Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-34681951

RESUMO

It is commonly recognized in the field that cancer cells exhibit changes in the size and shape of their nuclei. These features often serve as important biomarkers in the diagnosis and prognosis of cancer patients. Nuclear size can significantly impact cell migration due to its incredibly large size. Nuclear structural changes are predicted to regulate cancer cell migration. Nuclear abnormalities are common across a vast spectrum of cancer types, regardless of tissue source, mutational spectrum, and signaling dependencies. The pervasiveness of nuclear alterations suggests that changes in nuclear structure may be crucially linked to the transformation process. The factors driving these nuclear abnormalities, and the functional consequences, are not completely understood. Nuclear envelope proteins play an important role in regulating nuclear size and structure in cancer. Altered expression of nuclear lamina proteins, including emerin, is found in many cancers and this expression is correlated with better clinical outcomes. A model is emerging whereby emerin, as well as other nuclear lamina proteins, binding to the nucleoskeleton regulates the nuclear structure to impact metastasis. In this model, emerin and lamins play a central role in metastatic transformation, since decreased emerin expression during transformation causes the nuclear structural defects required for increased cell migration, intravasation, and extravasation. Herein, we discuss the cellular functions of nuclear lamina proteins, with a particular focus on emerin, and how these functions impact cancer progression and metastasis.


Assuntos
Proteínas de Membrana/metabolismo , Mutação , Neoplasias/patologia , Proteínas Nucleares/metabolismo , Animais , Humanos , Proteínas de Membrana/genética , Metástase Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética
2.
Muscle Nerve ; 62(1): 128-136, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32304242

RESUMO

INTRODUCTION: Emery-Dreifuss muscular dystrophy (EDMD) is a disease characterized by skeletal muscle wasting, major tendon contractures, and cardiac conduction defects. Mutations in the gene encoding emerin cause EDMD1. Our previous studies suggested that emerin activation of histone deacetylase 3 (HDAC3) to reduce histone 4-lysine 5 (H4K5) acetylation (ac) is important for myogenic differentiation. METHODS: Pharmacological inhibitors (Nu9056, L002) of histone acetyltransferases targeting acetylated H4K5 were used to test whether increased acetylated H4K5 was responsible for the impaired differentiation seen in emerin-deficient myogenic progenitors. RESULTS: Nu9056 and L002 rescued impaired differentiation in emerin deficiency. SRT1720, which inhibits the nicotinamide adenine dinucleotide (NAD)+ -dependent deacetylase sirtuin 1 (SIRT1), failed to rescue myotube formation. DISCUSSION: We conclude that emerin regulation of HDAC3 activity to affect H4K5 acetylation dynamics is important for myogenic differentiation. Targeting H4K5ac dynamics represents a potential new strategy for ameliorating the skeletal muscle wasting seen in EDMD1.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Histona Acetiltransferases/antagonistas & inibidores , Distrofia Muscular de Emery-Dreifuss/tratamento farmacológico , Distrofia Muscular de Emery-Dreifuss/patologia , Células-Tronco/efeitos dos fármacos , Tiazóis/uso terapêutico , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Histona Acetiltransferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco/patologia , Tiazóis/farmacologia
3.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-32824881

RESUMO

ß-dystroglycan (ß-DG) assembles with lamins A/C and B1 and emerin at the nuclear envelope (NE) to maintain proper nuclear architecture and function. To provide insight into the nuclear function of ß-DG, we characterized the interaction between ß-DG and emerin at the molecular level. Emerin is a major NE protein that regulates multiple nuclear processes and whose deficiency results in Emery-Dreifuss muscular dystrophy (EDMD). Using truncated variants of ß-DG and emerin, via a series of in vitro and in vivo binding experiments and a tailored computational analysis, we determined that the ß-DG-emerin interaction is mediated at least in part by their respective transmembrane domains (TM). Using surface plasmon resonance assays we showed that emerin binds to ß-DG with high affinity (KD in the nanomolar range). Remarkably, the analysis of cells in which DG was knocked out demonstrated that loss of ß-DG resulted in a decreased emerin stability and impairment of emerin-mediated processes. ß-DG and emerin are reciprocally required for their optimal targeting within the NE, as shown by immunofluorescence, western blotting and immunoprecipitation assays using emerin variants with mutations in the TM domain and B-lymphocytes of a patient with EDMD. In summary, we demonstrated that ß-DG plays a role as an emerin interacting partner modulating its stability and function.


Assuntos
Distroglicanas/metabolismo , Proteínas de Membrana/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Proteínas Nucleares/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linfócitos B/metabolismo , Sítios de Ligação , Linhagem Celular , Células Cultivadas , Distroglicanas/química , Distroglicanas/genética , Células HeLa , Humanos , Proteínas de Membrana/química , Proteínas de Membrana/genética , Camundongos , Distrofia Muscular de Emery-Dreifuss/genética , Mutação , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Ligação Proteica
4.
Semin Cell Dev Biol ; 29: 95-106, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24365856

RESUMO

Emery-Dreifuss muscular dystrophy (EDMD) is caused by mutations in the genes encoding emerin, lamins A and C and FHL1. Additional EDMD-like syndromes are caused by mutations in nesprins and LUMA. This review will specifically focus on emerin function and the current thinking for how loss or mutations in emerin cause EDMD. Emerin is a well-conserved, ubiquitously expressed protein of the inner nuclear membrane. Emerin has been shown to have diverse functions, including the regulation of gene expression, cell signaling, nuclear structure and chromatin architecture. This review will focus on the relationships between these functions and the EDMD disease phenotype. Additionally it will highlight open questions concerning emerin's roles in cell and nuclear biology and disease.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Lamina Tipo A/genética , Proteínas de Membrana/genética , Distrofia Muscular de Emery-Dreifuss/genética , Lâmina Nuclear/genética , Proteínas Nucleares/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Musculares/genética , Mutação , Lâmina Nuclear/fisiologia , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , beta Catenina/metabolismo
5.
Muscle Nerve ; 51(2): 222-8, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24825363

RESUMO

INTRODUCTION: Mutations in the inner nuclear envelope protein emerin cause Emery-Dreifuss muscular dystrophy (EDMD), which is characterized by progressive skeletal muscle wasting, cardiac conduction defects, and tendon contractures. We previously showed that emerin binds directly to the transcription regulator Lmo7 and attenuates its activity to regulate the proper temporal expression of important myogenic differentiation genes. METHODS: The skeletal muscle and cardiac phenotypes were analyzed in a newly generated Lmo7-null mouse using histological analysis, echocardiography, and various neuromuscular tests to determine if Lmo7 was important for skeletal muscle and cardiac function. RESULTS: Lmo7-null mice had growth retardation, decreased fiber size, and impaired skeletal muscle and cardiac function. Lmo7-null mice also had lower levels of phosphorylated retinoblastoma (Rb), extracellular signal-regulated kinase, and c-Jun N-terminal kinase, which is consistent with altered Rb and mitogen-activated protein kinase signaling. CONCLUSIONS: These findings demonstrate that loss of Lmo7 in mice causes myopathic phenotypes similar to those seen in other EDMD mouse models.


Assuntos
Proteínas com Domínio LIM/deficiência , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/fisiopatologia , Fatores de Transcrição/deficiência , Animais , Índice de Massa Corporal , Peso Corporal/genética , Modelos Animais de Doenças , Ecocardiografia , Regulação da Expressão Gênica/genética , Cardiopatias/genética , Humanos , Proteínas com Domínio LIM/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Contração Muscular/fisiologia , Doenças da Junção Neuromuscular/etiologia , Doenças da Junção Neuromuscular/genética , Fenótipo , Transdução de Sinais/genética , Fatores de Transcrição/genética
6.
Chromosome Res ; 21(8): 765-79, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24062260

RESUMO

The spatial organization of chromatin is critical in establishing cell-type dependent gene expression programs. The inner nuclear membrane protein emerin has been implicated in regulating global chromatin architecture. We show emerin associates with genomic loci of muscle differentiation promoting factors in murine myogenic progenitors, including Myf5 and MyoD. Prior to their transcriptional activation Myf5 and MyoD loci localized to the nuclear lamina in proliferating progenitors and moved to the nucleoplasm upon transcriptional activation during differentiation. The Pax7 locus, which is transcribed in proliferating progenitors, localized to the nucleoplasm and Pax7 moved to the nuclear lamina upon repression during differentiation. Localization of Myf5, MyoD, and Pax7 to the nuclear lamina and proper temporal expression of these genes required emerin and HDAC3. Interestingly, activation of HDAC3 catalytic activity rescued both Myf5 localization to the nuclear lamina and its expression. Collectively, these data support a model whereby emerin facilitates repressive chromatin formation at the nuclear lamina by activating the catalytic activity of HDAC3 to regulate the coordinated spatiotemporal expression of myogenic differentiation genes.


Assuntos
Histona Desacetilases/metabolismo , Proteínas de Membrana/metabolismo , Desenvolvimento Muscular/genética , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX7/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Núcleo Celular/genética , Núcleo Celular/metabolismo , Proliferação de Células , Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos , Histona Desacetilases/genética , Proteínas de Membrana/genética , Camundongos , Proteína MyoD/genética , Fator Regulador Miogênico 5/genética , Proteínas Nucleares/genética , Fator de Transcrição PAX7/genética , Ativação Transcricional
7.
bioRxiv ; 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38712242

RESUMO

During metastasis, cancer cells traverse the vasculature by squeezing through very small gaps in the endothelium. Thus, nuclei in metastatic cancer cells must become more malleable to move through these gaps. Our lab showed invasive breast cancer cells have 50% less emerin protein resulting in smaller, misshapen nuclei, and higher metastasis rates than non-cancerous controls. Thus, emerin deficiency was predicted to cause increased nuclear compliance, cell migration, and metastasis. We tested this hypothesis by downregulating emerin in noninvasive MCF7 cells and found emerin knockdown causes smaller, dysmorphic nuclei, resulting in increased impeded cell migration. Emerin reduction in invasive breast cancer cells showed similar results. Supporting the clinical relevance of emerin reduction in cancer progression, our analysis of 192 breast cancer patient samples showed emerin expression inversely correlates with cancer invasiveness. We conclude emerin loss is an important driver of invasive transformation and has utility as a biomarker for tumor progression.

8.
J Biol Chem ; 287(26): 22080-8, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22570481

RESUMO

Organization of the genome is critical for maintaining cell-specific gene expression, ensuring proper cell function. It is well established that the nuclear lamina preferentially associates with repressed chromatin. However, the molecular mechanisms underlying repressive chromatin formation and maintenance at the nuclear lamina remain poorly understood. Here we show that emerin binds directly to HDAC3, the catalytic subunit of the nuclear co-repressor (NCoR) complex, and recruits HDAC3 to the nuclear periphery. Emerin binding stimulated the catalytic activity of HDAC3, and emerin-null cells exhibit increased H4K5 acetylation, which is the preferred target of the NCoR complex. Emerin-null cells exhibit an epigenetic signature similar to that seen in HDAC3-null cells. Emerin-null cells also had significantly less HDAC3 at the nuclear lamina. Collectively, these data support a model whereby emerin facilitates repressive chromatin formation at the nuclear periphery by increasing the catalytic activity of HDAC3.


Assuntos
Histona Desacetilases/química , Proteínas de Membrana/química , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Animais , Catálise , Núcleo Celular/metabolismo , Cromatina/metabolismo , Ativação Enzimática , Epigênese Genética , Genoma , Histonas/química , Cinética , Camundongos , Microscopia Confocal/métodos , Distrofias Musculares/metabolismo , Ligação Proteica , Frações Subcelulares/metabolismo
9.
J Cell Sci ; 124(Pt 10): 1691-702, 2011 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-21525034

RESUMO

X-linked Emery-Dreifuss muscular dystrophy (X-EDMD) is caused by mutations in the inner nuclear membrane protein emerin. Previous studies have shown that emerin binds to and inhibits the activity of LIM domain only 7 (Lmo7), a transcription factor that regulates the expression of genes implicated in X-EDMD. Here, we analyzed Lmo7 function in C2C12 myoblast differentiation and its regulation by emerin. We found that Lmo7 was required for proper myoblast differentiation. Lmo7-downregulated myoblasts exhibited reduced expression of Pax3, Pax7, Myf5 and MyoD, whereas overexpression of GFP-Lmo7 increased the expression of MyoD and Myf5. Upon myotube formation, Lmo7 shuttled from the nucleus to the cytoplasm, concomitant with reduced expression of MyoD, Pax3 and Myf5. Importantly, we show that Lmo7 bound the Pax3, MyoD and Myf5 promoters both in C2C12 myoblasts and in vitro. Because emerin inhibited Lmo7 activity, we tested whether emerin competed with the MyoD promoter for binding to Lmo7 or whether emerin sequestered promoter-bound Lmo7 to the nuclear periphery. Supporting the competition model, emerin binding to Lmo7 inhibited Lmo7 binding to and activation of the MyoD and Pax3 promoters. These findings support the hypothesis that the functional interaction between emerin and Lmo7 is crucial for temporally regulating the expression of key myogenic differentiation genes.


Assuntos
Proteínas de Homeodomínio/antagonistas & inibidores , Proteínas de Membrana/genética , Proteína MyoD/genética , Mioblastos/fisiologia , Proteínas Nucleares/genética , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição/antagonistas & inibidores , Animais , Diferenciação Celular/genética , Processos de Crescimento Celular/genética , Linhagem Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Regulação para Baixo , Imunofluorescência , Proteínas de Homeodomínio/metabolismo , Proteínas com Domínio LIM , Proteínas de Membrana/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/metabolismo , Proteínas Nucleares/metabolismo , Fator de Transcrição PAX3 , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Transcrição Gênica , Ativação Transcricional
10.
Front Cell Dev Biol ; 10: 1007120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36274837

RESUMO

X-Linked Emery-Dreifuss muscular dystrophy is caused by mutations in the gene encoding emerin. Emerin is an inner nuclear membrane protein important for repressive chromatin organization at the nuclear periphery. Myogenic differentiation is a tightly regulated process characterized by genomic reorganization leading to coordinated temporal expression of key transcription factors, including MyoD, Pax7, and Myf5. Emerin was shown to interact with repressive histone modification machinery, including HDAC3 and EZH2. Using emerin-null myogenic progenitor cells we established several EDMD-causing emerin mutant lines in the effort to understand how the functional interaction of emerin with HDAC3 regulates histone methyltransferase localization or function to organize repressive chromatin at the nuclear periphery. We found that, in addition to its interaction with HDAC3, emerin interacts with the histone methyltransferases EZH2 and G9a in myogenic progenitor cells. Further, we show enhanced binding of emerin HDAC3-binding mutants S54F and Q133H to EZH2 and G9a. Treatment with small molecule inhibitors of EZH2 and G9a reduced H3K9me2 or H3K27me3 throughout differentiation. EZH2 and G9a inhibitors impaired cell cycle withdrawal, differentiation commitment, and myotube formation in wildtype progenitors, while they had no effect on emerin-null progenitors. Interestingly, these inhibitors exacerbated the impaired differentiation of emerin S54F and Q133H mutant progenitors. Collectively, these results suggest the functional interaction between emerin and HDAC3, EZH2, and G9a are important for myogenic differentiation.

11.
Hum Mol Genet ; 18(4): 607-20, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19008300

RESUMO

Mutations in the gene encoding the inner nuclear membrane proteins lamins A and C produce cardiac and skeletal muscle dysfunction referred to as Emery Dreifuss muscular dystrophy. Lamins A and C participate in the LINC complex that, along with the nesprin and SUN proteins, LInk the Nucleoskeleton with the Cytoskeleton. Nesprins 1 and 2 are giant spectrin-repeat containing proteins that have large and small forms. The nesprins contain a transmembrane anchor that tethers to the nuclear membrane followed by a short domain that resides within the lumen between the inner and outer nuclear membrane. Nesprin's luminal domain binds directly to SUN proteins. We generated mice where the C-terminus of nesprin-1 was deleted. This strategy produced a protein lacking the transmembrane and luminal domains that together are referred to as the KASH domain. Mice homozygous for this mutation exhibit lethality with approximately half dying at or near birth from respiratory failure. Surviving mice display hindlimb weakness and an abnormal gait. With increasing age, kyphoscoliosis, muscle pathology and cardiac conduction defects develop. The protein components of the LINC complex, including mutant nesprin-1alpha, lamin A/C and SUN2, are localized at the nuclear membrane in this model. However, the LINC components do not normally associate since coimmunoprecipitation experiments with SUN2 and nesprin reveal that mutant nesprin-1 protein no longer interacts with SUN2. These findings demonstrate the role of the LINC complex, and nesprin-1, in neuromuscular and cardiac disease.


Assuntos
Inativação Gênica , Distrofia Muscular de Emery-Dreifuss/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas Nucleares/genética , Animais , Proteínas do Citoesqueleto , Modelos Animais de Doenças , Feminino , Humanos , Laminas/genética , Laminas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Emery-Dreifuss/embriologia , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fenótipo , Ligação Proteica , Estrutura Terciária de Proteína
12.
Curr Opin Cell Biol ; 14(3): 357-64, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12067659

RESUMO

The nuclear lamina is composed of both A- and B-type lamins and lamin-binding proteins. Many lamin-binding proteins are integral proteins of the inner nuclear membrane. Lamins and inner nuclear membrane proteins are important for a variety of cell functions, including nuclear assembly, replication, transcription, and nuclear integrity. Recent advances in the field in the past year include the identification of a family of spectrin-repeat-containing inner nuclear membrane proteins and other novel inner-membrane proteins, and the discovery of a nuclear membrane fusion complex. There is also growing evidence that A- and B-type lamins and their binding partners have distinct roles during nuclear assembly and interphase.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Laminas/fisiologia , Membrana Nuclear/metabolismo , Proteínas de Ancoragem à Quinase A , Animais , Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica , Doenças Genéticas Inatas/etiologia , Humanos , Fusão de Membrana , Proteínas de Membrana/metabolismo , Poro Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Plantas/genética
13.
Mol Cancer Res ; 19(7): 1196-1207, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33771882

RESUMO

Nuclear envelope proteins play an important role in regulating nuclear size and structure in cancer. Altered expression of nuclear lamins are found in many cancers and its expression is correlated with better clinical outcomes. The nucleus is the largest organelle in the cell with a diameter between 10 and 20 µm. Nuclear size significantly impacts cell migration. Nuclear structural changes are predicted to impact cancer metastasis by regulating cancer cell migration. Here we show emerin regulates nuclear structure in invasive breast cancer cells to impact cancer metastasis. Invasive breast cancer cells had 40% to 50% less emerin than control cells, which resulted in decreased nuclear size. Overexpression of GFP-emerin in invasive breast cancer cells rescued nuclear size and inhibited migration through 3.0 and 8.0 µm pores. Mutational analysis showed emerin binding to nucleoskeletal proteins was important for its regulation of nuclear structure, migration, and invasion. Importantly, emerin expression inhibited lung metastasis by 91% in orthotopic mouse models of breast cancer. Emerin nucleoskeleton-binding mutants failed to inhibit metastasis. These results support a model whereby emerin binding to the nucleoskeleton regulates nuclear structure to impact metastasis. In this model, emerin plays a central role in metastatic transformation, because decreased emerin expression during transformation causes the nuclear structural defects required for increased cell migration, intravasation, and extravasation. IMPLICATIONS: Modulating emerin expression and function represents new targets for therapeutic interventions of metastasis, because increased emerin expression rescued cancer metastasis.


Assuntos
Neoplasias da Mama/genética , Movimento Celular/genética , Núcleo Celular/genética , Proteínas de Membrana/genética , Matriz Nuclear/genética , Proteínas Nucleares/genética , Animais , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Ciclo Celular/genética , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Proliferação de Células/genética , Células Cultivadas , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Proteínas de Membrana/metabolismo , Camundongos Nus , Microscopia Confocal/métodos , Metástase Neoplásica , Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Ligação Proteica , Transplante Heterólogo
14.
Circ Res ; 103(1): 16-23, 2008 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-18596264

RESUMO

The human genome is contained within the nucleus and is separated from the cytoplasm by the nuclear envelope. Mutations in the nuclear envelope proteins emerin and lamin A cause a number of diseases including premature aging syndromes, muscular dystrophy, and cardiomyopathy. Emerin and lamin A are implicated in regulating muscle- and heart-specific gene expression and nuclear architecture. For example, lamin A regulates the expression and localization of gap junction and intercalated disc components. Additionally, emerin and lamin A are also required to maintain nuclear envelope integrity. Demonstrating the importance of maintaining nuclear integrity in heart disease, atrioventricular node cells lacking lamin A exhibit increased nuclear deformation and apoptosis. This review highlights the present understanding of lamin A and emerin function in regulating nuclear architecture, gene expression, and cell signaling and discusses putative mechanisms for how specific mutations in lamin A and emerin cause cardiac- or muscle-specific disease.


Assuntos
Cardiomiopatias/metabolismo , Genoma Humano , Lamina Tipo A/metabolismo , Proteínas de Membrana/metabolismo , Miocárdio/metabolismo , Lâmina Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Senilidade Prematura/genética , Senilidade Prematura/metabolismo , Senilidade Prematura/patologia , Animais , Apoptose/genética , Nó Atrioventricular/metabolismo , Nó Atrioventricular/patologia , Cardiomiopatias/genética , Cardiomiopatias/patologia , Regulação da Expressão Gênica/genética , Humanos , Lamina Tipo A/genética , Proteínas de Membrana/genética , Distrofias Musculares/genética , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Mutação , Miocárdio/patologia , Lâmina Nuclear/genética , Lâmina Nuclear/patologia , Proteínas Nucleares/genética , Especificidade de Órgãos/genética , Transdução de Sinais/genética , Síndrome
15.
Cells ; 9(6)2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32549231

RESUMO

Mutations in the gene encoding emerin (EMD) cause Emery-Dreifuss muscular dystrophy (EDMD1), an inherited disorder characterized by progressive skeletal muscle wasting, irregular heart rhythms and contractures of major tendons. The skeletal muscle defects seen in EDMD are caused by failure of muscle stem cells to differentiate and regenerate the damaged muscle. However, the underlying mechanisms remain poorly understood. Most EDMD1 patients harbor nonsense mutations and have no detectable emerin protein. There are three EDMD-causing emerin mutants (S54F, Q133H, and D95-99) that localize correctly to the nuclear envelope and are expressed at wildtype levels. We hypothesized these emerin mutants would share in the disruption of key molecular pathways involved in myogenic differentiation. We generated myogenic progenitors expressing wildtype emerin and each EDMD1-causing emerin mutation (S54F, Q133H, D95-99) in an emerin-null (EMD-/y) background. S54F, Q133H, and D95-99 failed to rescue EMD-/y myogenic differentiation, while wildtype emerin efficiently rescued differentiation. RNA sequencing was done to identify pathways and networks important for emerin regulation of myogenic differentiation. This analysis significantly reduced the number of pathways implicated in EDMD1 muscle pathogenesis.


Assuntos
Diferenciação Celular/fisiologia , Desenvolvimento Muscular/fisiologia , Distrofia Muscular de Emery-Dreifuss/metabolismo , Mioblastos/metabolismo , Diferenciação Celular/genética , Humanos , Desenvolvimento Muscular/genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patologia , Membrana Nuclear/metabolismo , Regeneração/genética
17.
PLoS Biol ; 2(9): E231, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15328537

RESUMO

X-linked Emery-Dreifuss muscular dystrophy is caused by loss of emerin, a LEM-domain protein of the nuclear inner membrane. To better understand emerin function, we used affinity chromatography to purify emerin-binding proteins from nuclear extracts of HeLa cells. Complexes that included actin, alphaII-spectrin and additional proteins, bound specifically to emerin. Actin polymerization assays in the presence or absence of gelsolin or capping protein showed that emerin binds and stabilizes the pointed end of actin filaments, increasing the actin polymerization rate 4- to 12-fold. We propose that emerin contributes to the formation of an actin-based cortical network at the nuclear inner membrane, conceptually analogous to the actin cortical network at the plasma membrane. Thus, in addition to disrupting transcription factors that bind emerin, loss of emerin may destabilize nuclear envelope architecture by weakening a nuclear actin network.


Assuntos
Citoesqueleto de Actina/química , Actinas/química , Núcleo Celular/metabolismo , Proteínas de Membrana/química , Membrana Nuclear/metabolismo , Timopoietinas/química , Sítios de Ligação , Membrana Celular/metabolismo , Cromatografia de Afinidade , Gelsolina/química , Células HeLa , Humanos , Distrofias Musculares/patologia , Proteínas Nucleares , Ligação Proteica , Transcrição Gênica
18.
Mol Cell Biol ; 22(17): 6286-97, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12167720

RESUMO

We have characterized a pathway for nuclear export of the glucocorticoid receptor (GR) in mammalian cells. This pathway involves the Ca2+ -binding protein calreticulin (CRT), which directly contacts the DNA binding domain (DBD) of GR and facilitates its delivery from the nucleus to the cytoplasm. In the present study, we investigated the role of Ca2+ in CRT-dependent export of GR. We found that removal of Ca2+ from CRT inhibits its capacity to stimulate the nuclear export of GR in digitonin-permeabilized cells and that the inhibition is due to the failure of Ca2+-free CRT to bind the DBD. These effects are reversible, since DBD binding and nuclear export can be restored by Ca2+ addition. Depletion of intracellular Ca2+ inhibits GR export in intact cells under conditions that do not inhibit other nuclear transport pathways, suggesting that there is a Ca2+ requirement for GR export in vivo. We also found that the Ran GTPase is not required for GR export. These data show that the nuclear export pathway used by steroid hormone receptors such as GR is distinct from the Crm1 pathway. We suggest that signaling events that increase Ca2+ could positively regulate CRT and inhibit GR function through nuclear export.


Assuntos
Transporte Ativo do Núcleo Celular/fisiologia , Proteínas de Ligação ao Cálcio/fisiologia , Cálcio/fisiologia , Núcleo Celular/metabolismo , Receptores de Glucocorticoides/metabolismo , Ribonucleoproteínas/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Sítios de Ligação , Proteínas de Ligação ao Cálcio/química , Calreticulina , Compartimento Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citoplasma/metabolismo , DNA/metabolismo , Digoxina/farmacologia , Retículo Endoplasmático/metabolismo , Camundongos , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Ribonucleoproteínas/química , Especificidade por Substrato , Proteína ran de Ligação ao GTP/fisiologia
19.
Cells ; 6(4)2017 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-29065506

RESUMO

Mutations in the gene encoding emerin cause Emery-Dreifuss muscular dystrophy (EDMD), a disorder causing progressive skeletal muscle wasting, irregular heart rhythms and contractures of major tendons. RNA sequencing was performed on differentiating wildtype and emerin-null myogenic progenitors to identify molecular pathways implicated in EDMD, 340 genes were uniquely differentially expressed during the transition from day 0 to day 1 in wildtype cells. 1605 genes were uniquely expressed in emerin-null cells; 1706 genes were shared among both wildtype and emerin-null cells. One thousand and forty-seven transcripts showed differential expression during the transition from day 1 to day 2. Four hundred and thirty-one transcripts showed altered expression in both wildtype and emerin-null cells. Two hundred and ninety-five transcripts were differentially expressed only in emerin-null cells and 321 transcripts were differentially expressed only in wildtype cells. DAVID, STRING and Ingenuity Pathway Analysis identified pathways implicated in impaired emerin-null differentiation, including cell signaling, cell cycle checkpoints, integrin signaling, YAP/TAZ signaling, stem cell differentiation, and multiple muscle development and myogenic differentiation pathways. Functional enrichment analysis showed biological functions associated with the growth of muscle tissue and myogenesis of skeletal muscle were inhibited. The large number of differentially expressed transcripts upon differentiation induction suggests emerin functions during transcriptional reprograming of progenitors to committed myoblasts.

20.
Dis Model Mech ; 10(4): 385-397, 2017 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-28188262

RESUMO

Mutations in the gene encoding emerin cause Emery-Dreifuss muscular dystrophy (EDMD). Emerin is an integral inner nuclear membrane protein and a component of the nuclear lamina. EDMD is characterized by skeletal muscle wasting, cardiac conduction defects and tendon contractures. The failure to regenerate skeletal muscle is predicted to contribute to the skeletal muscle pathology of EDMD. We hypothesize that muscle regeneration defects are caused by impaired muscle stem cell differentiation. Myogenic progenitors derived from emerin-null mice were used to confirm their impaired differentiation and analyze selected myogenic molecular pathways. Emerin-null progenitors were delayed in their cell cycle exit, had decreased myosin heavy chain (MyHC) expression and formed fewer myotubes. Emerin binds to and activates histone deacetylase 3 (HDAC3). Here, we show that theophylline, an HDAC3-specific activator, improved myotube formation in emerin-null cells. Addition of the HDAC3-specific inhibitor RGFP966 blocked myotube formation and MyHC expression in wild-type and emerin-null myogenic progenitors, but did not affect cell cycle exit. Downregulation of emerin was previously shown to affect the p38 MAPK and ERK/MAPK pathways in C2C12 myoblast differentiation. Using a pure population of myogenic progenitors completely lacking emerin expression, we show that these pathways are also disrupted. ERK inhibition improved MyHC expression in emerin-null cells, but failed to rescue myotube formation or cell cycle exit. Inhibition of p38 MAPK prevented differentiation in both wild-type and emerin-null progenitors. These results show that each of these molecular pathways specifically regulates a particular stage of myogenic differentiation in an emerin-dependent manner. Thus, pharmacological targeting of multiple pathways acting at specific differentiation stages may be a better therapeutic approach in the future to rescue muscle regeneration in vivo.


Assuntos
Diferenciação Celular , Histona Desacetilases/metabolismo , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Desenvolvimento Muscular , Proteínas Nucleares/metabolismo , Células-Tronco/metabolismo , Acetilação , Animais , Butadienos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Flavonoides/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Histonas/metabolismo , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Modelos Biológicos , Desenvolvimento Muscular/efeitos dos fármacos , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Cadeias Pesadas de Miosina/metabolismo , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Células-Tronco/efeitos dos fármacos , Teofilina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA