor development in liver expressing CDK1AF is inhibited. Interestingly, the regulatory mechanisms that impede cell proliferation in CDK1AF expressing cells differ partially from the actions of the WEE1 inhibitor, MK-1775, with p53 expression determining the sensitivity of cells to the drug response. Thus, our work highlights the importance of improved therapeutic strategies for patients with various cancer types and may explain why some patients respond better to WEE1 inhibitors.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Pérdida del Embrión/enzimología , Embrión de Mamíferos/enzimología , Mitosis , Fase S , Sustitución de Aminoácidos , Animales , Proteína Quinasa CDC2/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Pérdida del Embrión/genética , Pérdida del Embrión/patología , Embrión de Mamíferos/patología , Activación Enzimática , Ratones , Ratones Transgénicos , Mutación Missense , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
20.
Nat Commun ; 10(1): 4990, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31740672

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a genetic disorder characterized by premature aging features. Cells from HGPS patients express progerin, a truncated form of Lamin A, which perturbs cellular homeostasis leading to nuclear shape alterations, genome instability, heterochromatin loss, telomere dysfunction and premature entry into cellular senescence. Recently, we reported that telomere dysfunction induces the transcription of telomeric non-coding RNAs (tncRNAs) which control the DNA damage response (DDR) at dysfunctional telomeres. Here we show that progerin-induced telomere dysfunction induces the transcription of tncRNAs. Their functional inhibition by sequence-specific telomeric antisense oligonucleotides (tASOs) prevents full DDR activation and premature cellular senescence in various HGPS cell systems, including HGPS patient fibroblasts. We also show in vivo that tASO treatment significantly enhances skin homeostasis and lifespan in a transgenic HGPS mouse model. In summary, our results demonstrate an important role for telomeric DDR activation in HGPS progeroid detrimental phenotypes in vitro and in vivo.


Asunto(s)
Daño del ADN , Progeria/patología , Telómero/metabolismo , Animales , Línea Celular , Proliferación Celular , Senescencia Celular , Reparación del ADN , Modelos Animales de Enfermedad , Homeostasis , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Ratones , Mutación/genética , Oligonucleótidos Antisentido/metabolismo , Fenotipo , ARN no Traducido/genética , ARN no Traducido/metabolismo , Piel/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA