Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
1.
Cell ; 156(3): 400-7, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485450

RESUMEN

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.


Asunto(s)
Progeria/tratamiento farmacológico , Progeria/fisiopatología , Investigación Biomédica Traslacional , Envejecimiento/genética , Envejecimiento/patología , Niño , Farnesiltransferasa/antagonistas & inhibidores , Humanos , Lamina Tipo A , Proteínas Nucleares/metabolismo , Progeria/genética , Progeria/patología , Precursores de Proteínas/metabolismo
2.
Nature ; 589(7843): 608-614, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33408413

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS or progeria) is typically caused by a dominant-negative C•G-to-T•A mutation (c.1824 C>T; p.G608G) in LMNA, the gene that encodes nuclear lamin A. This mutation causes RNA mis-splicing that produces progerin, a toxic protein that induces rapid ageing and shortens the lifespan of children with progeria to approximately 14 years1-4. Adenine base editors (ABEs) convert targeted A•T base pairs to G•C base pairs with minimal by-products and without requiring double-strand DNA breaks or donor DNA templates5,6. Here we describe the use of an ABE to directly correct the pathogenic HGPS mutation in cultured fibroblasts derived from children with progeria and in a mouse model of HGPS. Lentiviral delivery of the ABE to fibroblasts from children with HGPS resulted in 87-91% correction of the pathogenic allele, mitigation of RNA mis-splicing, reduced levels of progerin and correction of nuclear abnormalities. Unbiased off-target DNA and RNA editing analysis did not detect off-target editing in treated patient-derived fibroblasts. In transgenic mice that are homozygous for the human LMNA c.1824 C>T allele, a single retro-orbital injection of adeno-associated virus 9 (AAV9) encoding the ABE resulted in substantial, durable correction of the pathogenic mutation (around 20-60% across various organs six months after injection), restoration of normal RNA splicing and reduction of progerin protein levels. In vivo base editing rescued the vascular pathology of the mice, preserving vascular smooth muscle cell counts and preventing adventitial fibrosis. A single injection of ABE-expressing AAV9 at postnatal day 14 improved vitality and greatly extended the median lifespan of the mice from 215 to 510 days. These findings demonstrate the potential of in vivo base editing as a possible treatment for HGPS and other genetic diseases by directly correcting their root cause.


Asunto(s)
Adenina/metabolismo , Edición Génica/métodos , Mutación , Progeria/genética , Progeria/terapia , Alelos , Empalme Alternativo , Animales , Aorta/patología , Emparejamiento Base , Niño , ADN/genética , Modelos Animales de Enfermedad , Femenino , Fibroblastos/metabolismo , Humanos , Lamina Tipo A/química , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Longevidad , Masculino , Ratones , Ratones Transgénicos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Progeria/patología , ARN/genética
3.
Circulation ; 147(23): 1734-1744, 2023 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-36919608

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) is an ultrarare, fatal, premature aging disease caused by a toxic protein called progerin. Circulating progerin has not been previously detected, precluding research using readily available biological samples. This study aimed to develop a plasma progerin assay to evaluate progerin's quantity, response to progerin-targeted therapy, and relationship to patient survival. METHODS: Biological samples were collected by The Progeria Research Foundation Cell and Tissue Bank from a non-HGPS cohort cross-sectionally and a HGPS cohort longitudinally. HGPS donations occurred at baseline and intermittently while treated with farnesylation inhibitors lonafarnib±pravastatin and zoledronate, within 3 sequential open-label clinical trials at Boston Children's Hospital totaling >10 years of treatment. An ultrasensitive single-molecule counting progerin immunoassay was developed with prespecified performance parameters. Intra- and interpatient group statistics were descriptive. The relationship between progerin and survival was assessed by using joint modeling with time-dependent slopes parameterization. RESULTS: The assay's dynamic detection range was 59 to 30 000 pg/mL (R2=0.9987). There was no lamin A cross-reactivity. Mean plasma progerin in non-HGPS participants (n=69; 39 male, 30 female; age, 0.2-71.3 years) was 351±251 pg/mL, and in drug-naive participants with HGPS (n=74; 37 female, 37 male; age, 2.1-17.5 years) was 33 261±12 346 pg/mL, reflecting a 95-fold increase in affected children (P<0.0001). Progerin levels did not differ by sex (P=0.99). Lonafarnib treatment resulted in an average per-visit progerin decrease from baseline of between 35% to 62% (all P<0.005); effects were not augmented by adding pravastatin and zoledronate. Progerin levels fell within 4 months of therapy and remained lower for up to 10 years. The magnitude of progerin decrease positively associated with patient survival (P<0.0001; ie, 15 000 pg/mL decrease yields a 63.9% decreased risk of death). For any given decrease in progerin, life expectancy incrementally increased with longer treatment duration. CONCLUSIONS: A sensitive, quantitative immunoassay for progerin was developed and used to demonstrate high progerin levels in HGPS plasma that decreased with lonafarnib therapy. The extent of improved survival was associated with both the magnitude of progerin decrease and duration at lower levels. Thus, plasma progerin is a biomarker for HGPS whose reduction enables short- and long-term assessment of progerin-targeted treatment efficacy. REGISTRATION: URL: https://www. CLINICALTRIALS: gov. Unique identifiers: NCT00879034 and NCT00916747.


Asunto(s)
Progeria , Niño , Humanos , Masculino , Femenino , Lactante , Preescolar , Adolescente , Adulto Joven , Adulto , Persona de Mediana Edad , Anciano , Progeria/diagnóstico , Progeria/tratamiento farmacológico , Progeria/metabolismo , Ácido Zoledrónico/uso terapéutico , Pravastatina/uso terapéutico , Piperidinas/uso terapéutico , Lamina Tipo A/metabolismo
4.
Pediatr Res ; 95(5): 1356-1362, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38191824

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) and progeroid laminopathies (PL) are extremely rare genetic diseases with extremely poor prognoses. This study aims to investigate the epidemiological and genotypic characteristics of patients with HGPS/PL in China. METHODS: Using a cross-sectional study design, general characteristics and genotypic data of 46 patients with HGPS/PL from 17 provinces in China were analyzed. RESULTS: Among the 46 patients with HGPS/PL, 20 patients are HGPS, and the rest are PL; the identified total prevalence of HGPS/PL is 1/23 million. Among 42 patients with gene reports, 3 carried compound heterozygous mutations in the ZMPSTE24 while the other 39 carried LMNA mutations. Among PL, LMNA c.1579 C > T homozygous mutation was the most common. The onset of classic genotype HGPS is skin sclerosis in the first month after birth. The primary clinical manifestations of PL patients include skin abnormalities, growth retardation, and joint stiffness. The median age of onset for PL was 12 (6,12) months. CONCLUSIONS: In China, the identified total prevalence of HGPS/PL is 1/23 million. 92.8% of the genetic mutations of HGPS/PL were located in LMNA, and the rest in ZMPSTE24. Most patients of HGPS/PL have skin abnormalities as the earliest manifestation. Compared to PL, the classic genotype HGPS starts earlier. IMPACT STATEMENT: Hutchinson-Gilford progeria syndrome (HGPS) and progeroid laminopathies (PL) are extremely rare genetic diseases with extremely poor prognoses. To date, there is a paucity of epidemiological data related to HGPS/PL in China. This study first examined the genotypic, phenotypic, and prevalence characteristics of 40-50% of the cases of HGPS/PL in mainland China through a collaborative international registry effort. In China, the identified total prevalence of HGPS/PL is 1/23 million. 92.8% of the genetic mutations of HGPS/PL are located in LMNA. LMNA c.1579 C > T homozygous mutations are the most common form of gene mutations among the Chinese PL population.


Asunto(s)
Lamina Tipo A , Proteínas de la Membrana , Mutación , Progeria , Humanos , Progeria/genética , Progeria/epidemiología , China/epidemiología , Masculino , Femenino , Lamina Tipo A/genética , Estudios Transversales , Preescolar , Lactante , Prevalencia , Niño , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Genotipo , Adolescente , Laminopatías/genética , Laminopatías/epidemiología , Fenotipo
5.
Phys Occup Ther Pediatr ; 43(4): 482-501, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36628480

RESUMEN

AIMS: Limited information is available on impairments, activity limitations and participation restrictions in youth with Hutchinson-Gilford progeria syndrome (HGPS), a rare genetic premature aging disease. The purposes were to: (1) describe range of motion (ROM), grip, pinch and quadriceps strength, functional balance, walking endurance, and gross motor limitations and participation restrictions; (2) evaluate the association between ROM impairments and age; and (3) evaluate the association between the Gross Motor Function Measure-88 (GMFM) scores and lower extremity (LE) ROM, quadriceps strength, and age. METHODS: Upper and LE ROM, grip, pinch and quadriceps strength, Timed Up and Go (TUG), Six Minute Walk Test, GMFM-88, and Canadian Occupational Performance Measure data were recorded for 38 participants with HGPS. RESULTS: All youth exhibited ROM impairments and most displayed decreased grip and pinch strength, walking endurance, and gross motor skills when compared to same-aged peers. However, the majority had good functional balance with TUG scores in the normal range. Participation restrictions included difficulty keeping up with peers when walking and difficulty completing activities of daily living. Some ROM measurements were negatively associated with age indicating that older participants had more extensive ROM limitation than younger participants. CONCLUSIONS: Physical and occupational therapists can use this information when evaluating youth with HGPS, designing a plan of care, and providing treatment interventions.


Asunto(s)
Progeria , Humanos , Adolescente , Progeria/genética , Actividades Cotidianas , Canadá , Caminata , Rango del Movimiento Articular
6.
Am J Med Genet A ; 188(1): 216-223, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34611991

RESUMEN

Pathogenic biallelic variants in POL3RA have been associated with different disorders characterized by progressive neurological deterioration. These include the 4H leukodystrophy syndrome (hypomyelination, hypogonadotropic hypogonadism, and hypodontia) and adolescent-onset progressive spastic ataxia, as well as Wiedemann-Rautenstrauch syndrome (WRS), a recognizable neonatal progeroid syndrome. The phenotypic differences between these disorders are thought to occur mainly due to different functional effects of underlying POLR3A variants. Here we present the detailed clinical course of a 37-year-old woman in whom we identified a homozygous synonymous POLR3A variant c.3336G>A resulting in leaky splicing r.[3336ins192, =, 3243_3336del94]. She presented at birth with intrauterine growth retardation, lipodystrophy, muscular hypotonia, and several WRS-like facial features, albeit without sparse hair and prominent scalp veins. She had no signs of developmental delay or intellectual disability. Over the years, above characteristic facial features, she showed severe postnatal growth retardation, global lipodystrophy, joint contractures, thoracic hypoplasia, scoliosis, anodontia, spastic quadriplegia, bilateral hearing loss, aphonia, hypogonadotropic hypogonadism, and cerebellar peduncles hyperintensities in brain imaging. These manifestations partially overlap the clinical features of the previously reported POLR3A-associated disorders, mostly mimicking the WRS. Thus, our study expands the POLR3A-mediated phenotypic spectrum and suggests existence of a phenotypic continuum underlying biallelic POLR3A variants.


Asunto(s)
Atrofia Óptica , Progeria , Ataxias Espinocerebelosas , Adolescente , Adulto , Ataxia , Femenino , Humanos , Recién Nacido , Progeria/patología , ARN Polimerasa III/genética
8.
Proc Natl Acad Sci U S A ; 115(16): 4206-4211, 2018 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-29581305

RESUMEN

LMNA encodes the A-type lamins that are part of the nuclear scaffold. Mutations in LMNA can cause a variety of disorders called laminopathies, including Hutchinson-Gilford progeria syndrome (HGPS), atypical Werner syndrome, and Emery-Dreifuss muscular dystrophy. Previous work has shown that treatment of HGPS cells with the mTOR inhibitor rapamycin or with the rapamycin analog everolimus corrects several of the phenotypes seen at the cellular level-at least in part by increasing autophagy and reducing the amount of progerin, the toxic form of lamin A that is overproduced in HGPS patients. Since other laminopathies also result in production of abnormal and potentially toxic lamin proteins, we hypothesized that everolimus would also be beneficial in those disorders. To test this, we applied everolimus to fibroblast cell lines from six laminopathy patients, each with a different mutation in LMNA Everolimus treatment increased proliferative ability and delayed senescence in all cell lines. In several cell lines, we observed that with treatment, there is a significant improvement in nuclear blebbing, which is a cellular hallmark of HGPS and other lamin disorders. These preclinical results suggest that everolimus might have clinical benefit for multiple laminopathy syndromes.


Asunto(s)
Everolimus/farmacología , Fibroblastos/efectos de los fármacos , Lamina Tipo A/deficiencia , Distrofia Muscular de Emery-Dreifuss/genética , Progeria/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Síndrome de Werner/genética , Biomarcadores , División Celular/efectos de los fármacos , Línea Celular , Núcleo Celular/efectos de los fármacos , Núcleo Celular/ultraestructura , Senescencia Celular/efectos de los fármacos , Humanos , Lamina Tipo A/genética , Distrofia Muscular de Emery-Dreifuss/patología , Mutación , Fosforilación/efectos de los fármacos , Progeria/patología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteína S6 Ribosómica/metabolismo , Síndrome de Werner/patología
10.
Proc Natl Acad Sci U S A ; 113(46): E7250-E7259, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27799555

RESUMEN

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.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Trastorno del Sistema de Conducción Cardíaco/fisiopatología , Progeria/fisiopatología , Adolescente , Adulto , Animales , Arritmias Cardíacas/metabolismo , Calcio/fisiología , Trastorno del Sistema de Conducción Cardíaco/metabolismo , Niño , Preescolar , Conexina 43/metabolismo , Conexina 43/fisiología , Femenino , Corazón/fisiología , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Metaloendopeptidasas/genética , Metaloendopeptidasas/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/metabolismo , Lámina Nuclear/fisiología , Progeria/metabolismo , Retículo Sarcoplasmático/fisiología , Adulto Joven
11.
Hum Genet ; 137(11-12): 921-939, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30450527

RESUMEN

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.


Asunto(s)
Retardo del Crecimiento Fetal/genética , Progeria/genética , Pirrolina Carboxilato Reductasas/genética , ARN Polimerasa III/genética , Adolescente , Empalme Alternativo/genética , Niño , Femenino , Retardo del Crecimiento Fetal/diagnóstico , Retardo del Crecimiento Fetal/patología , Predisposición Genética a la Enfermedad , Humanos , Lactante , Lamina Tipo A/genética , Masculino , Mutación , Fenotipo , Progeria/diagnóstico , Progeria/patología , Progeria/fisiopatología , delta-1-Pirrolina-5-Carboxilato Reductasa
12.
Pediatr Res ; 83(5): 982-992, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29342131

RESUMEN

BackgroundHutchinson-Gilford progeria syndrome (HGPS) is an ultra-rare, fatal, segmental premature aging syndrome caused by the aberrant lamin A protein, progerin. The protein farnesyltransferase inhibitor, lonafarnib, ameliorates some aspects of cardiovascular and bone disease.MethodsWe performed a prospective longitudinal survey of plasma proteins in 24 children with HGPS (an estimated 10% of the world's population at the time) at baseline and on lonafarnib therapy, compared with age- and gender-matched controls using a multi-analyte, microsphere-based immunofluorescent assay.ResultsThe mean levels for 23/66 (34.8%) proteins were significantly lower and 7/66 (10.6%) were significantly higher in HGPS samples compared with those in controls (P≤0.05). Six proteins whose concentrations were initially lower normalized with lonafarnib therapy: interleukins 1α, 7, and 13, beta-2 microglobulin, C-reactive protein, and myoglobin. Alpha-2 macroglobulin, a protease inhibitor associated with stroke, was elevated at baseline and subsequently normalized with lonafarnib therapy.ConclusionThis is the first study to employ a multi-analyte array platform in HGPS. Novel potential biomarkers identified in this study should be further validated by correlations with clinical disease status, especially proteins associated with cardiovascular disease and those that normalized with lonafarnib therapy.


Asunto(s)
Proteínas Sanguíneas/análisis , Piperidinas/uso terapéutico , Progeria/sangre , Progeria/tratamiento farmacológico , Piridinas/uso terapéutico , Adolescente , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Estudios de Casos y Controles , Niño , Preescolar , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Interleucina-13/sangre , Interleucina-1alfa/sangre , Interleucina-7/sangre , Lamina Tipo A , Estudios Longitudinales , Masculino , Mutación , Mioglobina/sangre , Piperidinas/sangre , Estudios Prospectivos , Piridinas/sangre , Microglobulina beta-2/sangre
13.
J Med Genet ; 54(3): 212-216, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27920058

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome (HGPS) is a fatal sporadic autosomal dominant premature ageing disease caused by single base mutations that optimise a cryptic splice site within exon 11 of the LMNA gene. The resultant disease-causing protein, progerin, acts as a dominant negative. Disease severity relies partly on progerin levels. METHODS AND RESULTS: We report a novel form of somatic mosaicism, where a child possessed two cell populations with different HGPS disease-producing mutations of the same nucleotide-one producing severe HGPS and one mild HGPS. The proband possessed an intermediate phenotype. The mosaicism was initially discovered when Sanger sequencing showed a c.1968+2T>A mutation in blood DNA and a c.1968+2T>C in DNA from cultured fibroblasts. Deep sequencing of DNA from the proband's blood revealed 4.7% c.1968+2T>C mutation, and 41.3% c.1968+2T>A mutation. CONCLUSIONS: We hypothesise that the germline mutation was c.1968+2T>A, but a rescue event occurred during early development, where the somatic mutation from A to C at 1968+2 provided a selective advantage. This type of mosaicism where a partial phenotypic rescue event results from a second but milder disease-causing mutation in the same nucleotide has not been previously characterised for any disease.


Asunto(s)
Núcleo Celular/genética , Lamina Tipo A/genética , Progeria/genética , Adolescente , Núcleo Celular/patología , Células Cultivadas , Niño , Preescolar , Exones/genética , Femenino , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Masculino , Mosaicismo , Progeria/patología
14.
JAMA ; 319(16): 1687-1695, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29710166

RESUMEN

Importance: Hutchinson-Gilford progeria syndrome (HGPS) is an extremely rare fatal premature aging disease. There is no approved treatment. Objective: To evaluate the association of monotherapy using the protein farnesyltransferase inhibitor lonafarnib with mortality rate in children with HGPS. Design, Setting, and Participants: Cohort study comparing contemporaneous (birth date ≥1991) untreated patients with HGPS matched with treated patients by age, sex, and continent of residency using conditional Cox proportional hazards regression. Treatment cohorts included patients from 2 single-group, single-site clinical trials (ProLon1 [n = 27; completed] and ProLon2 [n = 36; ongoing]). Untreated patients originated from a separate natural history study (n = 103). The cutoff date for patient follow-up was January 1, 2018. Exposure: Treated patients received oral lonafarnib (150 mg/m2) twice daily. Untreated patients received no clinical trial medications. Main Outcomes and Measures: The primary outcome was mortality. The primary analysis compared treated patients from the first lonafarnib trial with matched untreated patients. A secondary analysis compared the combined cohorts from both lonafarnib trials with matched untreated patients. Results: Among untreated and treated patients (n = 258) from 6 continents, 123 (47.7%) were female; 141 (54.7%) had a known genotype, of which 125 (88.7%) were classic (c.1824C>T in LMNA). When identified (n = 73), the primary cause of death was heart failure (79.4%). The median treatment duration was 2.2 years. Median age at start of follow-up was 8.4 (interquartile range [IQR], 4.8-9.5) years in the first trial cohort and 6.5 (IQR, 3.7-9.0) years in the combined cohort. There was 1 death (3.7%) among 27 patients in the first trial group and there were 9 deaths (33.3%) among 27 patients in the matched untreated group. Treatment was associated with a lower mortality rate (hazard ratio, 0.12; 95% CI, 0.01-0.93; P = .04). In the combined cohort, there were 4 deaths (6.3%) among 63 patients in the treated group and 17 deaths (27.0%) among 63 patients in the matched untreated group (hazard ratio, 0.23; 95% CI, 0.06-0.90; P = .04). Conclusions and Relevance: Among patients with HGPS, lonafarnib monotherapy, compared with no treatment, was associated with a lower mortality rate after 2.2 years of follow-up. Study interpretation is limited by its observational design.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Fosfotransferasas (Aceptor del Grupo Fosfato)/antagonistas & inhibidores , Piperidinas/uso terapéutico , Progeria/tratamiento farmacológico , Piridinas/uso terapéutico , Adolescente , Adulto , Causas de Muerte , Niño , Estudios de Cohortes , Femenino , Humanos , Estimación de Kaplan-Meier , Lamina Tipo A/biosíntesis , Lamina Tipo A/metabolismo , Masculino , Progeria/genética , Progeria/mortalidad , Procesamiento Proteico-Postraduccional , Adulto Joven
15.
Circulation ; 134(2): 114-25, 2016 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-27400896

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome is an extremely rare, fatal, segmental premature aging syndrome caused by a mutation in LMNA yielding the farnesylated aberrant protein progerin. Without progerin-specific treatment, death occurs at an average age of 14.6 years from an accelerated atherosclerosis. A previous single-arm clinical trial demonstrated that the protein farnesyltransferase inhibitor lonafarnib ameliorates some aspects of cardiovascular and bone disease. This present trial sought to further improve disease by additionally inhibiting progerin prenylation. METHODS: Thirty-seven participants with Hutchinson-Gilford progeria syndrome received pravastatin, zoledronic acid, and lonafarnib. This combination therapy was evaluated, in addition to descriptive comparisons with the prior lonafarnib monotherapy trial. RESULTS: No participants withdrew because of side effects. Primary outcome success was predefined by improved per-patient rate of weight gain or carotid artery echodensity; 71.0% of participants succeeded (P<0.0001). Key cardiovascular and skeletal secondary variables were predefined. Secondary improvements included increased areal (P=0.001) and volumetric (P<0.001-0.006) bone mineral density and 1.5- to 1.8-fold increases in radial bone structure (P<0.001). Median carotid artery wall echodensity and carotid-femoral pulse wave velocity demonstrated no significant changes. Percentages of participants with carotid (5% to 50%; P=0.001) and femoral (0% to 12%; P=0.13) artery plaques and extraskeletal calcifications (34.4% to 65.6%; P=0.006) increased. Other than increased bone mineral density, no improvement rates exceeded those of the prior lonafarnib monotherapy treatment trial. CONCLUSIONS: Comparisons with lonafarnib monotherapy treatment reveal additional bone mineral density benefit but likely no added cardiovascular benefit with the addition of pravastatin and zoledronic acid. CLINICAL TRIAL REGISTRATION: URL: http://www.clinicaltrials.gov. Unique identifiers: NCT00879034 and NCT00916747.


Asunto(s)
Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Piperidinas/uso terapéutico , Pravastatina/uso terapéutico , Progeria/tratamiento farmacológico , Piridinas/uso terapéutico , Huesos/diagnóstico por imagen , Arterias Carótidas/diagnóstico por imagen , Preescolar , Difosfonatos/efectos adversos , Quimioterapia Combinada , Femenino , Humanos , Imidazoles/efectos adversos , Lactante , Masculino , Piperidinas/efectos adversos , Piperidinas/farmacocinética , Pravastatina/efectos adversos , Estudios Prospectivos , Prenilación de Proteína/efectos de los fármacos , Piridinas/efectos adversos , Piridinas/farmacocinética , Ácido Zoledrónico
16.
Circulation ; 130(1): 27-34, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24795390

RESUMEN

BACKGROUND: Hutchinson-Gilford progeria syndrome is an ultrarare segmental premature aging disease resulting in early death from heart attack or stroke. There is no approved treatment, but starting in 2007, several recent single-arm clinical trials administered inhibitors of protein farnesylation aimed at reducing toxicity of the disease-producing protein progerin. No study assessed whether treatments influence patient survival. The key elements necessary for this analysis are a robust natural history of survival and comparison with a sufficiently large patient population that has been treated for a sufficient time period with disease-targeting medications. METHODS AND RESULTS: We generated Kaplan-Meier survival analyses for the largest untreated Hutchinson-Gilford progeria syndrome cohort to date. Mean survival was 14.6 years. Comparing survival for treated versus age- and sex-matched untreated cohorts, hazard ratio was 0.13 (95% confidence interval, 0.04-0.37; P<0.001) with median follow-up of 5.3 years from time of treatment initiation. There were 21 of 43 deaths in untreated versus 5 of 43 deaths among treated subjects. Treatment increased mean survival by 1.6 years. CONCLUSIONS: This study provides a robust untreated disease survival profile that can be used for comparisons now and in the future to assess changes in survival with treatments for Hutchinson-Gilford progeria syndrome. The current comparisons estimating increased survival with protein farnesylation inhibitors provide the first evidence of treatments influencing survival for this fatal disease. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrials.gov. Unique Indentifiers: NCT00425607, NCT00879034, and NCT00916747.


Asunto(s)
Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Proteínas Nucleares/metabolismo , Piperidinas/uso terapéutico , Pravastatina/uso terapéutico , Progeria/tratamiento farmacológico , Precursores de Proteínas/metabolismo , Prenilación de Proteína/efectos de los fármacos , Piridinas/uso terapéutico , Adolescente , Adulto , Transferasas Alquil y Aril/antagonistas & inhibidores , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/prevención & control , Causas de Muerte , Niño , Preescolar , Ensayos Clínicos como Asunto/estadística & datos numéricos , Estudios de Cohortes , Dimetilaliltranstransferasa/antagonistas & inhibidores , Difosfonatos/administración & dosificación , Difosfonatos/farmacología , Quimioterapia Combinada , Femenino , Genes Dominantes , Genotipo , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/administración & dosificación , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/uso terapéutico , Imidazoles/administración & dosificación , Imidazoles/farmacología , Estimación de Kaplan-Meier , Lamina Tipo A , Masculino , Estudios Multicéntricos como Asunto/estadística & datos numéricos , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Piperidinas/administración & dosificación , Piperidinas/farmacología , Pravastatina/administración & dosificación , Pravastatina/farmacología , Progeria/complicaciones , Progeria/mortalidad , Modelos de Riesgos Proporcionales , Precursores de Proteínas/deficiencia , Precursores de Proteínas/genética , Piridinas/administración & dosificación , Piridinas/farmacología , Resultado del Tratamiento , Adulto Joven , Ácido Zoledrónico
17.
Proc Natl Acad Sci U S A ; 109(41): 16666-71, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23012407

RESUMEN

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.


Asunto(s)
Inhibidores Enzimáticos/uso terapéutico , Farnesiltransferasa/antagonistas & inhibidores , Piperidinas/uso terapéutico , Progeria/tratamiento farmacológico , Piridinas/uso terapéutico , Adolescente , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/patología , Niño , Preescolar , Diarrea/inducido químicamente , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Inhibidores Enzimáticos/efectos adversos , Inhibidores Enzimáticos/farmacocinética , Farnesiltransferasa/metabolismo , Fatiga/inducido químicamente , Femenino , Humanos , Masculino , Piperidinas/efectos adversos , Piperidinas/farmacocinética , Progeria/patología , Progeria/fisiopatología , Análisis de la Onda del Pulso , Piridinas/efectos adversos , Piridinas/farmacocinética , Resultado del Tratamiento , Vómitos/inducido químicamente , Aumento de Peso/efectos de los fármacos
18.
Pediatr Dermatol ; 31(2): 196-202, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24456199

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare, uniformly fatal, premature aging disease with distinct dermatologic features. We sought to identify and describe the initial skin and hair findings as potential diagnostic signs of the disease. We performed a chart review of the structured initial intake histories of 39 individuals with HGPS enrolled in clinical trials from 2007 to 2010 at Boston Children's Hospital, limited to cutaneous history from birth to 24 months. Medical photographs were provided through the clinical trials and the Progeria Research Foundation Medical and Research Database at Brown University Center for Gerontology and Healthcare Research. All 39 patients reported skin and hair abnormalities within the first 24 months of life. Pathologies included sclerodermoid change, prominent superficial veins, dyspigmentation, and alopecia. The mean age of presentation for each finding was <12 months. The most frequently reported skin feature was sclerodermoid change, which commonly involved the abdomen and bilateral lower extremities. Prominent superficial vasculature manifested as circumoral cyanosis and pronounced veins on the scalp and body. Hypo- and hyperpigmentation were observed over areas of sclerodermoid change. Scalp alopecia progressed in a distinct pattern, with preservation of the hair over the midscalp and vertex areas for the longest period of time. HGPS has distinct cutaneous manifestations during the first 2 years of life that may be the first signs of disease. Awareness of these findings could expedite diagnosis.


Asunto(s)
Progeria/patología , Piel/patología , Boston , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Humanos , Lactante , Masculino
19.
Front Cardiovasc Med ; 11: 1356010, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38725831

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is an ultra-rare genetic premature aging disease that is historically fatal in teenage years, secondary to severe accelerated atherosclerosis. The only approved treatment is the farnesyltransferase inhibitor lonafarnib, which improves vascular structure and function, extending average untreated lifespan of 14.5 years by 4.3 years (30%). With this longer lifespan, calcific aortic stenosis (AS) was identified as an emerging critical risk factor for cardiac death in older patients. Intervention to relieve critical AS has the potential for immediate improvement in healthspan and lifespan. However, HGPS patient-device size mismatch, pervasive peripheral arterial disease, skin and bone abnormalities, and lifelong failure to thrive present unique challenges to intervention. An international group of experts in HGPS, pediatric and adult cardiology, cardiac surgery, and pediatric critical care convened to identify strategies for successful treatment. Candidate procedures were evaluated by in-depth examination of 4 cases that typify HGPS clinical pathology. Modified transcatheter aortic valve replacement (TAVR) and left ventricular Apico-Aortic Conduit (AAC) placement were deemed high risk but viable options. Two cases received TAVR and 2 received AAC post-summit. Three were successful and 1 patient died perioperatively due to cardiovascular disease severity, highlighting the importance of intervention timing and comparative risk stratification. These breakthrough interventions for treating critical aortic stenosis in HGPS patients could rewrite the current clinical perspective on disease course by greatly improving late-stage quality of life and increasing lifespan. Expanding worldwide medical and surgical competency for this ultra-rare disease through expert information-sharing could have high impact on treatment success.

20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA