Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
Foot Ankle Surg ; 30(3): 258-262, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38185597

RESUMO

BACKGROUND: Minimally invasive surgical (MIS) osteotomies are increasing as a surgical option for treating midfoot and forefoot conditions. This study aimed to evaluate the impact of each burr pass on the degree of correction, gap size, and alignment in MIS Akin and first metatarsal dorsiflexion osteotomies (DFO). METHODS: MIS Akin and first metatarsal DFO were performed on ten cadaveric specimens. Fluoroscopic measurements included the metatarsal dorsiflexion angle (MDA), dorsal cortical length (MDCL), first phalangeal medial cortical length (PCML) and proximal to distal phalangeal articular angle (PDPAA). RESULTS: The average decrease in PCML with each burr pass was as follows: 1.53, 1.33, 1.27, 1.23 and 1.13 mm at the 1st to 5th pass, respectively. The MDCL sequentially decreased by 1.80, 1.59, 1.35, 0.75, and 0.60 mm. The MDA consistently decreased, and the PDPAA incrementally became more valgus oriented. CONCLUSION: On average, a first metatarsal dorsal wedge resection of 4.7 mm and first phalangeal medial wedge resection of 2.9 mm was achieved after 3 and 2 burr passes, respectively. This data may aid surgeons determine the optimal number of burr passes required to achieve the desired patient-specific surgical correction.


Assuntos
Hallux Valgus , Lamina Tipo A/deficiência , Ossos do Metatarso , Distrofias Musculares , Humanos , Ossos do Metatarso/diagnóstico por imagem , Ossos do Metatarso/cirurgia , Hallux Valgus/cirurgia , Osteotomia , , Resultado do Tratamento
2.
J Biochem ; 171(5): 509-518, 2022 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-35137145

RESUMO

Nuclear lamina is a fundamental structure of the cell nucleus and regulates a wide range of molecular pathways. Defects of components of the nuclear lamina cause ageing-like physiological disorders, called laminopathy. Generally, ageing and diseases are often associated with perturbation of various time-of-day-dependent regulations, but it remains elusive whether laminopathy induces any changes of the circadian clock and physiological rhythms. Here, we demonstrated that deficiency of Lmna gene in mice caused an obvious shift of locomotor activities to the daytime. The abnormal activity profile was accompanied by a remarkable change in phase angle between the central clock in the suprachiasmatic nucleus (SCN) and the lung peripheral clocks, leaving the phase of the SCN clock unaffected by the mutation. These observations suggest that Lmna deficiency causes a change of the habitat from nocturnal to diurnal behaviours. On the other hand, molecular oscillation and its phase resetting mechanism were intact in both the Lmna-deficient cells and progeria-mimicking cells. Intriguingly, high-fat diet feeding extended the short lifespan and ameliorated the abnormalities of the behaviours and the phase of the peripheral clock in the Lmna-deficient mice. The present study supports the important contribution of the energy conditions to a shift between the diurnal and nocturnal activities.


Assuntos
Relógios Circadianos , Lamina Tipo A , Laminopatias , Envelhecimento/genética , Animais , Relógios Circadianos/genética , Ritmo Circadiano/genética , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Camundongos , Núcleo Supraquiasmático/metabolismo
3.
Int. j. cardiovasc. sci. (Impr.) ; 35(1): 136-139, Jan.-Feb. 2022. graf
Artigo em Inglês | LILACS | ID: biblio-1356313

RESUMO

Abstract Isolated left ventricular apical hypoplasia is a rare cardiomyopathy, with a broad range of clinical presentations. Since this entity was already described in association with osteomuscular diseases, mutation in the Lamin A/C gene has been regarded as a possible cause of this disease. This study describes the case of an asymptomatic teenager with isolated left ventricular apical hypoplasia and arthrogriposis but with no mutations in the entire Lamin A/C gene.


Assuntos
Humanos , Masculino , Adolescente , Lamina Tipo A/genética , Miocárdio Ventricular não Compactado Isolado/fisiopatologia , Artrogripose , Lamina Tipo A/deficiência , Miocárdio Ventricular não Compactado Isolado/diagnóstico , Miocárdio Ventricular não Compactado Isolado/etiologia
4.
Nat Commun ; 12(1): 4722, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34354059

RESUMO

Mutations in the LaminA gene are a common cause of monogenic dilated cardiomyopathy. Here we show that mice with a cardiomyocyte-specific Lmna deletion develop cardiac failure and die within 3-4 weeks after inducing the mutation. When the same Lmna mutations are induced in mice genetically deficient in the LINC complex protein SUN1, life is extended to more than one year. Disruption of SUN1's function is also accomplished by transducing and expressing a dominant-negative SUN1 miniprotein in Lmna deficient cardiomyocytes, using the cardiotrophic Adeno Associated Viral Vector 9. The SUN1 miniprotein disrupts binding between the endogenous LINC complex SUN and KASH domains, displacing the cardiomyocyte KASH complexes from the nuclear periphery, resulting in at least a fivefold extension in lifespan. Cardiomyocyte-specific expression of the SUN1 miniprotein prevents cardiomyopathy progression, potentially avoiding the necessity of developing a specific therapeutic tailored to treating each different LMNA cardiomyopathy-inducing mutation of which there are more than 450.


Assuntos
Cardiomiopatia Dilatada/genética , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Animais , Cardiomiopatia Dilatada/patologia , Cardiomiopatia Dilatada/fisiopatologia , Dependovirus/genética , Feminino , Humanos , Lamina Tipo A/deficiência , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/deficiência , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Transdução Genética
5.
Sci Rep ; 11(1): 13195, 2021 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162976

RESUMO

Chromatin organization within the nuclear volume is essential to regulate many aspects of its function and to safeguard its integrity. A key player in this spatial scattering of chromosomes is the nuclear envelope (NE). The NE tethers large chromatin domains through interaction with the nuclear lamina and other associated proteins. This organization is perturbed in cells from Hutchinson-Gilford progeria syndrome (HGPS), a genetic disorder characterized by premature aging features. Here, we show that HGPS-related lamina defects trigger an altered 3D telomere organization with increased contact sites between telomeres and the nuclear lamina, and an altered telomeric chromatin state. The genome-wide replication timing signature of these cells is perturbed, with a shift to earlier replication for regions that normally replicate late. As a consequence, we detected a higher density of replication forks traveling simultaneously on DNA fibers, which relies on limiting cellular dNTP pools to support processive DNA synthesis. Remarkably, increasing dNTP levels in HGPS cells rescued fragile telomeres, and improved the replicative capacity of the cells. Our work highlights a functional connection between NE dysfunction and telomere homeostasis in the context of premature aging.


Assuntos
Cromatina/ultraestrutura , Desoxirribonucleotídeos/metabolismo , Lamina Tipo A/fisiologia , Lâmina Nuclear/patologia , Progéria/genética , Homeostase do Telômero/genética , Telômero/patologia , Adulto , Animais , Células Cultivadas , Senescência Celular/genética , Dano ao DNA , Replicação do DNA , Fibroblastos , Genes Reporter , Proteínas de Fluorescência Verde , Código das Histonas , Humanos , Recém-Nascido , Lamina Tipo A/análise , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Lamina Tipo B/análise , Camundongos , Camundongos Knockout , Progéria/patologia , Proteínas Recombinantes de Fusão/metabolismo , Pele/patologia
6.
Proc Natl Acad Sci U S A ; 118(25)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34161290

RESUMO

Defects or deficiencies in nuclear lamins cause pathology in many cell types, and recent studies have implicated nuclear membrane (NM) ruptures as a cause of cell toxicity. We previously observed NM ruptures and progressive cell death in the developing brain of lamin B1-deficient mouse embryos. We also observed frequent NM ruptures and DNA damage in nuclear lamin-deficient fibroblasts. Factors modulating susceptibility to NM ruptures remain unclear, but we noted low levels of LAP2ß, a chromatin-binding inner NM protein, in fibroblasts with NM ruptures. Here, we explored the apparent link between LAP2ß and NM ruptures in nuclear lamin-deficient neurons and fibroblasts, and we tested whether manipulating LAP2ß expression levels would alter NM rupture frequency. In cortical plate neurons of lamin B1-deficient embryos, we observed a strong correlation between low LAP2ß levels and NM ruptures. We also found low LAP2ß levels and frequent NM ruptures in neurons of cultured Lmnb1-/- neurospheres. Reducing LAP2ß expression in Lmnb1-/- neurons with an siRNA markedly increased the NM rupture frequency (without affecting NM rupture duration), whereas increased LAP2ß expression eliminated NM ruptures and reduced DNA damage. Consistent findings were observed in nuclear lamin-deficient fibroblasts. Reduced LAP2ß expression increased NM ruptures, whereas increased LAP2ß expression virtually abolished NM ruptures. Increased LAP2ß expression nearly abolished NM ruptures in cells subjected to mechanical stress (an intervention that increases NM ruptures). Our studies showed that increasing LAP2ß expression bolsters NM integrity in nuclear lamin-deficient cells and markedly reduces NM rupture frequency.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Fibroblastos/metabolismo , Lamina Tipo B/deficiência , Proteínas de Membrana/metabolismo , Neurônios/metabolismo , Membrana Nuclear/metabolismo , Animais , Morte Celular , Diferenciação Celular , Córtex Cerebral/patologia , Dano ao DNA , Embrião de Mamíferos/metabolismo , Lamina Tipo A/deficiência , Lamina Tipo A/metabolismo , Lamina Tipo B/metabolismo , Camundongos Knockout , Especificidade de Órgãos
7.
Biomolecules ; 11(4)2021 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-33917623

RESUMO

The Cdkn2a locus is one of the most studied tumor suppressor loci in the context of several cancer types. However, in the last years, its expression has also been linked to terminal differentiation and the activation of the senescence program in different cellular subtypes. Knock-out (KO) of the entire locus enhances the capability of stem cells to proliferate in some tissues and respond to severe physiological and non-physiological damages in different organs, including the heart. Emery-Dreifuss muscular dystrophy (EDMD) is characterized by severe contractures and muscle loss at the level of skeletal muscles of the elbows, ankles and neck, and by dilated cardiomyopathy. We have recently demonstrated, using the LMNA Δ8-11 murine model of Emery-Dreifuss muscular dystrophy (EDMD), that dystrophic muscle stem cells prematurely express non-lineage-specific genes early on during postnatal growth, leading to rapid exhaustion of the muscle stem cell pool. Knock-out of the Cdkn2a locus in EDMD dystrophic mice partially restores muscle stem cell properties. In the present study, we describe the cardiac phenotype of the LMNA Δ8-11 mouse model and functionally characterize the effects of KO of the Cdkn2a locus on heart functions and life expectancy.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Distrofia Muscular de Emery-Dreifuss/patologia , Animais , Apoptose , Inibidor p16 de Quinase Dependente de Ciclina/deficiência , Modelos Animais de Doenças , Loci Gênicos , Genótipo , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Longevidade , Camundongos , Camundongos Knockout , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/mortalidade , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Fenótipo , Células-Tronco/citologia , Células-Tronco/metabolismo , Taxa de Sobrevida
8.
Int J Mol Sci ; 21(22)2020 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-33233602

RESUMO

Lipodystrophies are a heterogeneous group of physiological changes characterized by a selective loss of fatty tissue. Here, no fat cells are present, either through lack of differentiation, loss of function or premature apoptosis. As a consequence, lipids can only be stored ectopically in non-adipocytes with the major health consequences as fatty liver and insulin resistance. This is a crucial difference to being slim where the fat cells are present and store lipids if needed. A simple clinical classification of lipodystrophies is based on congenital vs. acquired and generalized vs. partial disturbance of fat distribution. Complications in patients with lipodystrophy depend on the clinical manifestations. For example, in diabetes mellitus microangiopathic complications such as nephropathy, retinopathy and neuropathy may develop. In addition, due to ectopic lipid accumulation in the liver, fatty liver hepatitis may also develop, possibly with cirrhosis. The consequences of extreme hypertriglyceridemia are typically acute pancreatitis or eruptive xanthomas. The combination of severe hyperglycemia with dyslipidemia and signs of insulin resistance can lead to premature atherosclerosis with its associated complications of coronary heart disease, peripheral vascular disease and cerebrovascular changes. Overall, lipodystrophy is rare with an estimated incidence for congenital (<1/1.000.000) and acquired (1-9/100.000) forms. Due to the rarity of the syndrome and the phenotypic range of metabolic complications, only studies with limited patient numbers can be considered. Experimental animal models are therefore useful to understand the molecular mechanisms in lipodystrophy and to identify possible therapeutic approaches.


Assuntos
Aterosclerose/genética , Doença das Coronárias/genética , Diabetes Mellitus/genética , Fígado Gorduroso/genética , Hipertrigliceridemia/genética , Lipodistrofia/genética , Aciltransferases/deficiência , Aciltransferases/genética , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Aterosclerose/etiologia , Aterosclerose/metabolismo , Aterosclerose/patologia , Distribuição da Gordura Corporal , Doença das Coronárias/etiologia , Doença das Coronárias/metabolismo , Doença das Coronárias/patologia , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Diabetes Mellitus/patologia , Modelos Animais de Doenças , Fígado Gorduroso/complicações , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Humanos , Hipertrigliceridemia/complicações , Hipertrigliceridemia/metabolismo , Hipertrigliceridemia/patologia , Resistência à Insulina , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Metabolismo dos Lipídeos/genética , Lipodistrofia/complicações , Lipodistrofia/metabolismo , Lipodistrofia/patologia , Pancreatite/etiologia , Pancreatite/genética , Pancreatite/metabolismo , Pancreatite/patologia , Xantomatose/etiologia , Xantomatose/genética , Xantomatose/metabolismo , Xantomatose/patologia
9.
Mol Biol Cell ; 31(21): 2363-2378, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32816614

RESUMO

Nuclear envelope proteins influence cell cytoarchitecure by poorly understood mechanisms. Here we show that small interfering RNA-mediated silencing of lamin A/C (LMNA) promotes contrasting stress fiber assembly and disassembly in individual cells and within cell populations. We show that LMNA-deficient cells have elevated myosin-II bipolar filament accumulations, irregular formation of actin comet tails and podosome-like adhesions, increased steady state nuclear localization of the mechanosensitive transcription factors MKL1 and YAP, and induced expression of some MKL1/serum response factor-regulated genes such as that encoding myosin-IIA (MYH9). Our studies utilizing live cell imaging and pharmacological inhibition of myosin-II support a mechanism of deregulated myosin-II self-organizing activity at the nexus of divergent actin cytoskeletal aberrations resulting from LMNA loss. In light of our results, we propose a model of how the nucleus, via linkage to the cytoplasmic actomyosin network, may act to control myosin-II contractile behavior through both mechanical and transcriptional feedback mechanisms.


Assuntos
Citoesqueleto de Actina/metabolismo , Lamina Tipo A/metabolismo , Miosina Tipo II/metabolismo , Membrana Nuclear/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HeLa , Humanos , Lamina Tipo A/deficiência , Miosina Tipo II/genética
10.
Cells ; 9(6)2020 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-32517247

RESUMO

Reactive Oxygen Species (ROS) are reactive molecules required for the maintenance of physiological functions. Oxidative stress arises when ROS production exceeds the cellular ability to eliminate such molecules. In this study, we showed that oxidative stress induces post-translational modification of the inner nuclear membrane protein emerin. In particular, emerin is phosphorylated at the early stages of the oxidative stress response, while protein phosphorylation is abolished upon recovery from stress. A finely tuned balance between emerin phosphorylation and O-GlcNAcylation seems to govern this dynamic and modulates emerin-BAF interaction and BAF nucleoplasmic localization during the oxidative stress response. Interestingly, emerin post-translational modifications, similar to those observed during the stress response, are detected in cells bearing LMNA gene mutations and are characterized by a free radical generating environment. On the other hand, under oxidative stress conditions, a delay in DNA damage repair and cell cycle progression is found in cells from Emery-Dreifuss Muscular Dystrophy type 1, which do not express emerin. These results suggest a role of the emerin-BAF protein platform in the DNA damage response aimed at counteracting the detrimental effects of elevated levels of ROS.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Estresse Oxidativo , Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Células HEK293 , Células HeLa , Humanos , Lamina Tipo A/deficiência , Lamina Tipo A/metabolismo , Peso Molecular , Distrofia Muscular de Emery-Dreifuss/patologia , Fosforilação , Ligação Proteica , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo
11.
PLoS Biol ; 18(6): e3000731, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32479501

RESUMO

The nuclear lamina protein lamin A/C is a key component of the nuclear envelope. Mutations in the lamin A/C gene (LMNA) are identified in patients with various types of laminopathy-containing diseases, which have features of accelerated aging and osteoporosis. However, the underlying mechanisms for laminopathy-associated osteoporosis remain largely unclear. Here, we provide evidence that loss of lamin A/C in skeletal muscles, but not osteoblast (OB)-lineage cells, results in not only muscle aging-like deficit but also trabecular bone loss, a feature of osteoporosis. The latter is due in large part to elevated bone resorption. Further cellular studies show an increase of osteoclast (OC) differentiation in cocultures of bone marrow macrophages/monocytes (BMMs) and OBs after treatment with the conditioned medium (CM) from lamin A/C-deficient muscle cells. Antibody array screening analysis of the CM proteins identifies interleukin (IL)-6, whose expression is markedly increased in lamin A/C-deficient muscles. Inhibition of IL-6 by its blocking antibody in BMM-OB cocultures diminishes the increase of osteoclastogenesis. Knockout (KO) of IL-6 in muscle lamin A/C-KO mice diminishes the deficits in trabecular bone mass but not muscle. Further mechanistic studies reveal an elevation of cellular senescence marked by senescence-associated beta-galactosidase (SA-ß-gal), p16Ink4a, and p53 in lamin A/C-deficient muscles and C2C12 muscle cells, and the p16Ink4a may induce senescence-associated secretory phenotype (SASP) and IL-6 expression. Taken together, these results suggest a critical role for skeletal muscle lamin A/C to prevent cellular senescence, IL-6 expression, hyperosteoclastogenesis, and trabecular bone loss, uncovering a pathological mechanism underlying the link between muscle aging/senescence and osteoporosis.


Assuntos
Envelhecimento/patologia , Lamina Tipo A/deficiência , Músculo Esquelético/patologia , Osteoporose/patologia , Animais , Anticorpos Bloqueadores/farmacologia , Fenômenos Biomecânicos , Reabsorção Óssea/complicações , Reabsorção Óssea/patologia , Osso Esponjoso/efeitos dos fármacos , Osso Esponjoso/patologia , Diferenciação Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Interleucina-6/metabolismo , Camundongos Knockout , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Osteoporose/sangue , Fenótipo
12.
J Clin Invest ; 130(9): 4740-4758, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32484798

RESUMO

Mutation in the LMNA gene, encoding lamin A/C, causes a diverse group of diseases called laminopathies. Cardiac involvement is the major cause of death and manifests as dilated cardiomyopathy, heart failure, arrhythmias, and sudden death. There is no specific therapy for LMNA-associated cardiomyopathy. We report that deletion of Lmna in cardiomyocytes in mice leads to severe cardiac dysfunction, conduction defect, ventricular arrhythmias, fibrosis, apoptosis, and premature death within 4 weeks. The phenotype is similar to LMNA-associated cardiomyopathy in humans. RNA sequencing, performed before the onset of cardiac dysfunction, led to identification of 2338 differentially expressed genes (DEGs) in Lmna-deleted cardiomyocytes. DEGs predicted activation of bromodomain-containing protein 4 (BRD4), a regulator of chromatin-associated proteins and transcription factors, which was confirmed by complementary approaches, including chromatin immunoprecipitation sequencing. Daily injection of JQ1, a specific BET bromodomain inhibitor, partially reversed the DEGs, including those encoding secretome; improved cardiac function; abrogated cardiac arrhythmias, fibrosis, and apoptosis; and prolonged the median survival time 2-fold in the myocyte-specific Lmna-deleted mice. The findings highlight the important role of LMNA in cardiomyocytes and identify BET bromodomain inhibition as a potential therapeutic target in LMNA-associated cardiomyopathy, for which there is no specific effective therapy.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Regulação da Expressão Gênica , Lamina Tipo A/deficiência , Miócitos Cardíacos/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Animais , Azepinas/farmacologia , Cardiomiopatia Dilatada/tratamento farmacológico , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/patologia , Lamina Tipo A/metabolismo , Camundongos , Camundongos Knockout , Miócitos Cardíacos/patologia , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Triazóis/farmacologia
13.
Sci Rep ; 10(1): 6632, 2020 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-32313136

RESUMO

Lamin A/C (LMNA) gene mutations are a known cause of familial dilated cardiomyopathy, but the precise mechanisms triggering disease progression remain unknown. We hypothesize that analysis of differentially expressed genes (DEGs) throughout the course of Lmna knockout (Lmna-/-)-induced cardiomyopathy may reveal novel Lmna-mediated alterations of signaling pathways leading to dilated cardiomyopathy. Although Lmna was the only DEG down-regulated at 1 week of age, we identified 730 and 1004 DEGs in Lmna-/- mice at 2 weeks and 1 month of age, respectively. At 2 weeks, Lmna-/- mice demonstrated both down- and up-regulation of the key genes involving cell cycle control, mitochondrial dysfunction, and oxidative phosphorylation, as well as down-regulated genes governing DNA damage repair and up-regulated genes involved in oxidative stress response, cell survival, and cardiac hypertrophy. At 1 month, the down-regulated genes included those involved in oxidative phosphorylation, mitochondrial dysfunction, nutrient metabolism, cardiac ß-adrenergic signaling, action potential generation, and cell survival. We also found 96 overlapping DEGs at both ages involved in oxidative phosphorylation, mitochondrial function, and calcium signaling. Impaired oxidative phosphorylation was observed at early disease stage, even before the appearance of disease phenotypes, and worsened with disease progression, suggesting its importance in the pathogenesis and progression of LMNA cardiomyopathy. Reduction of oxidative stress might therefore prevent or delay the development from Lmna mutation to LMNA cardiomyopathy.


Assuntos
Cardiomiopatia Dilatada/genética , Ciclo Celular/genética , Reparo do DNA , Lamina Tipo A/genética , Mitocôndrias/metabolismo , Potenciais de Ação/fisiologia , Animais , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Sobrevivência Celular , Dano ao DNA , Modelos Animais de Doenças , Progressão da Doença , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Lamina Tipo A/deficiência , Lamina Tipo A/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/patologia , Fosforilação Oxidativa , Estresse Oxidativo , Transdução de Sinais , Estresse Fisiológico/genética
14.
Cells ; 9(4)2020 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-32244403

RESUMO

LMNA encodes for Lamin A/C, type V intermediate filaments that polymerize under the inner nuclear membrane to form the nuclear lamina. A small fraction of Lamin A/C, less polymerized, is also found in the nucleoplasm. Lamin A/C functions include roles in nuclear resistance to mechanical stress and gene regulation. LMNA mutations are responsible for a wide variety of pathologies, including Emery-Dreifuss (EDMD) and LMNA-related congenital muscular dystrophies (L-CMD) without clear genotype-phenotype correlations. Both diseases presented with striated muscle disorders although L-CMD symptoms appear much earlier and are more severe. Seeking for pathomechanical differences to explain the severity of L-CMD mutations, we performed an in silico analysis of the UMD-LMNA database and found that L-CMD mutations mainly affect residues involved in Lamin dimer and tetramer stability. In line with this, we found increased nucleoplasmic Lamin A/C in L-CMD patient fibroblasts and mouse myoblasts compared to the control and EDMD. L-CMD myoblasts show differentiation defects linked to their inability to upregulate muscle specific nuclear envelope (NE) proteins expression. NE proteins were mislocalized, leading to misshapen nuclei. We conclude that these defects are due to both the absence of Lamin A/C from the nuclear lamina and its maintenance in the nucleoplasm of myotubes.


Assuntos
Lamina Tipo A/deficiência , Lamina Tipo A/metabolismo , Distrofias Musculares/patologia , Distrofia Muscular de Emery-Dreifuss/patologia , Índice de Gravidade de Doença , Animais , Células Cultivadas , Simulação por Computador , Bases de Dados Genéticas , Modelos Animais de Doenças , Humanos , Lamina Tipo A/genética , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Distrofias Musculares/genética , Distrofia Muscular de Emery-Dreifuss/genética , Mutação/genética , Mioblastos/metabolismo , Fenótipo
15.
Muscle Nerve ; 60(6): 668-672, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31498906

RESUMO

INTRODUCTION: The identification of LMNA-related muscular dystrophy is important because it poses life-threatening cardiac complications. However, diagnosis of LMNA-related muscular dystrophy based on clinical features is challenging. METHODS: We reviewed the clinical phenotypes of 14 children with LMNA variants, focusing on the cardiac function and genotypes. RESULTS: Most patients presented with motor developmental delay or gait abnormalities. Eight (57%) patients had prominent neck extensor weakness or contractures. All patients showed ankle contractures at an early stage. Regular cardiac surveillance allowed for the detection of dysrhythmias in 57% of patients at a mean age of 14 years (range, 5-26). All patients had missense variants; however, there were no clear phenotype-genotype correlations. DISCUSSION: Early diagnosis of LMNA-related muscular dystrophy provides an opportunity for cardiac surveillance, potentially leading to the prevention of cardiac mortality in children.


Assuntos
Arritmias Cardíacas/diagnóstico , Cardiomiopatias/diagnóstico , Lamina Tipo A/deficiência , Distrofias Musculares/diagnóstico , Adolescente , Adulto , Arritmias Cardíacas/fisiopatologia , Cardiomiopatias/fisiopatologia , Criança , Pré-Escolar , Diagnóstico Precoce , Ecocardiografia , Eletrocardiografia , Eletrocardiografia Ambulatorial , Feminino , Humanos , Lamina Tipo A/genética , Masculino , Músculo Esquelético/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Distrofias Musculares/fisiopatologia , Mutação de Sentido Incorreto , Estudos Retrospectivos , Adulto Jovem
16.
J Pathol ; 249(4): 509-522, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31372995

RESUMO

The mechanisms by which lamin A/C in CD4+ T-cells control intestinal homeostasis and can cause inflammatory bowel disease (IBD) are unknown. Here, we explore lamin A/C in a mouse model of IBD. Adoptive transfer to Rag1-/- mice of Lmna-/- CD4+ T-cells, which have enhanced regulatory T-cells (Treg) differentiation and function, induced less severe IBD than wild-type T-cells. Lamin A/C deficiency in CD4+ T-cells enhanced transcription of the Treg master regulator FOXP3, thus promoting Treg differentiation, and reduced Th1 polarization, due to epigenetic changes in the Th1 master regulator T-bet. In mesenteric lymph nodes, retinoic acid (RA) released by CD103+ dendritic cells downregulated lamin A/C in CD4+ T-cells, enhancing Treg differentiation. However, non-RA-producing CD103- dendritic cells predominated in peripheral lymph nodes, facilitating lamin A/C expression in CD4+ T-cells and therefore Th1 differentiation. Our findings establish lamin A/C as a key regulator of Th differentiation in physiological conditions and show it as a potential immune-regulatory target in IBD. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Diferenciação Celular , Colite/prevenção & controle , Colo/metabolismo , Lamina Tipo A/deficiência , Linfócitos T Reguladores/metabolismo , Células Th1/metabolismo , Transferência Adotiva , Animais , Colite/imunologia , Colite/metabolismo , Colite/patologia , Colo/imunologia , Colo/patologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Lamina Tipo A/genética , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos Knockout , Transdução de Sinais , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/transplante , Células Th1/imunologia , Tretinoína/metabolismo
17.
PLoS One ; 13(10): e0205878, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30379953

RESUMO

Hutchinson-Gilford progeria syndrome (HGPS) is a very rare fatal disease characterized for accelerated aging. Although the causal agent, a point mutation in LMNA gene, was identified more than a decade ago, the molecular mechanisms underlying HGPS are still not fully understood and, currently, there is no cure for the patients, which die at a mean age of thirteen. With the aim of unraveling non-previously altered molecular pathways in the premature aging process, human cell lines from HGPS patients and from healthy parental controls were studied in parallel using Next-Generation Sequencing (RNAseq) and High-Resolution Quantitative Proteomics (iTRAQ) techniques. After selection of significant proteins and transcripts and crosschecking of the results a small set of protein/transcript pairs were chosen for validation. One of those proteins, ribose-phosphate pyrophosphokinase 1 (PRPS1), is essential for nucleotide synthesis. PRPS1 loss-of-function mutants present lower levels of purine. PRPS1 protein and transcript levels are detected as significantly decreased in HGPS cell lines vs. healthy parental controls. This modulation was orthogonally confirmed by targeted techniques in cell lines and also in an animal model of Progeria, the ZMPSTE24 knock-out mouse. In addition, functional experiments through supplementation with S-adenosyl-methionine (SAMe), a metabolite that is an alternative source of purine, were done. Results indicate that SAMe has a positive effect in the proliferative capacity and reduces senescence-associated Beta-galactosidase staining of the HPGS cell lines. Altogether, our data suggests that nucleotide and, specifically, purine-metabolism, are altered in premature aging, opening a new window for the therapeutic treatment of the disease.


Assuntos
Lamina Tipo A/genética , Progéria/genética , Purinas/metabolismo , RNA Mensageiro/genética , Ribose-Fosfato Pirofosfoquinase/genética , Adulto , Animais , Linhagem Celular , Proliferação de Células , Criança , Biologia Computacional/métodos , Modelos Animais de Doenças , Feminino , Efeito Fundador , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Lamina Tipo A/deficiência , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Metaloendopeptidases/deficiência , Metaloendopeptidases/genética , Camundongos , Camundongos Knockout , Progéria/tratamento farmacológico , Progéria/metabolismo , Progéria/patologia , RNA Mensageiro/metabolismo , Ribose-Fosfato Pirofosfoquinase/deficiência , S-Adenosilmetionina/farmacologia , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
18.
Sci Rep ; 8(1): 16302, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30389963

RESUMO

Congenital muscular dystrophy with laminin α2 chain-deficiency (LAMA2-CMD) is a severe muscle disorder with complex underlying pathogenesis. We have previously employed profiling techniques to elucidate molecular patterns and demonstrated significant metabolic impairment in skeletal muscle from LAMA2-CMD patients and mouse models. Thus, we hypothesize that skeletal muscle metabolism may be a promising pharmacological target to improve muscle function in LAMA2-CMD. Here, we have investigated whether the multifunctional medication metformin could be used to reduce disease in the dy2J/dy2J mouse model of LAMA2-CMD. First, we show gender disparity for several pathological hallmarks of LAMA2-CMD. Second, we demonstrate that metformin treatment significantly increases weight gain and energy efficiency, enhances muscle function and improves skeletal muscle histology in female dy2J/dy2J mice (and to a lesser extent in dy2J/dy2J males). Thus, our current data suggest that metformin may be a potential future supportive treatment that improves many of the pathological characteristics of LAMA2-CMD.


Assuntos
Disparidades nos Níveis de Saúde , Lamina Tipo A/deficiência , Metformina/administração & dosagem , Músculo Esquelético/efeitos dos fármacos , Distrofias Musculares/tratamento farmacológico , Distrofia Muscular Animal/tratamento farmacológico , Administração Oral , Animais , Modelos Animais de Doenças , Progressão da Doença , Feminino , Humanos , Lamina Tipo A/genética , Laminina/genética , Masculino , Camundongos , Músculo Esquelético/patologia , Distrofias Musculares/genética , Distrofias Musculares/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/patologia , Fatores Sexuais , Resultado do Tratamento
19.
Contrast Media Mol Imaging ; 2018: 8327089, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30116163

RESUMO

Brown adipose tissue (BAT) is an important energy metabolic organ that is highly implicated in obesity, type 2 diabetes, and atherosclerosis. Aging is one of the most important determinants of BAT activity. In this study, we used 18F-FDG PET/CT imaging to assess BAT aging in Lmna-/- mice. The maximum standardized uptake value (SUVMax) of the BAT was measured, and the target/nontarget (T/NT) values of BAT were calculated. The transcription and the protein expression levels of the uncoupling protein 1 (UCP1), beta3-adrenergic receptor (ß3-AR), and the PR domain-containing 16 (PRDM16) were measured by quantitative real-time polymerase chain reaction (RT-PCR) and Western blotting or immunohistochemical analysis. Apoptosis and cell senescence rates in the BAT of WT and Lmna-/- mice were determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and by CDKN2A/p16INK4a immunohistochemical staining, respectively. At 14 weeks of age, the BAT SUVMax and the expression levels of UCP1, ß3-AR, and PRDM16 in Lmna-/- mice were significantly reduced relative to WT mice. At the same time, the number of p16INK4a and TUNEL positively stained cells (%) increased in Lmna-/- mice. Collectively, our results indicate that the aging characteristics and the aging regulatory mechanism in the BAT of Lmna-/- mice can mimic the normal BAT aging process.


Assuntos
Tecido Adiposo Marrom/diagnóstico por imagem , Envelhecimento/patologia , Fluordesoxiglucose F18/química , Lamina Tipo A/deficiência , Tomografia por Emissão de Pósitrons combinada à Tomografia Computadorizada , Progéria/diagnóstico por imagem , Progéria/patologia , Animais , Peso Corporal , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Lamina Tipo A/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Fatores de Transcrição/metabolismo , Proteína Desacopladora 1/metabolismo
20.
J Cell Biochem ; 119(10): 8146-8162, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29923310

RESUMO

We studied how deficiency in lamins A/C and lamina-associated polypeptide 2α (Lap2α) affects DNA repair after irradiation. A-type lamins and Lap2α were not recruited to local DNA lesions and did not accumulate to γ-irradiation-induced foci (IRIF), as it is generally observed for well-known marker of DNA lesions, 53BP1 protein. At micro-irradiated chromatin of lmna double knockout (dn) and Lap2α dn cells, 53BP1 protein levels were reduced, compared to locally irradiated wild-type counterpart. Decreased levels of 53BP1 we also observed in whole populations of lmna dn and Lap2α dn cells, irradiated by UV light. We also studied distribution pattern of 53BP1 protein in a genome outside micro-irradiated region. In Lap2α deficient cells, identical fluorescence of mCherry-tagged 53BP1 protein was found at both microirradiated region and surrounding chromatin. However, a well-known marker of double strand breaks, γH2AX, was highly abundant in the lesion-surrounding genome of Lap2α deficient cells. Described changes, induced by irradiation in Lap2α dn cells, were not accompanied by cell cycle changes. In Lap2α dn cells, we additionally performed analysis by FLIM (Fluorescence Lifetime Imaging Microscopy) that showed different dynamic behavior of mCherry-tagged 53BP1 protein pools when it was compared with wild-type (wt) fibroblasts. This analysis revealed three different fractions of mCherry-53BP1 protein. Two of them showed identical exponential decay times (τ1 and τ3), but the decay rate of τ2 and amplitudes of fluorescence decays (A1-A3) were statistically different in wt and Lap2α dn fibroblasts. Moreover, γ-irradiation weakened an interaction between A-type lamins and Lap2α. Together, our results demonstrate how depletion of Lap2α affects DNA damage response (DDR) and how chromatin compactness is changed in Lap2α deficient cells exposed to radiation.


Assuntos
Cromatina/efeitos da radiação , Reparo do DNA , Proteínas de Ligação a DNA/genética , Fibroblastos/efeitos da radiação , Lamina Tipo A/genética , Proteínas de Membrana/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Animais , Linhagem Celular Transformada , Cromatina/química , Cromatina/ultraestrutura , Dano ao DNA , Proteínas de Ligação a DNA/deficiência , Embrião de Mamíferos , Fibroblastos/citologia , Fibroblastos/metabolismo , Recuperação de Fluorescência Após Fotodegradação , Raios gama , Regulação da Expressão Gênica , Genes Reporter , Histonas/genética , Histonas/metabolismo , Lamina Tipo A/deficiência , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas de Membrana/deficiência , Camundongos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo , Raios Ultravioleta
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...