Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
1.
J Cell Sci ; 137(16)2024 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-39092499

RESUMEN

Lamins are intermediate filament proteins that contribute to numerous cellular functions, including nuclear morphology and mechanical stability. The N-terminal head domain of lamin is crucial for higher order filament assembly and function, yet the effects of commonly used N-terminal tags on lamin function remain largely unexplored. Here, we systematically studied the effect of two differently sized tags on lamin A (LaA) function in a mammalian cell model engineered to allow for precise control of expression of tagged lamin proteins. Untagged, FLAG-tagged and GFP-tagged LaA completely rescued nuclear shape defects when expressed at similar levels in lamin A/C-deficient (Lmna-/-) MEFs, and all LaA constructs prevented increased nuclear envelope ruptures in these cells. N-terminal tags, however, altered the nuclear localization of LaA and impaired the ability of LaA to restore nuclear deformability and to recruit emerin to the nuclear membrane in Lmna-/- MEFs. Our finding that tags impede some LaA functions but not others might explain the partial loss of function phenotypes when tagged lamins are expressed in model organisms and should caution researchers using tagged lamins to study the nucleus.


Asunto(s)
Núcleo Celular , Lamina Tipo A , Membrana Nuclear , Lamina Tipo A/metabolismo , Lamina Tipo A/genética , Animales , Ratones , Núcleo Celular/metabolismo , Membrana Nuclear/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética
2.
Dev Biol ; 515: 139-150, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39038593

RESUMEN

Stem cell loss in aging and disease is associated with nuclear deformation. Yet, how nuclear shape influences stem cell homeostasis is poorly understood. We investigated this connection using Drosophila germline stem cells, as survival of these stem cells is compromised by dysfunction of the nuclear lamina, the extensive protein network that lines the inner nuclear membrane and gives shape to the nucleus. To induce nuclear distortion in germline stem cells, we used the GAL4-UAS system to increase expression of the permanently farnesylated nuclear lamina protein, Kugelkern, a rate limiting factor for nuclear growth. We show that elevated Kugelkern levels cause severe nuclear distortion in germline stem cells, including extensive thickening and lobulation of the nuclear envelope and nuclear lamina, as well as alteration of internal nuclear compartments. Despite these changes, germline stem cell number, proliferation, and female fertility are preserved, even as females age. Collectively, these data demonstrate that disruption of nuclear architecture does not cause a failure of germline stem cell survival or homeostasis, revealing that nuclear deformation does not invariably promote stem cell loss.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Células Germinativas , Homeostasis , Lámina Nuclear , Células Madre , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Femenino , Células Germinativas/metabolismo , Drosophila melanogaster/metabolismo , Células Madre/metabolismo , Lámina Nuclear/metabolismo , Núcleo Celular/metabolismo , Proliferación Celular , Drosophila/metabolismo , Membrana Nuclear/metabolismo
3.
J Cell Sci ; 135(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35178558

RESUMEN

Emerin is an integral nuclear envelope protein that participates in the maintenance of nuclear shape. When mutated or absent, emerin causes X-linked Emery-Dreifuss muscular dystrophy (EDMD). To understand how emerin takes part in molecular --scaffolding at the nuclear envelope and helps protect the nucleus against mechanical stress, we established its nanoscale organization using single-molecule tracking and super-resolution microscopy. We show that emerin monomers form localized oligomeric nanoclusters stabilized by both lamin A/C and the SUN1-containing linker of nucleoskeleton and cytoskeleton (LINC) complex. Interactions of emerin with nuclear actin and BAF (also known as BANF1) additionally modulate its membrane mobility and its ability to oligomerize. In nuclei subjected to mechanical challenges, the mechanotransduction functions of emerin are coupled to changes in its oligomeric state, and the incremental self-assembly of emerin determines nuclear shape adaptation against mechanical forces. We also show that the abnormal nuclear envelope deformations induced by EDMD emerin mutants stem from improper formation of lamin A/C and LINC complex-stabilized emerin oligomers. These findings place emerin at the center of the molecular processes that regulate nuclear shape remodeling in response to mechanical challenges.


Asunto(s)
Distrofia Muscular de Emery-Dreifuss , Membrana Nuclear , Humanos , Mecanotransducción Celular , Proteínas de la Membrana , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
4.
J Cell Sci ; 135(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-35362531

RESUMEN

When metastasizing, tumor cells must traverse environments with diverse physicochemical properties. Recently, the cell nucleus has emerged as a major regulator of the transition from mesenchymal to fast amoeboid (leader bleb-based) migration. Here, we demonstrate that increasing nuclear stiffness through elevating lamin A, inhibits fast amoeboid migration in melanoma cells. Importantly, nuclei may respond to force through stiffening. A key factor in this process is the inner nuclear membrane (INM) protein emerin. Accordingly, we determined the role of emerin in regulating fast amoeboid migration. Strikingly, we found that both the up- and downregulation of emerin results in an inhibition of fast amoeboid migration. However, when key Src phosphorylation sites were removed, upregulation of emerin no longer inhibited fast amoeboid migration. Interestingly, as measured by using a Src biosensor, activity of Src was low in cells within a confined environment. Thus, the fast amoeboid migration of melanoma cells depends on the precise calibration of emerin activity.


Asunto(s)
Amoeba , Melanoma , Amoeba/metabolismo , Núcleo Celular/metabolismo , Humanos , Melanoma/patología , Proteínas de la Membrana , Membrana Nuclear/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo
5.
Mol Biol Rep ; 51(1): 898, 2024 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-39115711

RESUMEN

BACKGROUND: The nuclear envelope (NE), which is composed of the outer and inner nuclear membranes, the nuclear pore complex and the nuclear lamina, regulates a plethora of cellular processes, including those that restrict cancer development (genomic stability, cell cycle regulation, and cell migration). Thus, impaired NE is functionally related to tumorigenesis, and monitoring of NE alterations is used to diagnose cancer. However, the chronology of NE changes occurring during cancer evolution and the connection between them remained to be precisely defined, due to the lack of appropriate cell models. METHODS: The expression and subcellular localization of NE proteins (lamins A/C and B1 and the inner nuclear membrane proteins emerin and ß-dystroglycan [ß-DG]) during prostate cancer progression were analyzed, using confocal microscopy and western blot assays, and a prostate cancer cell system comprising RWPE-1 epithelial prostate cells and several prostate cancer cell lines with different invasiveness. RESULTS: Deformed nuclei and the mislocalization and low expression of lamin A/C, lamin B1, and emerin became more prominent as the invasiveness of the prostate cancer lines increased. Suppression of lamin A/C expression was an early event during prostate cancer evolution, while a more extensive deregulation of NE proteins, including ß-DG, occurred in metastatic prostate cells. CONCLUSIONS: The RWPE-1 cell line-based system was found to be suitable for the correlation of NE impairment with prostate cancer invasiveness and determination of the chronology of NE alterations during prostate carcinogenesis. Further study of this cell system would help to identify biomarkers for prostate cancer prognosis and diagnosis.


Asunto(s)
Lamina Tipo A , Lamina Tipo B , Proteínas de la Membrana , Membrana Nuclear , Proteínas Nucleares , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/genética , Membrana Nuclear/metabolismo , Línea Celular Tumoral , Proteínas de la Membrana/metabolismo , Lamina Tipo B/metabolismo , Lamina Tipo A/metabolismo , Lamina Tipo A/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Distroglicanos/metabolismo , Regulación Neoplásica de la Expresión Génica , Núcleo Celular/metabolismo
6.
Eur Heart J ; 44(48): 5064-5073, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37639473

RESUMEN

BACKGROUND AND AIMS: Emery-Dreifuss muscular dystrophy (EDMD) is caused by variants in EMD (EDMD1) and LMNA (EDMD2). Cardiac conduction defects and atrial arrhythmia are common to both, but LMNA variants also cause end-stage heart failure (ESHF) and malignant ventricular arrhythmia (MVA). This study aimed to better characterize the cardiac complications of EMD variants. METHODS: Consecutively referred EMD variant-carriers were retrospectively recruited from 12 international cardiomyopathy units. MVA and ESHF incidences in male and female variant-carriers were determined. Male EMD variant-carriers with a cardiac phenotype at baseline (EMDCARDIAC) were compared with consecutively recruited male LMNA variant-carriers with a cardiac phenotype at baseline (LMNACARDIAC). RESULTS: Longitudinal follow-up data were available for 38 male and 21 female EMD variant-carriers [mean (SD) ages 33.4 (13.3) and 43.3 (16.8) years, respectively]. Nine (23.7%) males developed MVA and five (13.2%) developed ESHF during a median (inter-quartile range) follow-up of 65.0 (24.3-109.5) months. No female EMD variant-carrier had MVA or ESHF, but nine (42.8%) developed a cardiac phenotype at a median (inter-quartile range) age of 58.6 (53.2-60.4) years. Incidence rates for MVA were similar for EMDCARDIAC and LMNACARDIAC (4.8 and 6.6 per 100 person-years, respectively; log-rank P = .49). Incidence rates for ESHF were 2.4 and 5.9 per 100 person-years for EMDCARDIAC and LMNACARDIAC, respectively (log-rank P = .09). CONCLUSIONS: Male EMD variant-carriers have a risk of progressive heart failure and ventricular arrhythmias similar to that of male LMNA variant-carriers. Early implantable cardioverter defibrillator implantation and heart failure drug therapy should be considered in male EMD variant-carriers with cardiac disease.


Asunto(s)
Cardiopatías , Insuficiencia Cardíaca , Distrofia Muscular de Emery-Dreifuss , Distrofia Muscular de Emery-Dreifuss Ligada a X , Humanos , Masculino , Femenino , Persona de Mediana Edad , Distrofia Muscular de Emery-Dreifuss Ligada a X/complicaciones , Estudios Retrospectivos , Arritmias Cardíacas/epidemiología , Arritmias Cardíacas/genética , Arritmias Cardíacas/complicaciones , Cardiopatías/complicaciones , Distrofia Muscular de Emery-Dreifuss/complicaciones , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/complicaciones , Mutación
7.
Exp Cell Res ; 418(1): 113265, 2022 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-35716785

RESUMEN

Understanding the mechanisms of colorectal cancer (CRC) progression is critical for developing innovative treatment strategies. As an endoplasmic reticulum-located protein, B cell receptor-associated protein 31 (BCAP31) has been identified to be highly expressed in multiple cancers. However, its function and molecular mechanism in CRC remain not fully understood. In the present study, BCAP31 expression and its correlation with the clinical stage were analyzed based on TCGA database. We demonstrated that loss of BCAP31 suppressed CRC cell proliferation in vitro and tumor growth in vivo. Mechanistically, we demonstrated that Emerin was an interaction partner and downstream molecule of BCAP31. Knockdown of BCAP31 promoted the nuclear envelope localization of Emerin, leading to a reduction of ß-catenin accumulation in the nucleus, which resulted in downregulation of Wnt/ß-catenin downstream target genes, including c-Myc, cyclin D1, Survivin, and Mcl-1. Moreover, downregulation of Emerin partially restored the BCAP31 depletion-mediated ß-catenin protein level and tumor suppressive effects in CRC cells.Our data highlights the pivotal role of BCAP31 depletion in inhibiting cell proliferation in CRC cells, and mechanistically via Emerin/ß-catenin signaling, which may serve as a promising target for CRC treatment.


Asunto(s)
Neoplasias Colorrectales , beta Catenina , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas Nucleares , Receptores de Antígenos de Linfocitos B/genética , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
8.
J Proteome Res ; 21(9): 2197-2210, 2022 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-35972904

RESUMEN

Emerin and lamin B receptor (LBR) are abundant transmembrane proteins of the nuclear envelope that are concentrated at the inner nuclear membrane (INM). Although both proteins interact with chromatin and nuclear lamins, they have distinctive biochemical and functional properties. Here, we have deployed proximity labeling using the engineered biotin ligase TurboID (TbID) and quantitative proteomics to compare the neighborhoods of emerin and LBR in cultured mouse embryonic fibroblasts. Our analysis revealed 232 high confidence proximity partners that interact selectively with emerin and/or LBR, 49 of which are shared by both. These included previously characterized NE-concentrated proteins, as well as a host of additional proteins not previously linked to emerin or LBR functions. Many of these are TM proteins of the ER, including two E3 ubiquitin ligases. Supporting these results, we found that 11/12 representative proximity relationships identified by TbID also were detected at the NE with the proximity ligation assay. Overall, this work presents methodology that may be used for large-scale mapping of the landscape of the INM and reveals a group of new proteins with potential functional connections to emerin and LBR.


Asunto(s)
Lamina Tipo A , Proteómica , Animales , Fibroblastos/metabolismo , Lamina Tipo A/metabolismo , Proteínas de la Membrana , Ratones , Proteínas Nucleares , Receptores Citoplasmáticos y Nucleares , Receptor de Lamina B
9.
J Cell Mol Med ; 26(19): 5054-5066, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36106556

RESUMEN

Emerin is an inner nuclear envelope protein encoded by the EMD gene, mutations in which cause Emery-Dreifuss muscular dystrophy type 1 (EDMD1). Cardiac involvement has become a major threat to patients with EDMD1; however, the cardiovascular phenotype spectrums of emerinopathy and the mechanisms by which emerin regulates cardiac pathophysiology remain unclear. Here, we identified a novel nonsense mutation (c.C57G, p.Y19X) in the EMD gene in a Han Chinese family through high-throughput sequencing. Two family members were found to have EDMD1 with muscle weakness and cardiac arrhythmia. Mechanistically, we first discovered that knockdown of emerin in HL-1 or H9C2 cardiomyocytes lead to impaired mitochondrial oxidative phosphorylation capacity with downregulation of electron transport chain complex I and IV and upregulation of complex III and V. Moreover, loss of emerin in HL-1 cells resulted in collapsed mitochondrial membrane potential, altered mitochondrial networks and downregulated multiple factors in RNA and protein level, such as PGC1α, DRP1, MFF, MFN2, which are involved in regulation of mitochondrial biogenesis, fission and fusion. Our findings suggest that targeting mitochondrial bioenergetics might be an effective strategy against cardiac disorders caused by EMD mutations.


Asunto(s)
Distrofias Musculares , Distrofia Muscular de Emery-Dreifuss , Distrofia Muscular de Emery-Dreifuss Ligada a X , Codón sin Sentido , Complejo III de Transporte de Electrones/genética , Humanos , Proteínas de la Membrana , Mitocondrias/genética , Distrofias Musculares/genética , Distrofia Muscular de Emery-Dreifuss/genética , Mutación/genética , Miocitos Cardíacos , Proteínas Nucleares , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética
10.
Int J Mol Sci ; 23(1)2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-35008948

RESUMEN

Myotonic dystrophy type 1 (DM1) is a hereditary and multisystemic disease characterized by myotonia, progressive distal muscle weakness and atrophy. The molecular mechanisms underlying this disease are still poorly characterized, although there are some hypotheses that envisage to explain the multisystemic features observed in DM1. An emergent hypothesis is that nuclear envelope (NE) dysfunction may contribute to muscular dystrophies, particularly to DM1. Therefore, the main objective of the present study was to evaluate the nuclear profile of DM1 patient-derived and control fibroblasts and to determine the protein levels and subcellular distribution of relevant NE proteins in these cell lines. Our results demonstrated that DM1 patient-derived fibroblasts exhibited altered intracellular protein levels of lamin A/C, LAP1, SUN1, nesprin-1 and nesprin-2 when compared with the control fibroblasts. In addition, the results showed an altered location of these NE proteins accompanied by the presence of nuclear deformations (blebs, lobes and/or invaginations) and an increased number of nuclear inclusions. Regarding the nuclear profile, DM1 patient-derived fibroblasts had a larger nuclear area and a higher number of deformed nuclei and micronuclei than control-derived fibroblasts. These results reinforce the evidence that NE dysfunction is a highly relevant pathological characteristic observed in DM1.


Asunto(s)
Biomarcadores , Fibroblastos/metabolismo , Membrana Nuclear/metabolismo , Núcleo Celular/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Espacio Intracelular/metabolismo , Lamina Tipo A/metabolismo , Proteínas de la Membrana/metabolismo , Distrofia Miotónica/genética , Distrofia Miotónica/metabolismo , Proteína Quinasa de Distrofia Miotónica/metabolismo , Proteínas Nucleares/metabolismo
11.
Neurogenetics ; 22(1): 33-41, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33405017

RESUMEN

The nuclear envelope (NE) separates the nucleus from the cytoplasm in all eukaryotic cells. A disruption of the NE structure compromises normal gene regulation and leads to severe human disorders collectively classified as nuclear envelopathies and affecting skeletal muscle, heart, brain, skin, and bones. The ubiquitous NE component LAP1B is encoded by TOR1AIP1, and the use of an alternative start codon gives rise to the shorter LAP1C isoform. TOR1AIP1 mutations have been identified in patients with diverging clinical presentations such as muscular dystrophy, progressive dystonia with cerebellar atrophy, and a severe multi-systemic disorder, but the correlation between the mutational effect and the clinical spectrum remains to be determined. Here, we describe a novel TOR1AIP1 patient manifesting childhood-onset muscle weakness and contractures, and we provide clinical, histological, ultrastructural, and genetic data. We demonstrate that the identified TOR1AIP1 frameshift mutation leads to the selective loss of the LAP1B isoform, while the expression of LAP1C was preserved. Through comparative review of all previously reported TOR1AIP1 cases, we delineate a genotype/phenotype correlation and conclude that LAP1B-specific mutations cause a progressive skeletal muscle phenotype, while mutations involving a loss of both LAP1B and LAP1C isoforms induce a syndromic disorder affecting skeletal muscle, brain, eyes, ear, skin, and bones.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/genética , Mutación/genética , Membrana Nuclear/genética , Isoformas de Proteínas/genética , Niño , Femenino , Mutación del Sistema de Lectura/genética , Humanos , Masculino , Músculos/metabolismo , Músculos/patología , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestructura , Proteínas Nucleares/genética , Fenotipo
12.
Int J Mol Sci ; 22(20)2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34681951

RESUMEN

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.


Asunto(s)
Proteínas de la Membrana/metabolismo , Mutación , Neoplasias/patología , Proteínas Nucleares/metabolismo , Animales , Humanos , Proteínas de la Membrana/genética , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética
13.
Int J Mol Sci ; 22(13)2021 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-34206382

RESUMEN

Emerin is the inner nuclear membrane protein involved in maintaining the mechanical integrity of the nuclear membrane. Mutations in EMD encoding emerin cause Emery-Dreifuss muscular dystrophy (EDMD). Evidence is accumulating that emerin regulation of specific gene expression is associated with this disease, but the exact function of emerin has not been fully elucidated. Here, we show that emerin downregulates Signal transducer and activators of transcription 3 (STAT3) signaling, activated exclusively by Janus kinase (JAK). Deletion mutation experiments show that the lamin-binding domain of emerin is essential for the inhibition of STAT3 signaling. Emerin interacts directly and co-localizes with STAT3 in the nuclear membrane. Emerin knockdown induces STAT3 target genes Bcl2 and Survivin to increase cell survival signals and suppress hydrogen peroxide-induced cell death in HeLa cells. Specifically, downregulation of BAF or lamin A/C increases STAT3 signaling, suggesting that correct-localized emerin, by assembling with BAF and lamin A/C, acts as an intrinsic inhibitor against STAT3 signaling. In C2C12 cells, emerin knockdown induces STAT3 target gene, Pax7, and activated abnormal myoblast proliferation associated with muscle wasting in skeletal muscle homeostasis. Our results indicate that emerin downregulates STAT3 signaling by inducing retention of STAT3 and delaying STAT3 signaling in the nuclear membrane. This mechanism provides clues to the etiology of emerin-related muscular dystrophy and may be a new therapeutic target for treatment.


Asunto(s)
Proteínas de la Membrana/metabolismo , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células A549 , Núcleo Celular/metabolismo , Supervivencia Celular , Regulación de la Expresión Génica , Humanos , Quinasas Janus/metabolismo , Músculo Esquelético/metabolismo , Distrofias Musculares/etiología , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Mutación , Unión Proteica , Factor de Transcripción STAT3/genética , Factores de Transcripción/metabolismo
14.
J Biol Chem ; 294(44): 16241-16254, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31519755

RESUMEN

Vesicle-associated membrane protein-associated protein B (VAPB) is a tail-anchored protein that is present at several contact sites of the endoplasmic reticulum (ER). We now show by immunoelectron microscopy that VAPB also localizes to the inner nuclear membrane (INM). Using a modified enhanced ascorbate peroxidase 2 (APEX2) approach with rapamycin-dependent targeting of the peroxidase to a protein of interest, we searched for proteins that are in close proximity to VAPB, particularly at the INM. In combination with stable isotope labeling with amino acids in cell culture (SILAC), we confirmed many well-known interaction partners at the level of the ER with a clear distinction between specific and nonspecific hits. Furthermore, we identified emerin, TMEM43, and ELYS as potential interaction partners of VAPB at the INM and the nuclear pore complex, respectively.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas/metabolismo , Enzimas Multifuncionales/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Proteínas de Unión al ADN/metabolismo , Retículo Endoplásmico/metabolismo , Células HeLa , Humanos , Marcaje Isotópico , Proteínas de la Membrana/metabolismo , Microscopía Inmunoelectrónica/métodos , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica , Mapeo de Interacción de Proteínas/métodos , Mapas de Interacción de Proteínas , Transporte de Proteínas , Proteómica , Sirolimus/metabolismo , Factores de Transcripción/metabolismo
15.
Muscle Nerve ; 61(4): 436-448, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31840275

RESUMEN

Emery-Dreifuss muscular dystrophy (EDMD) is a rare muscular dystrophy, but is particularly important to diagnose due to frequent life-threatening cardiac complications. EDMD classically presents with muscle weakness, early contractures, cardiac conduction abnormalities and cardiomyopathy, although the presence and severity of these manifestations vary by subtype and individual. Associated genes include EMD, LMNA, SYNE1, SYNE2, FHL1, TMEM43, SUN1, SUN2, and TTN, encoding emerin, lamin A/C, nesprin-1, nesprin-2, FHL1, LUMA, SUN1, SUN2, and titin, respectively. The Online Mendelian Inheritance in Man database recognizes subtypes 1 through 7, which captures most but not all of the associated genes. Genetic diagnosis is essential whenever available, but traditional diagnostic tools can help steer the evaluation toward EDMD and assist with interpretation of equivocal genetic test results. Management is primarily supportive, but it is important to monitor patients closely, especially for potential cardiac complications. There is a high potential for progress in the treatment of EDMD in the coming years.


Asunto(s)
Músculo Esquelético/patología , Distrofia Muscular de Emery-Dreifuss/diagnóstico , Bases de Datos Genéticas , Humanos , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/patología
16.
Muscle Nerve ; 62(1): 128-136, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32304242

RESUMEN

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.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Histona Acetiltransferasas/antagonistas & inhibidores , Distrofia Muscular de Emery-Dreifuss/tratamiento farmacológico , Distrofia Muscular de Emery-Dreifuss/patología , Células Madre/efectos de los fármacos , Tiazoles/uso terapéutico , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Histona Acetiltransferasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre/patología , Tiazoles/farmacología
17.
Int J Mol Sci ; 21(17)2020 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-32824881

RESUMEN

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.


Asunto(s)
Distroglicanos/metabolismo , Proteínas de la Membrana/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Proteínas Nucleares/metabolismo , Transporte Activo de Núcleo Celular , Animales , Linfocitos B/metabolismo , Sitios de Unión , Línea Celular , Células Cultivadas , Distroglicanos/química , Distroglicanos/genética , Células HeLa , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Distrofia Muscular de Emery-Dreifuss/genética , Mutación , Membrana Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Unión Proteica
18.
J Cell Sci ; 130(13): 2111-2118, 2017 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-28576971

RESUMEN

The complex comprising serum response factor (SRF) and megakaryoblastic leukemia 1 protein (Mkl1) promotes myofibroblast differentiation during wound healing. SRF-Mkl1 is sensitive to the mechanical properties of the extracellular environment; but how cells sense and transduce mechanical cues to modulate SRF-Mkl1-dependent gene expression is not well understood. Here, we demonstrate that the nuclear lamina-associated inner nuclear membrane protein Emerin stimulates SRF-Mkl1-dependent gene activity in a substrate stiffness-dependent manner. Specifically, Emerin was required for Mkl1 nuclear accumulation and maximal SRF-Mkl1-dependent gene expression in response to serum stimulation of cells grown on stiff substrates but was dispensable on more compliant substrates. Focal adhesion area was also reduced in cells lacking Emerin, consistent with a role for Emerin in sensing substrate stiffness. Expression of a constitutively active form of Mkl1 bypassed the requirement for Emerin in SRF-Mkl1-dependent gene expression and reversed the focal adhesion defects evident in EmdKO fibroblasts. Together, these data indicate that Emerin, a conserved nuclear lamina protein, couples extracellular matrix mechanics and SRF-Mkl1-dependent transcription.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas Nucleares/genética , Factor de Respuesta Sérica/genética , Transactivadores/genética , Cicatrización de Heridas/genética , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Adhesiones Focales/genética , Regulación de la Expresión Génica/genética , Humanos , Ratones , Ratones Noqueados , Miofibroblastos/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Lámina Nuclear/genética , Transducción de Señal/genética
19.
Proc Natl Acad Sci U S A ; 113(19): 5293-8, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27114541

RESUMEN

Cyclic stretch is an important inducer of vascular smooth muscle cell (VSMC) proliferation, which is crucial in vascular remodeling during hypertension. However, the molecular mechanism remains unclear. We studied the effects of emerin and lamin A/C, two important nuclear envelope proteins, on VSMC proliferation in hypertension and the underlying mechano-mechanisms. In common carotid artery of hypertensive rats in vivo and in cultured cells subjected to high (15%) cyclic stretch in vitro, VSMC proliferation was increased significantly, and the expression of emerin and lamin A/C was repressed compared with normotensive or normal (5%) cyclic stretch controls. Using targeted siRNA to mimic the repressed expression of emerin or lamin A/C induced by 15% stretch, we found that VSMC proliferation was enhanced under static and 5%-stretch conditions. Overexpression of emerin or lamin A/C reversed VSMC proliferation induced by 15% stretch. Hence, emerin and lamin A/C play critical roles in suppressing VSMC hyperproliferation induced by hyperstretch. ChIP-on-chip and MOTIF analyses showed that the DNAs binding with emerin contain three transcription factor motifs: CCNGGA, CCMGCC, and ABTTCCG; DNAs binding with lamin A/C contain the motifs CVGGAA, GCCGCYGC, and DAAGAAA. Protein/DNA array proved that altered emerin or lamin A/C expression modulated the activation of various transcription factors. Furthermore, accelerating local expression of emerin or lamin A/C reversed cell proliferation in the carotid artery of hypertensive rats in vivo. Our findings establish the pathogenetic role of emerin and lamin A/C repression in stretch-induced VSMC proliferation and suggest mechanobiological mechanism underlying this process that involves the sequence-specific binding of emerin and lamin A/C to specific transcription factor motifs.


Asunto(s)
Proliferación Celular/fisiología , Lamina Tipo A/metabolismo , Mecanotransducción Celular/fisiología , Proteínas de la Membrana/metabolismo , Miocitos del Músculo Liso/fisiología , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Animales , Células Cultivadas , Masculino , Músculo Liso Vascular/citología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/citología , Ratas , Ratas Sprague-Dawley , Estrés Mecánico , Resistencia a la Tracción/fisiología
20.
Biochim Biophys Acta Mol Cell Res ; 1864(2): 303-313, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27865926

RESUMEN

Emerin is an inner nuclear membrane protein that is involved in maintaining the mechanical integrity of the nuclear membrane. Increasing evidence supports the involvement of emerin in the regulation of gene expression; however, its precise function remains to be elucidated. Here, we show that emerin downregulated genes downstream of Notch signaling, which are activated exclusively by the Notch intracellular domain (NICD). Deletion mutant experiments revealed that the transmembrane domain of emerin is important for the inhibition of Notch signaling. Emerin interacted directly and colocalized with the NICD at the nuclear membrane. Emerin knockdown induced the phosphorylation of ERK and AKT, increased endogenous Notch signaling, and inhibited hydrogen peroxide-induced apoptosis in HeLa cells. Notably, the downregulation of barrier-to-autointegration factor (BAF) or lamin A/C increased Notch signaling by inducing the release of emerin into the cytosol, implying that nuclear membrane-bound emerin acts as an endogenous inhibitor of Notch signaling. Taken together, our results indicate that emerin negatively regulates Notch signaling by promoting the retention of the NICD at the nuclear membrane. This mechanism could constitute a new therapeutic target for the treatment of emerin-related diseases.


Asunto(s)
Proteínas de la Membrana/fisiología , Membrana Nuclear/metabolismo , Proteínas Nucleares/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Supervivencia Celular , Regulación de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Proteínas de la Membrana/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda