Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 156(3): 400-7, 2014 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-24485450

RESUMO

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


Assuntos
Progéria/tratamento farmacológico , Progéria/fisiopatologia , Pesquisa Translacional Biomédica , Envelhecimento/genética , Envelhecimento/patologia , Criança , Farnesiltranstransferase/antagonistas & inibidores , Humanos , Lamina Tipo A , Proteínas Nucleares/metabolismo , Progéria/genética , Progéria/patologia , Precursores de Proteínas/metabolismo
2.
Genes Dev ; 29(19): 2022-36, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26443848

RESUMO

Lamina-associated polypeptide 2α (LAP2α) localizes throughout the nucleoplasm and interacts with the fraction of lamins A/C that is not associated with the peripheral nuclear lamina. The LAP2α-lamin A/C complex negatively affects cell proliferation. Lamins A/C are encoded by LMNA, a single heterozygous mutation of which causes Hutchinson-Gilford progeria syndrome (HGPS). This mutation generates the lamin A variant progerin, which we show here leads to loss of LAP2α and nucleoplasmic lamins A/C, impaired proliferation, and down-regulation of extracellular matrix components. Surprisingly, contrary to wild-type cells, ectopic expression of LAP2α in cells expressing progerin restores proliferation and extracellular matrix expression but not the levels of nucleoplasmic lamins A/C. We conclude that, in addition to its cell cycle-inhibiting function with lamins A/C, LAP2α can also regulate extracellular matrix components independently of lamins A/C, which may help explain the proliferation-promoting function of LAP2α in cells expressing progerin.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo , Proteínas da Matriz Extracelular/genética , Proteínas de Membrana/metabolismo , Progéria/fisiopatologia , Linhagem Celular , Proliferação de Células/genética , Células Cultivadas , Proteínas de Ligação a DNA/genética , Humanos , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas de Membrana/genética , Progéria/genética , Regulação para Cima
3.
Nat Rev Mol Cell Biol ; 11(8): 567-78, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20651707

RESUMO

One of the many debated topics in ageing research is whether progeroid syndromes are really accelerated forms of human ageing. The answer requires a better understanding of the normal ageing process and the molecular pathology underlying these rare diseases. Exciting recent findings regarding a severe human progeria, Hutchinson-Gilford progeria syndrome, have implicated molecular changes that are also linked to normal ageing, such as genome instability, telomere attrition, premature senescence and defective stem cell homeostasis in disease development. These observations, coupled with genetic studies of longevity, lead to a hypothesis whereby progeria syndromes accelerate a subset of the pathological changes that together drive the normal ageing process.


Assuntos
Envelhecimento , Progéria/etiologia , Envelhecimento/genética , Envelhecimento/fisiologia , Animais , Evolução Biológica , Dano ao DNA , Reparo do DNA , Humanos , Lamina Tipo A/genética , Longevidade/efeitos dos fármacos , Longevidade/genética , Longevidade/fisiologia , Células-Tronco Mesenquimais/patologia , Células-Tronco Mesenquimais/fisiologia , Modelos Biológicos , Progéria/genética , Progéria/fisiopatologia , Progéria/terapia , Transdução de Sinais , Sirolimo/farmacologia , Síndrome
4.
Proc Natl Acad Sci U S A ; 116(9): 3578-3583, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30808750

RESUMO

Studies of the accelerated aging disorder Hutchinson-Gilford progeria syndrome (HGPS) can potentially reveal cellular defects associated with physiological aging. HGPS results from expression and abnormal nuclear envelope association of a farnesylated, truncated variant of prelamin A called "progerin." We surveyed the diffusional mobilities of nuclear membrane proteins to identify proximal effects of progerin expression. The mobilities of three proteins-SUN2, nesprin-2G, and emerin-were reduced in fibroblasts from children with HGPS compared with those in normal fibroblasts. These proteins function together in nuclear movement and centrosome orientation in fibroblasts polarizing for migration. Both processes were impaired in fibroblasts from children with HGPS and in NIH 3T3 fibroblasts expressing progerin, but were restored by inhibiting protein farnesylation. Progerin affected both the coupling of the nucleus to actin cables and the oriented flow of the cables necessary for nuclear movement and centrosome orientation. Progerin overexpression increased levels of SUN1, which couples the nucleus to microtubules through nesprin-2G and dynein, and microtubule association with the nucleus. Reducing microtubule-nuclear connections through SUN1 depletion or dynein inhibition rescued the polarity defects. Nuclear movement and centrosome orientation were also defective in fibroblasts from normal individuals over 60 y, and both defects were rescued by reducing the increased level of SUN1 in these cells or inhibiting dynein. Our results identify imbalanced nuclear engagement of the cytoskeleton (microtubules: high; actin filaments: low) as the basis for intrinsic cell polarity defects in HGPS and physiological aging and suggest that rebalancing the connections can ameliorate the defects.


Assuntos
Envelhecimento/genética , Lamina Tipo A/genética , Proteínas de Membrana/genética , Proteínas dos Microfilamentos/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Progéria/genética , Envelhecimento/patologia , Animais , Núcleo Celular/genética , Polaridade Celular/genética , Dineínas/química , Dineínas/genética , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Lamina Tipo A/química , Proteínas de Membrana/química , Camundongos , Proteínas dos Microfilamentos/química , Células NIH 3T3 , Proteínas do Tecido Nervoso/química , Membrana Nuclear/genética , Proteínas Nucleares/química , Progéria/fisiopatologia , Prenilação de Proteína
5.
Annu Rev Physiol ; 80: 27-48, 2018 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-28934587

RESUMO

Aging, the main risk factor for cardiovascular disease (CVD), is becoming progressively more prevalent in our societies. A better understanding of how aging promotes CVD is therefore urgently needed to develop new strategies to reduce disease burden. Atherosclerosis and heart failure contribute significantly to age-associated CVD-related morbimortality. CVD and aging are both accelerated in patients suffering from Hutchinson-Gilford progeria syndrome (HGPS), a rare genetic disorder caused by the prelamin A mutant progerin. Progerin causes extensive atherosclerosis and cardiac electrophysiological alterations that invariably lead to premature aging and death. This review summarizes the main structural and functional alterations to the cardiovascular system during physiological and premature aging and discusses the mechanisms underlying exaggerated CVD and aging induced by prelamin A and progerin. Because both proteins are expressed in normally aging non-HGPS individuals, and most hallmarks of normal aging occur in progeria, research on HGPS can identify mechanisms underlying physiological aging.


Assuntos
Envelhecimento/metabolismo , Doenças Cardiovasculares/metabolismo , Sistema Cardiovascular/metabolismo , Progéria/metabolismo , Calcificação Vascular/metabolismo , Animais , Doenças Cardiovasculares/fisiopatologia , Sistema Cardiovascular/fisiopatologia , Humanos , Progéria/fisiopatologia , Calcificação Vascular/fisiopatologia
6.
Clin Genet ; 99(1): 3-28, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32860237

RESUMO

Progeroid disorders make up a heterogeneous group of very rare hereditary diseases characterized by clinical signs that often mimic physiological aging in a premature manner. Apart from Hutchinson-Gilford progeria syndrome, one of the best-investigated progeroid disorders, a wide spectrum of other premature aging phenotypes exist, which differ significantly in their clinical presentation and molecular pathogenesis. Next-generation sequencing (NGS)-based approaches have made it feasible to determine the molecular diagnosis in the early stages of a disease. Nevertheless, a broad clinical knowledge on these disorders and their associated symptoms is still fundamental for a comprehensive patient management and for the interpretation of variants of unknown significance from NGS data sets. This review provides a detailed overview on characteristic clinical features and underlying molecular genetics of well-known as well as only recently identified premature aging disorders and also highlights novel findings towards future therapeutic options.


Assuntos
Senilidade Prematura/genética , Envelhecimento/genética , Progéria/genética , Envelhecimento/patologia , Senilidade Prematura/diagnóstico , Senilidade Prematura/fisiopatologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Patologia Molecular , Fenótipo , Progéria/diagnóstico , Progéria/fisiopatologia
7.
Nephrol Dial Transplant ; 35(12): 2036-2045, 2020 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31302696

RESUMO

The cytoprotective transcriptor factor nuclear factor erythroid 2- related factor 2 (NRF2) is part of a complex regulatory network that responds to environmental cues. To better understand its role in a cluster of inflammatory and pro-oxidative burden of lifestyle diseases that accumulate with age, lessons can be learned from evolution, the animal kingdom and progeroid syndromes. When levels of oxygen increased in the atmosphere, mammals required ways to protect themselves from the metabolic toxicity that arose from the production of reactive oxygen species. The evolutionary origin of the NRF2-Kelch-like ECH-associated protein 1 (KEAP1) signalling pathway from primitive origins has been a prerequisite for a successful life on earth, with checkpoints in antioxidant gene expression, inflammation, detoxification and protein homoeostasis. Examples from the animal kingdom suggest that superior antioxidant defense mechanisms with enhanced NRF2 expression have been developed during evolution to protect animals during extreme environmental conditions, such as deep sea diving, hibernation and habitual hypoxia. The NRF2-KEAP1 signalling pathway is repressed in progeroid (accelerated ageing) syndromes and a cluster of burden of lifestyle disorders that accumulate with age. Compelling links exist between tissue hypoxia, senescence and a repressed NRF2 system. Effects of interventions that activate NRF2, including nutrients, and more potent (semi)synthetic NRF2 agonists on clinical outcomes are of major interest. Given the broad-ranging actions of NRF2, we need to better understand the mechanisms of activation, biological function and regulation of NRF2 and its inhibitor, KEAP1, in different clinical conditions to ensure that modulation of this thiol-based system will not result in major adverse effects. Lessons from evolution, the animal kingdom and conditions of accelerated ageing clarify a major role of a controlled NRF2-KEAP1 system in healthy ageing and well-being.


Assuntos
Evolução Biológica , Citoproteção , Fator 2 Relacionado a NF-E2/metabolismo , Progéria/prevenção & controle , Progéria/fisiopatologia , Doenças Raras/prevenção & controle , Doenças Raras/fisiopatologia , Animais , Humanos , Estresse Oxidativo , Progéria/metabolismo , Doenças Raras/metabolismo , Síndrome
8.
Pak J Pharm Sci ; 33(3): 1179-1182, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-33191246

RESUMO

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


Assuntos
Senilidade Prematura , Lamina Tipo A/genética , Mutação Puntual , Progéria/terapia , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Fenótipo , Progéria/genética , Progéria/patologia , Progéria/fisiopatologia , Prognóstico
9.
Proc Natl Acad Sci U S A ; 113(46): E7250-E7259, 2016 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-27799555

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a rare genetic disease caused by defective prelamin A processing, leading to nuclear lamina alterations, severe cardiovascular pathology, and premature death. Prelamin A alterations also occur in physiological aging. It remains unknown how defective prelamin A processing affects the cardiac rhythm. We show age-dependent cardiac repolarization abnormalities in HGPS patients that are also present in the Zmpste24-/- mouse model of HGPS. Challenge of Zmpste24-/- mice with the ß-adrenergic agonist isoproterenol did not trigger ventricular arrhythmia but caused bradycardia-related premature ventricular complexes and slow-rate polymorphic ventricular rhythms during recovery. Patch-clamping in Zmpste24-/- cardiomyocytes revealed prolonged calcium-transient duration and reduced sarcoplasmic reticulum calcium loading and release, consistent with the absence of isoproterenol-induced ventricular arrhythmia. Zmpste24-/- progeroid mice also developed severe fibrosis-unrelated bradycardia and PQ interval and QRS complex prolongation. These conduction defects were accompanied by overt mislocalization of the gap junction protein connexin43 (Cx43). Remarkably, Cx43 mislocalization was also evident in autopsied left ventricle tissue from HGPS patients, suggesting intercellular connectivity alterations at late stages of the disease. The similarities between HGPS patients and progeroid mice reported here strongly suggest that defective cardiac repolarization and cardiomyocyte connectivity are important abnormalities in the HGPS pathogenesis that increase the risk of arrhythmia and premature death.


Assuntos
Arritmias Cardíacas/fisiopatologia , Doença do Sistema de Condução Cardíaco/fisiopatologia , Progéria/fisiopatologia , Adolescente , Adulto , Animais , Arritmias Cardíacas/metabolismo , Cálcio/fisiologia , Doença do Sistema de Condução Cardíaco/metabolismo , Criança , Pré-Escolar , Conexina 43/metabolismo , Conexina 43/fisiologia , Feminino , Coração/fisiologia , Humanos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Metaloendopeptidases/genética , Metaloendopeptidases/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Lâmina Nuclear/fisiologia , Progéria/metabolismo , Retículo Sarcoplasmático/fisiologia , Adulto Jovem
10.
Hum Genet ; 137(11-12): 921-939, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30450527

RESUMO

Juvenile segmental progeroid syndromes are rare, heterogeneous disorders characterized by signs of premature aging affecting more than one tissue or organ starting in childhood. Hutchinson-Gilford progeria syndrome (HGPS), caused by a recurrent de novo synonymous LMNA mutation resulting in aberrant splicing and generation of a mutant product called progerin, is a prototypical example of such disorders. Here, we performed a joint collaborative study using massively parallel sequencing and targeted Sanger sequencing, aimed at delineating the underlying genetic cause of 14 previously undiagnosed, clinically heterogeneous, non-LMNA-associated juvenile progeroid patients. The molecular diagnosis was achieved in 11 of 14 cases (~ 79%). Furthermore, we firmly establish biallelic mutations in POLR3A as the genetic cause of a recognizable, neonatal, Wiedemann-Rautenstrauch-like progeroid syndrome. Thus, we suggest that POLR3A mutations are causal for a portion of under-diagnosed early-onset segmental progeroid syndromes. We additionally expand the clinical spectrum associated with PYCR1 mutations by showing that they can somewhat resemble HGPS in the first year of life. Moreover, our results lead to clinical reclassification in one single case. Our data emphasize the complex genetic and clinical heterogeneity underlying progeroid disorders.


Assuntos
Retardo do Crescimento Fetal/genética , Progéria/genética , Pirrolina Carboxilato Redutases/genética , RNA Polimerase III/genética , Adolescente , Processamento Alternativo/genética , Criança , Feminino , Retardo do Crescimento Fetal/diagnóstico , Retardo do Crescimento Fetal/patologia , Predisposição Genética para Doença , Humanos , Lactente , Lamina Tipo A/genética , Masculino , Mutação , Fenótipo , Progéria/diagnóstico , Progéria/patologia , Progéria/fisiopatologia , delta-1-Pirrolina-5-Carboxilato Redutase
11.
Small ; 13(15)2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28211642

RESUMO

Organ-on-a-chip platforms seek to recapitulate the complex microenvironment of human organs using miniaturized microfluidic devices. Besides modeling healthy organs, these devices have been used to model diseases, yielding new insights into pathophysiology. Hutchinson-Gilford progeria syndrome (HGPS) is a premature aging disease showing accelerated vascular aging, leading to the death of patients due to cardiovascular diseases. HGPS targets primarily vascular cells, which reside in mechanically active tissues. Here, a progeria-on-a-chip model is developed and the effects of biomechanical strain are examined in the context of vascular aging and disease. Physiological strain induces a contractile phenotype in primary smooth muscle cells (SMCs), while a pathological strain induces a hypertensive phenotype similar to that of angiotensin II treatment. Interestingly, SMCs derived from human induced pluripotent stem cells of HGPS donors (HGPS iPS-SMCs), but not from healthy donors, show an exacerbated inflammatory response to strain. In particular, increased levels of inflammation markers as well as DNA damage are observed. Pharmacological intervention reverses the strain-induced damage by shifting gene expression profile away from inflammation. The progeria-on-a-chip is a relevant platform to study biomechanics in vascular biology, particularly in the setting of vascular disease and aging, while simultaneously facilitating the discovery of new drugs and/or therapeutic targets.


Assuntos
Progressão da Doença , Inflamação/patologia , Dispositivos Lab-On-A-Chip , Progéria/fisiopatologia , Angiotensina II/farmacologia , Fenômenos Biomecânicos , Vasos Sanguíneos/patologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lovastatina/farmacologia , Microfluídica , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Fenótipo
12.
BMC Med Genet ; 18(1): 116, 2017 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-29047356

RESUMO

BACKGROUND: Mutations in LMNA gene, encoding Lamin A/C, cause a diverse array of phenotypes, collectively referred to as laminopathies. The most common manifestation is dilated cardiomyopathy (DCM), occurring in conjunction with variable skeletal muscle involvement but without involvement of the coronary arteries. Much less commonly, LMNA mutations cause progeroid syndromes, whereby an early-onset coronary artery disease (CAD) is the hallmark of the disease. We report a hitherto unreported compound cardiac phenotype, dubbed as "non-syndromic cardiac progeria", in a young patient who carried a rare pathogenic variant in the LMNA gene and developed progressive degeneration of various cardiac structures, as seen in the elderly. The phenotype resembled the progeroid syndromes, except that it was restricted to the heart and did not involve other organs. CASE PRESENTATION: The patient was a well-developed Caucasian female who presented at age 29 years with an acute myocardial infarction (MI) and was found to have extensive CAD. She had none of the conventional risk factors for atherosclerosis. She underwent coronary artery bypass surgery but continued to require multiple percutaneous coronary interventions for symptomatic obstructive coronary lesions. During the course of next 10 years, she developed mitral regurgitation, degenerative mitral and aortic valve diseases, atrial flutter, and progressive conduction defects. She died from progressive heart failure with predominant involvement of the right ventricle and severe tricuspid regurgitation. Cardiac phenotype in this young patient resembled degenerative cardiac diseases of the elderly and the progeroid syndromes. However, in contrast to the progeroid syndromes, the phenotype was restricted to the heart and did not involve other organs. Thus, the phenotype was dubbed as a non-syndromic cardiac progeria. Genetic screening of several cardiomyopathy genes, including LMNA, which is a causal gene for progeroid syndromes, led to identification of a very rare pathogenic p.Asp300Asn variant in the LMNA gene. CONCLUSIONS: We infer that the LMNA p.Asp300Asn mutation is pathogenic in non-syndromic cardiac progeria. Mutations involving codon 300 in the LMNA gene have been associated with progeroid syndromes involving multiple organs. Collectively, the data provide credence to the causal role of p.Asp300Asn mutation in the pathogenesis of non-syndromic cardiac progeria.


Assuntos
Lamina Tipo A/genética , Mutação de Sentido Incorreto , Progéria/genética , Adulto , Sequência de Aminoácidos , Asparagina/genética , Ácido Aspártico/genética , Sequência de Bases , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/fisiopatologia , Ecocardiografia/métodos , Eletrocardiografia/métodos , Evolução Fatal , Feminino , Humanos , Infarto do Miocárdio/diagnóstico , Infarto do Miocárdio/genética , Infarto do Miocárdio/fisiopatologia , Linhagem , Progéria/diagnóstico , Progéria/fisiopatologia , Homologia de Sequência de Aminoácidos
13.
Exp Dermatol ; 26(10): 889-895, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28192606

RESUMO

Metformin is a popular antidiabetic biguanide, which has been considered as a candidate drug for cancer treatment and ageing prevention. Hutchinson-Gilford progeria syndrome (HGPS) is a devastating disease characterized by premature ageing and severe age-associated complications leading to death. The effects of metformin on HGPS dermal fibroblasts remain largely undefined. In this study, we investigated whether metformin could exert a beneficial effect on nuclear abnormalities and delay senescence in fibroblasts derived from HGPS patients. Metformin treatment partially restored normal nuclear phenotypes, delayed senescence, activated the phosphorylation of AMP-activated protein kinase and decreased reactive oxygen species formation in HGPS dermal fibroblasts. Interestingly, metformin reduced the number of phosphorylated histone variant H2AX-positive DNA damage foci and suppressed progerin protein expression, compared to the control. Furthermore, metformin-supplemented aged mice showed higher splenocyte proliferation and mRNA expression of the antioxidant enzyme, superoxide dismutase 2 than the control mice. Collectively, our results show that metformin treatment alleviates the nuclear defects and premature ageing phenotypes in HGPS fibroblasts. Thus, metformin can be considered a promising therapeutic approach for life extension in HGPS.


Assuntos
Proliferação de Células/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Progéria/fisiopatologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Núcleo Celular/metabolismo , Criança , Pré-Escolar , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Lamina Tipo A/metabolismo , Camundongos , Proteínas Nucleares/metabolismo , Fenótipo , Fosforilação/efeitos dos fármacos , Progéria/patologia , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Pele/patologia , Baço/citologia , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo
14.
Am J Med Genet A ; 173(9): 2422-2427, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28639748

RESUMO

Skeletal overgrowth is a characteristic of several genetic disorders that are linked to specific molecular signaling cascades. Recently, we established a novel overgrowth syndrome (Kosaki overgrowth syndrome, OMIM #616592) arising from a de novo mutation in PDGFRB, that is, c.1751C>G p.(Pro584Arg). Subsequently, other investigators provided in vitro molecular evidence that this specific mutation in the juxtamembrane domain of PDGFRB causes an overgrowth phenotype and is the first gain-of-function point mutation of PDGFRB to be reported in humans. Here, we report the identification of a mutation in PDGFRB, c.1696T>C p.(Trp566Arg), in two unrelated patients with skeletal overgrowth, further confirming the existence of PDGFRB-related overgrowth syndrome arising from mutations in the juxtamembrane domain of PDGFRB. A review of all four of these patients with an overgrowth phenotype and PDGFRB mutations revealed postnatal skeletal overgrowth, premature aging, cognitive impairment, neurodegeneration, and a prominent connective tissue component to this complex phenotype. From a functional standpoint, hypermorphic mutations in PDGFRB lead to Kosaki overgrowth syndrome, infantile myofibromatosis (OMIM #228550), and Penttinen syndrome (OMIM #601812), whereas hypomorphic mutations lead to idiopathic basal ganglia calcification (OMIM #615007). In conclusion, a specific class of mutations in PDGFRB causes a clinically recognizable syndromic form of skeletal overgrowth.


Assuntos
Acro-Osteólise/genética , Doenças dos Gânglios da Base/genética , Calcinose/genética , Deformidades Congênitas dos Membros/genética , Miofibromatose/congênito , Progéria/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética , Acro-Osteólise/fisiopatologia , Doenças dos Gânglios da Base/fisiopatologia , Osso e Ossos/fisiopatologia , Calcinose/fisiopatologia , Feminino , Humanos , Deformidades Congênitas dos Membros/fisiopatologia , Masculino , Miofibromatose/genética , Miofibromatose/fisiopatologia , Fenótipo , Mutação Puntual , Progéria/fisiopatologia , Transdução de Sinais/genética
15.
Nature ; 472(7342): 221-5, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21346760

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a rare and fatal human premature ageing disease, characterized by premature arteriosclerosis and degeneration of vascular smooth muscle cells (SMCs). HGPS is caused by a single point mutation in the lamin A (LMNA) gene, resulting in the generation of progerin, a truncated splicing mutant of lamin A. Accumulation of progerin leads to various ageing-associated nuclear defects including disorganization of nuclear lamina and loss of heterochromatin. Here we report the generation of induced pluripotent stem cells (iPSCs) from fibroblasts obtained from patients with HGPS. HGPS-iPSCs show absence of progerin, and more importantly, lack the nuclear envelope and epigenetic alterations normally associated with premature ageing. Upon differentiation of HGPS-iPSCs, progerin and its ageing-associated phenotypic consequences are restored. Specifically, directed differentiation of HGPS-iPSCs to SMCs leads to the appearance of premature senescence phenotypes associated with vascular ageing. Additionally, our studies identify DNA-dependent protein kinase catalytic subunit (DNAPKcs, also known as PRKDC) as a downstream target of progerin. The absence of nuclear DNAPK holoenzyme correlates with premature as well as physiological ageing. Because progerin also accumulates during physiological ageing, our results provide an in vitro iPSC-based model to study the pathogenesis of human premature and physiological vascular ageing.


Assuntos
Células-Tronco Pluripotentes Induzidas/patologia , Envelhecimento/metabolismo , Envelhecimento/patologia , Envelhecimento/fisiologia , Senilidade Prematura/genética , Senilidade Prematura/patologia , Senilidade Prematura/fisiopatologia , Proteínas de Ligação ao Cálcio/análise , Diferenciação Celular , Linhagem Celular , Reprogramação Celular , Senescência Celular , Proteína Quinase Ativada por DNA/metabolismo , Epigênese Genética , Fibroblastos/patologia , Holoenzimas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Lamina Tipo A , Proteínas dos Microfilamentos/análise , Modelos Biológicos , Músculo Liso Vascular/patologia , Membrana Nuclear/patologia , Proteínas Nucleares/análise , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fenótipo , Progéria/genética , Progéria/patologia , Progéria/fisiopatologia , Precursores de Proteínas/análise , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Especificidade por Substrato , Calponinas
16.
Biochem Biophys Res Commun ; 473(3): 710-8, 2016 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-26474704

RESUMO

Induced pluripotent stem cells (iPSCs) represent today an invaluable tool to create disease cell models for modeling and drug screening. Several lines of iPSCs have been generated in the last 7 years that changed the paradigm for studying diseases and the discovery of new drugs to treat them. In this article we focus our attention to vascular diseases in particular Hutchinson-Gilford Progeria Syndrome (HGPS), a devastating premature aging disease caused by a mutation in the lamin A gene. In general, patients die because of myocardial infarction or stroke. Because the patients are fragile the isolation of a particular type of cells is very difficult. Therefore in the last 5 years, researchers have used cells derived from iPSCs to model aspects of the HGPS and to screen libraries of chemicals to retard or treat the disease.


Assuntos
Estenose da Valva Aórtica/fisiopatologia , Células-Tronco Pluripotentes Induzidas/citologia , Progéria/fisiopatologia , Doenças Vasculares/metabolismo , Animais , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Doença de Depósito de Glicogênio Tipo II/fisiopatologia , Humanos , Lamina Tipo A/genética , Engenharia Tecidual/métodos , Síndrome de Williams/fisiopatologia
17.
IUBMB Life ; 68(9): 717-21, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27364681

RESUMO

DNA damage caused by various sources remains one of the most researched topics in the area of aging and neurodegeneration. Increased DNA damage causes premature aging. Aging is plastic and is characterised by the decline in the ability of a cell/organism to maintain genomic stability. Lifespan can be modulated by various interventions like calorie restriction, a balanced diet of macro and micronutrients or supplementation with nutrients/nutrient formulations such as Amalaki rasayana, docosahexaenoic acid, resveratrol, curcumin, etc. Increased levels of DNA damage in the form of double stranded and single stranded breaks are associated with decreased longevity in animal models like WNIN/Ob obese rats. Erroneous DNA repair can result in accumulation of DNA damage products, which in turn result in premature aging disorders such as Hutchinson-Gilford progeria syndrome. Epigenomic studies of the aging process have opened a completely new arena for research and development of drugs and therapeutic agents. We propose here that agents or interventions that can maintain epigenomic stability and facilitate the DNA repair process can slow down the progress of premature aging, if not completely prevent it. © 2016 IUBMB Life, 68(9):717-721, 2016.


Assuntos
Envelhecimento/genética , Dano ao DNA/genética , Epigênese Genética/genética , Progéria/dietoterapia , Envelhecimento/efeitos dos fármacos , Envelhecimento/patologia , Animais , Curcumina/uso terapêutico , Reparo do DNA/efeitos dos fármacos , Ácidos Docosa-Hexaenoicos/uso terapêutico , Instabilidade Genômica/efeitos dos fármacos , Humanos , Longevidade/efeitos dos fármacos , Extratos Vegetais/uso terapêutico , Progéria/fisiopatologia , Ratos , Resveratrol , Estilbenos/uso terapêutico
18.
Dev Growth Differ ; 58(1): 116-30, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26691051

RESUMO

Progeria is a devastating disorder in which patients exhibit signs of premature aging. The most well-known progeroid syndromes include Hutchinson-Gilford Progeria Syndrome (HGPS) and Werner Syndrome (WS). While HGPS and WS are rare, they often result in severe age-associated complications starting in the early developmental period or after the pubertal growth spurt during adolescence, respectively. In addition, patients with HGPS ultimately die of diseases normally seen in the elderly population, with stroke and myocardial infarction as the leading causes of death. Many WS patients develop similar cardiovascular complications but also have an increased predisposition to developing multiple rare malignancies. These premature aging disorders, as well as animal and cell culture models that recapitulate these diseases, have provided insight into the genetics and cellular pathways that underlie these human conditions and have also uncovered possible mechanisms behind normal aging. Here we discuss the history, the types of progeria, and the various pathophysiological mechanisms that drive these diseases. We also address recent medical advances and treatment modalities for patients with progeria.


Assuntos
Desenvolvimento do Adolescente , Progéria , Puberdade , Síndrome de Werner , Adolescente , Animais , Feminino , Humanos , Masculino , Progéria/genética , Progéria/metabolismo , Progéria/fisiopatologia , Progéria/terapia , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Síndrome de Werner/fisiopatologia , Síndrome de Werner/terapia
19.
Cell Mol Life Sci ; 71(22): 4373-86, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25080110

RESUMO

Cellular senescence, a permanent state of cell cycle arrest accompanied by a complex phenotype, is an essential mechanism that limits tumorigenesis and tissue damage. In physiological conditions, senescent cells can be removed by the immune system, facilitating tumor suppression and wound healing. However, as we age, senescent cells accumulate in tissues, either because an aging immune system fails to remove them, the rate of senescent cell formation is elevated, or both. If senescent cells persist in tissues, they have the potential to paradoxically promote pathological conditions. Cellular senescence is associated with an enhanced pro-survival phenotype, which most likely promotes persistence of senescent cells in vivo. This phenotype may have evolved to favor facilitation of a short-term wound healing, followed by the elimination of senescent cells by the immune system. In this review, we provide a perspective on the triggers, mechanisms and physiological as well as pathological consequences of senescent cells.


Assuntos
Senescência Celular , Transformação Celular Neoplásica , Dano ao DNA , Matriz Extracelular/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Progéria/metabolismo , Progéria/fisiopatologia , Transdução de Sinais , Síndrome de Werner/metabolismo , Síndrome de Werner/fisiopatologia
20.
Proc Natl Acad Sci U S A ; 109(41): 16666-71, 2012 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-23012407

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare, fatal, segmental premature aging syndrome caused by a mutation in LMNA that produces the farnesylated aberrant lamin A protein, progerin. This multisystem disorder causes failure to thrive and accelerated atherosclerosis leading to early death. Farnesyltransferase inhibitors have ameliorated disease phenotypes in preclinical studies. Twenty-five patients with HGPS received the farnesyltransferase inhibitor lonafarnib for a minimum of 2 y. Primary outcome success was predefined as a 50% increase over pretherapy in estimated annual rate of weight gain, or change from pretherapy weight loss to statistically significant on-study weight gain. Nine patients experienced a ≥50% increase, six experienced a ≥50% decrease, and 10 remained stable with respect to rate of weight gain. Secondary outcomes included decreases in arterial pulse wave velocity and carotid artery echodensity and increases in skeletal rigidity and sensorineural hearing within patient subgroups. All patients improved in one or more of these outcomes. Results from this clinical treatment trial for children with HGPS provide preliminary evidence that lonafarnib may improve vascular stiffness, bone structure, and audiological status.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Farnesiltranstransferase/antagonistas & inibidores , Piperidinas/uso terapêutico , Progéria/tratamento farmacológico , Piridinas/uso terapêutico , Adolescente , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Criança , Pré-Escolar , Diarreia/induzido quimicamente , Relação Dose-Resposta a Droga , Esquema de Medicação , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Farnesiltranstransferase/metabolismo , Fadiga/induzido quimicamente , Feminino , Humanos , Masculino , Piperidinas/efeitos adversos , Piperidinas/farmacocinética , Progéria/patologia , Progéria/fisiopatologia , Análise de Onda de Pulso , Piridinas/efeitos adversos , Piridinas/farmacocinética , Resultado do Tratamento , Vômito/induzido quimicamente , Aumento de Peso/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA