Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 134
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 152(3): 584-98, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-23374351

RESUMO

Eukaryotic cells have a layer of heterochromatin at the nuclear periphery. To investigate mechanisms regulating chromatin distribution, we analyzed heterochromatin organization in different tissues and species, including mice with mutations in the lamin B receptor (Lbr) and lamin A (Lmna) genes that encode nuclear envelope (NE) proteins. We identified LBR- and lamin-A/C-dependent mechanisms tethering heterochromatin to the NE. The two tethers are sequentially used during cellular differentiation and development: first the LBR- and then the lamin-A/C-dependent tether. The absence of both LBR and lamin A/C leads to loss of peripheral heterochromatin and an inverted architecture with heterochromatin localizing to the nuclear interior. Myoblast transcriptome analyses indicated that selective disruption of the LBR- or lamin-A-dependent heterochromatin tethers have opposite effects on muscle gene expression, either increasing or decreasing, respectively. These results show how changes in NE composition contribute to regulating heterochromatin positioning, gene expression, and cellular differentiation during development.


Assuntos
Heterocromatina/metabolismo , Lamina Tipo A/metabolismo , Desenvolvimento Muscular , Mioblastos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Perfilação da Expressão Gênica , Camundongos , Mioblastos/citologia , Membrana Nuclear/metabolismo , Receptor de Lamina B
2.
Cell ; 149(3): 565-77, 2012 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-22541428

RESUMO

Human LMNA gene mutations result in laminopathies that include Emery-Dreifuss muscular dystrophy (AD-EDMD) and Hutchinson-Gilford progeria, the premature aging syndrome (HGPS). The Lmna null (Lmna(-/-)) and progeroid LmnaΔ9 mutant mice are models for AD-EDMD and HGPS, respectively. Both animals develop severe tissue pathologies with abbreviated life spans. Like HGPS cells, Lmna(-/-) and LmnaΔ9 fibroblasts have typically misshapen nuclei. Unexpectedly, Lmna(-/-) or LmnaΔ9 mice that are also deficient for the inner nuclear membrane protein Sun1 show markedly reduced tissue pathologies and enhanced longevity. Concordantly, reduction of SUN1 overaccumulation in LMNA mutant fibroblasts and in cells derived from HGPS patients corrected nuclear defects and cellular senescence. Collectively, these findings implicate Sun1 protein accumulation as a common pathogenic event in Lmna(-/-), LmnaΔ9, and HGPS disorders.


Assuntos
Lamina Tipo A/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Distrofia Muscular de Emery-Dreifuss/metabolismo , Distrofia Muscular de Emery-Dreifuss/patologia , Proteínas Nucleares/metabolismo , Progéria/metabolismo , Animais , Linhagem Celular , Senescência Celular , Modelos Animais de Doenças , Fibroblastos/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Progéria/patologia
3.
PLoS Biol ; 21(5): e3001746, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37134077

RESUMO

Extramedullary hematopoiesis (EMH) expands hematopoietic capacity outside of the bone marrow in response to inflammatory conditions, including infections and cancer. Because of its inducible nature, EMH offers a unique opportunity to study the interaction between hematopoietic stem and progenitor cells (HSPCs) and their niche. In cancer patients, the spleen frequently serves as an EMH organ and provides myeloid cells that may worsen pathology. Here, we examined the relationship between HSPCs and their splenic niche in EMH in a mouse breast cancer model. We identify tumor produced IL-1α and leukemia inhibitory factor (LIF) acting on splenic HSPCs and splenic niche cells, respectively. IL-1α induced TNFα expression in splenic HSPCs, which then activated splenic niche activity, while LIF induced proliferation of splenic niche cells. IL-1α and LIF display cooperative effects in activating EMH and are both up-regulated in some human cancers. Together, these data expand avenues for developing niche-directed therapies and further exploring EMH accompanying inflammatory pathologies like cancer.


Assuntos
Doenças Hematológicas , Hematopoese Extramedular , Neoplasias , Humanos , Animais , Camundongos , Hematopoese Extramedular/fisiologia , Fator Inibidor de Leucemia/farmacologia , Interleucina-1alfa/farmacologia , Hematopoese
4.
Hum Mol Genet ; 32(2): 177-191, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-35925868

RESUMO

Mutations in LMNA, the gene encoding A-type lamins, cause laminopathies-diseases of striated muscle and other tissues. The aetiology of laminopathies has been attributed to perturbation of chromatin organization or structural weakening of the nuclear envelope (NE) such that the nucleus becomes more prone to mechanical damage. The latter model requires a conduit for force transmission to the nucleus. NE-associated Linker of Nucleoskeleton and Cytoskeleton (LINC) complexes are one such pathway. Using clustered regularly interspaced short palindromic repeats to disrupt the Nesprin-1 KASH (Klarsicht, ANC-1, Syne Homology) domain, we identified this LINC complex protein as the predominant NE anchor for microtubule cytoskeleton components, including nucleation activities and motor complexes, in mouse cardiomyocytes. Loss of Nesprin-1 LINC complexes resulted in loss of microtubule cytoskeleton proteins at the nucleus and changes in nuclear morphology and positioning in striated muscle cells, but with no overt physiological defects. Disrupting the KASH domain of Nesprin-1 suppresses Lmna-linked cardiac pathology, likely by reducing microtubule cytoskeleton activities at the nucleus. Nesprin-1 LINC complexes thus represent a potential therapeutic target for striated muscle laminopathies.


Assuntos
Laminopatias , Músculo Estriado , Animais , Camundongos , Proteínas dos Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Membrana/genética , Citoesqueleto/genética , Citoesqueleto/metabolismo , Matriz Nuclear/genética , Microtúbulos/metabolismo , Membrana Nuclear/genética , Membrana Nuclear/metabolismo , Proteínas de Filamentos Intermediários/metabolismo , Músculo Estriado/metabolismo , Laminopatias/metabolismo
5.
Nat Rev Mol Cell Biol ; 14(1): 13-24, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23212477

RESUMO

The nuclear lamina is an important structural determinant for the nuclear envelope as a whole, attaching chromatin domains to the nuclear periphery and localizing some nuclear envelope proteins. The major components of the lamina are the A-type and B-type lamins, which are members of the intermediate filament protein family. Whereas the expression of A-type lamins is developmentally regulated, B-type lamins, as a class, are found in all cells. The association of B-type lamins with many aspects of nuclear function has led to the view that these are essential proteins, and there is growing evidence suggesting that they regulate cellular senescence. However, B-type lamins are dispensable in certain cell types in vivo, and neither A-type nor B-type lamins may be required in early embryos or embryonic stem cells. The picture that is beginning to emerge is of a complex network of interactions at the nuclear periphery that may be defined by cell- and tissue-specific functions.


Assuntos
Senescência Celular , Lâmina Nuclear/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Senilidade Prematura/genética , Animais , Osso e Ossos/anormalidades , Núcleo Celular/genética , Núcleo Celular/metabolismo , Senescência Celular/genética , Cromatina , Células-Tronco Embrionárias , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Filamentos Intermediários/genética , Filamentos Intermediários/metabolismo , Camundongos , Distrofia Muscular Animal/genética , Mutação , Lâmina Nuclear/genética
6.
Hum Mol Genet ; 31(18): 3161-3180, 2022 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-35567414

RESUMO

RTL1/PEG11, which has been associated with anxiety disorders, is a retrotransposon-derived imprinted gene in the placenta. However, imprinting patterns and functions of RTL1 in the brain have not been well-investigated. We found Rtl1 was paternally, but not maternally, expressed in brain stem, thalamus, and hypothalamus of mice, and imprinting status of RTL1 was maintained in human brain. Paternal Rtl1 knockout (Rtl1m+/p-) mice had higher neonatal death rates due to impaired suckling, and low body weights beginning on embryonic day 16.5. High paternal expression of Rtl1 was detected in the locus coeruleus (LC) and Rtl1m+/p- mice showed an increased delay in time of onset for action potentials and inward currents with decreased neuronal excitability of LC neurons. Importantly, Rtl1m+/p- mice exhibited behaviors associated with anxiety, depression, fear-related learning and memory, social dominance, and low locomotor activity. Taken together, our findings demonstrate RTL1 is imprinted in brain, mediates emotional and social behaviors, and regulates excitability in LC neurons.


Assuntos
Proteínas da Gravidez , Retroelementos , Animais , Ansiedade/genética , Transtornos de Ansiedade/genética , Feminino , Impressão Genômica , Humanos , Locus Cerúleo/metabolismo , Camundongos , Neurônios/metabolismo , Gravidez , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Comportamento Social
7.
PLoS Genet ; 17(8): e1009757, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34449766

RESUMO

To complete mitosis, the bridge that links the two daughter cells needs to be cleaved. This step is carried out by the endosomal sorting complex required for transport (ESCRT) machinery. AKTIP, a protein discovered to be associated with telomeres and the nuclear membrane in interphase cells, shares sequence similarities with the ESCRT I component TSG101. Here we present evidence that during mitosis AKTIP is part of the ESCRT machinery at the midbody. AKTIP interacts with the ESCRT I subunit VPS28 and forms a circular supra-structure at the midbody, in close proximity with TSG101 and VPS28 and adjacent to the members of the ESCRT III module CHMP2A, CHMP4B and IST1. Mechanistically, the recruitment of AKTIP is dependent on MKLP1 and independent of CEP55. AKTIP and TSG101 are needed together for the recruitment of the ESCRT III subunit CHMP4B and in parallel for the recruitment of IST1. Alone, the reduction of AKTIP impinges on IST1 and causes multinucleation. Our data altogether reveal that AKTIP is a component of the ESCRT I module and functions in the recruitment of ESCRT III components required for abscission.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Mitose/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Proteínas Reguladoras de Apoptose/fisiologia , Proteínas de Ciclo Celular/metabolismo , Citocinese , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte/genética , Células HeLa , Humanos , Transporte Proteico , Fuso Acromático/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
8.
Annu Rev Genomics Hum Genet ; 21: 263-288, 2020 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-32428417

RESUMO

In recent years, our perspective on the cell nucleus has evolved from the view that it is a passive but permeable storage organelle housing the cell's genetic material to an understanding that it is in fact a highly organized, integrative, and dynamic regulatory hub. In particular, the subcompartment at the nuclear periphery, comprising the nuclear envelope and the underlying lamina, is now known to be a critical nexus in the regulation of chromatin organization, transcriptional output, biochemical and mechanosignaling pathways, and, more recently, cytoskeletal organization. We review the various functional roles of the nuclear periphery and their deregulation in diseases of the nuclear envelope, specifically the laminopathies, which, despite their rarity, provide insights into contemporary health-care issues.


Assuntos
Núcleo Celular/genética , Cromatina/química , Cromatina/genética , Laminopatias/patologia , Animais , Humanos , Laminopatias/genética
9.
Hum Mol Genet ; 28(15): 2486-2500, 2019 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31009944

RESUMO

Mutations in LMNA encoding lamin A/C and EMD encoding emerin cause cardiomyopathy and muscular dystrophy. Lmna null mice develop these disorders and have a lifespan of 7-8 weeks. Emd null mice show no overt pathology and have normal skeletal muscle but with regeneration defects. We generated mice with germline deletions of both Lmna and Emd to determine the effects of combined loss of the encoded proteins. Mice without lamin A/C and emerin are born at the expected Mendelian ratio, are grossly normal at birth but have shorter lifespans than those lacking only lamin A/C. However, there are no major differences between these mice with regards to left ventricular function, heart ultrastructure or electrocardiographic parameters except for slower heart rates in the mice lacking both lamin A/C and emerin. Skeletal muscle is similarly affected in both of these mice. Lmna+/- mice also lacking emerin live to at least 1 year and have no significant differences in growth, heart or skeletal muscle compared to Lmna+/- mice. Deletion of the mouse gene encoding lamina-associated protein 1 leads to prenatal death; however, mice with heterozygous deletion of this gene lacking both lamin A/C and emerin are born at the expected Mendelian ratio but had a shorter lifespan than those only lacking lamin A/C and emerin. These results show that mice with combined deficiencies of three interacting nuclear envelope proteins have normal embryonic development and that early postnatal defects are primarily driven by loss of lamin A/C or lamina-associated polypeptide 1 rather than emerin.


Assuntos
Proteínas de Transporte/genética , Coração/fisiopatologia , Lamina Tipo A/genética , Proteínas de Membrana/genética , Músculo Esquelético/fisiopatologia , Distrofia Muscular de Emery-Dreifuss/genética , Mutação , Proteínas Nucleares/genética , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Haploinsuficiência , Masculino , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Distrofia Muscular de Emery-Dreifuss/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia
10.
Gastroenterology ; 154(6): 1625-1629.e8, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29366840

RESUMO

Lamins have important roles in nuclear structure and cell signaling. Several diseases are associated with mutations in the lamin A/C gene (LMNA in humans). Patients with familial partial lipodystrophy caused by LMNA mutations develop pancreatitis, but lamin function in the pancreas and how these mutations affect pancreatic regulation are unknown. We generated mice with inducible exocrine pancreas-specific disruption of Lmna and showed that LMNA is lost from most exocrine pancreas cells. LMNA-knockout pancreata develop endoplasmic reticulum stress with loss of acinar cell markers, increased autophagy, apoptosis, and cell proliferation, compared to CreERT2- mice (littermate controls). Disruption of Lmna led to a phenotype that resembled chronic pancreatitis, with increased Sirius Red staining and α-smooth muscle actin in male LMNA-knockout mice compared to littermate males, but not in female mice. LMNA-knockout pancreata have reduced levels of RB and activation of E2F, based on increased expression of E2F target genes. Therefore, lamins maintain pancreatic homeostasis by regulating RB stability and E2F activity.


Assuntos
Fatores de Transcrição E2F/fisiologia , Homeostase/genética , Lamina Tipo A/fisiologia , Pâncreas Exócrino/metabolismo , Proteína do Retinoblastoma/fisiologia , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transdução de Sinais/genética
11.
Development ; 142(14): 2425-30, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26138477

RESUMO

The paternally expressed imprinted retrotransposon-like 1 (Rtl1) is a retrotransposon-derived gene that has evolved a function in eutherian placentation. Seven miRNAs, including miR-127, are processed from a maternally expressed antisense Rtl1 transcript (Rtl1as) and regulate Rtl1 levels through RNAi-mediated post-transcriptional degradation. To determine the relative functional role of Rtl1as miRNAs in Rtl1 dosage, we generated a mouse specifically deleted for miR-127. The miR-127 knockout mice exhibit placentomegaly with specific defects within the labyrinthine zone involved in maternal-fetal nutrient transfer. Although fetal weight is unaltered, specific Rtl1 transcripts and protein levels are increased in both the fetus and placenta. Phenotypic analysis of single (ΔmiR-127/Rtl1 or miR-127/ΔRtl1) and double (ΔmiR-127/ΔRtl1) heterozygous miR-127- and Rtl1-deficient mice indicate that Rtl1 is the main target gene of miR-127 in placental development. Our results demonstrate that miR-127 is an essential regulator of Rtl1, mediated by a trans-homologue interaction between reciprocally imprinted genes on the maternally and paternally inherited chromosomes.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Placenta/fisiologia , Proteínas da Gravidez/metabolismo , Animais , Cromossomos/metabolismo , Cromossomos/ultraestrutura , Cruzamentos Genéticos , Éxons , Feminino , Deleção de Genes , Impressão Genômica , Heterozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Família Multigênica , Fenótipo , Placenta/metabolismo , Placentação/genética , Gravidez , Interferência de RNA
12.
Differentiation ; 94: 58-70, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28056360

RESUMO

Peripheral heterochromatin in mammalian nuclei is tethered to the nuclear envelope by at least two mechanisms here referred to as the A- and B-tethers. The A-tether includes lamins A/C and additional unknown components presumably INM protein(s) interacting with both lamins A/C and chromatin. The B-tether includes the inner nuclear membrane (INM) protein Lamin B-receptor, which binds B-type lamins and chromatin. Generally, at least one of the tethers is always present in the nuclear envelope of mammalian cells. Deletion of both causes the loss of peripheral heterochromatin and consequently inversion of the entire nuclear architecture, with this occurring naturally in rod photoreceptors of nocturnal mammals. The tethers are differentially utilized during development, regulate gene expression in opposite manners, and play an important role during cell differentiation. Here we aimed to identify the unknown chromatin binding component(s) of the A-tether. We analyzed 10 mouse tissues by immunostaining with antibodies against 7 INM proteins and found that every cell type has specific, although differentially and developmentally regulated, sets of these proteins. In particular, we found that INM protein LEMD2 is concomitantly expressed with A-type lamins in various cell types but is lacking in inverted nuclei of rod cells. Truncation or deletion of Lmna resulted in the downregulation and mislocalization of LEMD2, suggesting that the two proteins interact and pointing at LEMD2 as a potential chromatin binding mediator of the A-tether. Using nuclei of mouse rods as an experimental model lacking peripheral heterochromatin, we expressed a LEMD2 transgene alone or in combination with lamin C in these cells and observed no restoration of peripheral heterochromatin in either case. We conclude that in contrary to the B-tether, the A-tether has a more intricate composition and consists of multiple components that presumably vary, at differing degrees of redundancy, between cell types and differentiation stages.


Assuntos
Núcleo Celular/genética , Lamina Tipo A/genética , Proteínas de Membrana/genética , Membrana Nuclear/genética , Proteínas Nucleares/genética , Animais , Diferenciação Celular/genética , Núcleo Celular/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , Lamina Tipo A/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Membrana Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Transgenes
13.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L548-L558, 2017 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-28522567

RESUMO

In bacterial pneumonia, lung damage resulting from epithelial cell injury is a major contributor to the severity of disease and, in some cases, can lead to long-term sequelae, especially in the setting of severe lung injury or acute respiratory distress syndrome. Leukemia inhibitory factor (LIF), a member of the IL-6 cytokine family, is a critical determinant of lung tissue protection during pneumonia, but the cellular sources of LIF and the signaling pathways leading to its production in the infected lung are not known. Here, we demonstrate that lung epithelium, specifically alveolar type II cells, is the predominant site of LIF transcript induction in pneumonic mouse lungs. Epithelial cell cultures were induced to express LIF by bacteria and by sterile bronchoalveolar lavage fluid from pneumonic mice. Reciprocal bone marrow chimera studies demonstrated that LIF deficiency in the nonhematopoietic compartment, but not LIF deficiency in hematopoietic cells, eliminated LIF induction during pneumonia. Although NF-κB RelA (p65) is essential for the expression of many cytokines during pneumonia, its targeted mutation in the lung epithelium was inconsequential for pneumonia-driven LIF induction. However, maximal expression of this epithelial-derived cytokine was dependent on NF-κB RelA in myeloid cells. Overall, our data suggest a signaling axis whereby activation of NF-κB RelA in myeloid cells promotes epithelial LIF induction during lung infections, representing a means through which these two cell types collaborate to improve tissue resilience during pneumonia.


Assuntos
Células Epiteliais/metabolismo , Fator Inibidor de Leucemia/biossíntese , Células Mieloides/metabolismo , Pneumonia Bacteriana/metabolismo , Células Epiteliais Alveolares/metabolismo , Células Epiteliais Alveolares/patologia , Animais , Líquido da Lavagem Broncoalveolar , Linhagem Celular , Escherichia coli/fisiologia , Feminino , Hematopoese , Fator Inibidor de Leucemia/genética , Pulmão/metabolismo , Pulmão/patologia , Macrófagos Alveolares/metabolismo , Masculino , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumonia Bacteriana/microbiologia , Pneumonia Bacteriana/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição RelA/metabolismo
14.
Differentiation ; 91(4-5): 42-9, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26651425

RESUMO

Embryo implantation, a key critical feature of mammalian pregnancy, involves co-ordinate interplay between an incoming blastocyst and a receptive uterus. Aberrations in signaling cascades during this process result in pregnancy loss in mammals, including women. Analysis of the complete uterus at any given point either during preparation for implantation or during and after embryo attachment and invasion makes it difficult to assign specific signaling mechanism to the individual cellular compartments of the uterus. Here, we describe methods for the specific isolation of the luminal epithelium (LE) and subsequent analysis of gene expression/signaling pathways during embryo attachment. We further describe the analysis of RNA and proteins by specific techniques of quantitative PCR (qPCR), immunostaining and Western blotting of uterine tissues. These methods can be applied to the other cellular compartments of the uterus and embryo invasion and endometrial development. These techniques will be beneficial to investigators for delineating the mechanisms involved during embryo attachment and female reproduction as well as providing a means to studying highly dynamic changes in gene expression in tissues.


Assuntos
Implantação do Embrião/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Biossíntese de Proteínas/genética , Útero/crescimento & desenvolvimento , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Feminino , Camundongos , Gravidez , RNA Mensageiro/biossíntese , Transdução de Sinais/genética , Útero/metabolismo
15.
Dev Biol ; 407(2): 321-30, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26417726

RESUMO

One of the more dramatic examples of cellular reorganization occurs during spermiogenesis in which a roughly spherical spermatid is transformed into a mature sperm cell. A highlight of this process involves nuclear remodeling whereby the round spermatid nucleus is sculpted into an elongated and polar structure. This transformation in nuclear architecture features chromatin condensation, changes in the composition and organization of the nuclear lamina and redistribution and elimination of nuclear pore complexes. The manchette, a cytoplasmic microtubule-based structure is thought to play a crucial role in the remodeling process. Here we show that SUN4, a spermatid nuclear membrane protein has an essential function in coupling the manchette to the nuclear periphery. In the absence of SUN4, manchette microtubules appear highly disorganized and the nucleus itself fails to elongate. Consequently, mice deficient in SUN4 display globozoospermia with associated infertility.


Assuntos
Núcleo Celular/metabolismo , Mamíferos/metabolismo , Proteínas Nucleares/metabolismo , Espermatogênese , Animais , Núcleo Celular/ultraestrutura , Feminino , Imunofluorescência , Deleção de Genes , Células HeLa , Recombinação Homóloga/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/metabolismo , Membrana Nuclear/metabolismo , Membrana Nuclear/ultraestrutura , Ligação Proteica , Isoformas de Proteínas/metabolismo , Espermátides/metabolismo , Testículo/metabolismo , Testículo/ultraestrutura
16.
Differentiation ; 89(1-2): 11-21, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25578479

RESUMO

The nuclear lamina, comprised of the A and B-type lamins, is important in maintaining nuclear shape and in regulating key nuclear functions such as chromatin organization and transcription. Deletion of the A-type lamins results in genome instability and many cancers show altered levels of A-type lamin expression. Loss of function mutations in the mouse Lmna gene result in early postnatal lethality, usually within 3-5 weeks of birth making an analysis of the role of lamins in carcinogenesis difficult. To circumvent early lethality, and determine the role of the A-type lamins in specific tissues in older mice we derived a conditional allele of Lmna(FL/FL) (floxed). Lmna(FL/FL) was specifically deleted in the gastrointestinal (GI) epithelium by crossing the Lmna(FL/FL) mice with Villin-Cre mice. Mice lacking Lmna in the GI are overtly normal with no effects on overall growth, longevity or GI morphology. On a GI specific sensitized (Apc(Min/+)) background, polyp numbers are unchanged, but polyp size is slightly increased, and only in the duodenum. Our findings reveal that although A-type lamins are dispensable in the postnatal GI epithelium, loss of Lmna under malignant conditions may, to a limited extent, enhance polyp size indicating that A-type lamins may regulate cell proliferation in the transformed GI epithelium.


Assuntos
Transformação Celular Neoplásica/genética , Instabilidade Genômica , Pólipos Intestinais/genética , Lamina Tipo A/genética , Animais , Proliferação de Células/genética , Epitélio/crescimento & desenvolvimento , Epitélio/patologia , Trato Gastrointestinal/crescimento & desenvolvimento , Trato Gastrointestinal/patologia , Pólipos Intestinais/patologia , Lamina Tipo A/metabolismo , Camundongos , Especificidade de Órgãos
17.
Dev Biol ; 385(2): 155-9, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24269765

RESUMO

Primordial germ cells (PGCs) are the founder cells of the germline. Via gametogenesis and fertilisation this lineage generates a new embryo in the next generation. PGCs are also the cell of origin of multilineage teratocarcinomas. In vitro, mouse PGCs can give rise to embryonic germ (EG) cells - pluripotent stem cells that can contribute to primary chimaeras when introduced into pre-implantation embryos. Thus, PGCs can give rise to pluripotent cells in the course of the developmental cycle, during teratocarcinogenesis and by in vitro culture. However, there is no evidence that PGCs can differentiate directly into somatic cell types. Furthermore, it is generally assumed that PGCs do not contribute to chimaeras following injection into the early mouse embryo. However, these data have never been formally published. Here, we present the primary data from the original PGC-injection experiments performed 40 years ago, alongside results from more recent studies in three separate laboratories. These results have informed and influenced current models of the relationship between pluripotency and the germline cycle. Current technologies allow further experiments to confirm and expand upon these findings and allow definitive conclusions as to the developmental potency of PGCs.


Assuntos
Embrião de Mamíferos/citologia , Células Germinativas/citologia , Animais , Linhagem da Célula , Feminino , Camundongos , Gravidez
18.
Hum Mol Genet ; 22(14): 2852-69, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23535822

RESUMO

Mutations in lamin A/C result in a range of tissue-specific disorders collectively called laminopathies. Of these, Emery-Dreifuss and Limb-Girdle muscular dystrophy 1B mainly affect striated muscle. A useful model for understanding both laminopathies and lamin A/C function is the Lmna(-/-) mouse. We found that skeletal muscle growth and muscle satellite (stem) cell proliferation were both reduced in Lmna(-/-) mice. Lamins A and C associate with lamina-associated polypeptide 2 alpha (Lap2α) and the retinoblastoma gene product, pRb, to regulate cell cycle exit. We found Lap2α to be upregulated in Lmna(-/-) myoblasts (MBs). To specifically test the contribution of elevated Lap2α to the phenotype of Lmna(-/-) mice, we generated Lmna(-/-)Lap2α(-/-) mice. Lifespan and body mass were increased in Lmna(-/-)Lap2α(-/-) mice compared with Lmna(-/-). Importantly, the satellite cell proliferation defect was rescued, resulting in improved myogenesis. Lmna(-/-) MBs also exhibited increased levels of Smad2/3, which were abnormally distributed in the cell and failed to respond to TGFß1 stimulation as in control cells. However, using SIS3 to inhibit signaling via Smad3 reduced cell death and augmented MB fusion. Together, our results show that perturbed Lap2α/pRb and Smad2/3 signaling are important regulatory pathways mediating defective muscle growth in Lmna(-/-) mice, and that inhibition of either pathway alone or in combination can ameliorate this deleterious phenotype.


Assuntos
Proteínas de Ligação a DNA/deficiência , Lamina Tipo A/deficiência , Proteínas de Membrana/deficiência , Músculo Esquelético/crescimento & desenvolvimento , Distrofia Muscular de Emery-Dreifuss/metabolismo , Animais , Proliferação de Células , Proteínas de Ligação a DNA/genética , Humanos , Lamina Tipo A/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo , Distrofia Muscular de Emery-Dreifuss/genética , Distrofia Muscular de Emery-Dreifuss/fisiopatologia , Mioblastos/metabolismo , Células Satélites de Músculo Esquelético/citologia , Células Satélites de Músculo Esquelético/metabolismo , Proteína Smad2/metabolismo , Proteína Smad3/metabolismo
19.
EMBO J ; 30(16): 3383-96, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21750527

RESUMO

Genomic instability due to telomere dysfunction and defective repair of DNA double-strand breaks (DSBs) is an underlying cause of ageing-related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild-type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A-type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Assuntos
Catepsina L/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Reparo do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Lamina Tipo A/fisiologia , Vitamina D/fisiologia , Animais , Calcitriol/farmacologia , Catepsina L/antagonistas & inibidores , Catepsina L/biossíntese , Catepsina L/genética , Linhagem Celular , Precursores Enzimáticos/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Lamina Tipo A/deficiência , Lamina Tipo A/genética , Leupeptinas/farmacologia , Camundongos , Complexo de Endopeptidases do Proteassoma/metabolismo , Estabilidade Proteica , Proteínas Recombinantes de Fusão/fisiologia , Especificidade da Espécie , Transfecção , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
20.
Lipids Health Dis ; 14: 160, 2015 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-26637310

RESUMO

BACKGROUND: DDX5/p68 RNA helicase is a member of the DEAD (Asp-Glu-Ala-Asp) box proteins. Apart from RNA unwinding, DDX5 is an important transcriptional factor and co-activator in cell proliferation and differentiation. FINDINGS: Here, we have characterised the role of DDX5 in adipogenesis in 3T3-L1 cells using siRNA mediated suppression. Transient inhibition of Ddx5 mRNA expression at the start of adipogenesis impairs the differentiation programme even when DDX5 expression is restored later in adipogenesis. However transient suppression of Ddx5 at the later stages of adipogenesis do not impair adipogenesis or triglyceride accumulation suggesting Ddx5 expression is dispensable in a mature adipocyte. CONCLUSION: These results implicate DDX5 as a crucial factor involved in the complex transcriptional cascade of events that regulate adipogenesis and essential to the initiation of adipogenesis.


Assuntos
Adipogenia , RNA Helicases DEAD-box/metabolismo , Células-Tronco Mesenquimais/enzimologia , Células 3T3-L1 , Adipócitos/enzimologia , Animais , RNA Helicases DEAD-box/genética , Expressão Gênica , Técnicas de Silenciamento de Genes , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Endogâmicos C3H
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA