Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 102021 02 19.
Article in English | MEDLINE | ID: mdl-33605210

ABSTRACT

Lamins form stable filaments at the nuclear periphery in metazoans. Unlike B-type lamins, lamins A and C localize also in the nuclear interior, where they interact with lamin-associated polypeptide 2 alpha (LAP2α). Using antibody labeling, we previously observed a depletion of nucleoplasmic A-type lamins in mouse cells lacking LAP2α. Here, we show that loss of LAP2α actually causes formation of larger, biochemically stable lamin A/C structures in the nuclear interior that are inaccessible to lamin A/C antibodies. While nucleoplasmic lamin A forms from newly expressed pre-lamin A during processing and from soluble mitotic lamins in a LAP2α-independent manner, binding of LAP2α to lamin A/C during interphase inhibits formation of higher order structures, keeping nucleoplasmic lamin A/C in a mobile state independent of lamin A/C S22 phosphorylation. We propose that LAP2α is essential to maintain a mobile lamin A/C pool in the nuclear interior, which is required for proper nuclear functions.


Subject(s)
DNA-Binding Proteins/genetics , Lamin Type A/genetics , Membrane Proteins/genetics , Animals , Cell Line , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Lamin Type A/metabolism , Membrane Proteins/metabolism , Mice
2.
J Cell Sci ; 131(3)2018 02 08.
Article in English | MEDLINE | ID: mdl-29361532

ABSTRACT

A-type lamins are components of the peripheral nuclear lamina but also localize in the nuclear interior in a complex with lamina-associated polypeptide (LAP) 2α. Loss of LAP2α and nucleoplasmic lamins in wild-type cells increases cell proliferation, but in cells expressing progerin (a mutant lamin A that causes Hutchinson-Gilford progeria syndrome), low LAP2α levels result in proliferation defects. Here, the aim was to understand the molecular mechanism governing how relative levels of LAP2α, progerin and nucleoplasmic lamins affect cell proliferation. Cells from progeria patients and inducible progerin-expressing cells expressing low levels of progerin proliferate faster than wild-type or lamin A-expressing control cells, and ectopic expression of LAP2α impairs proliferation. In contrast, cells expressing high levels of progerin and lacking lamins in the nuclear interior proliferate more slowly, and ectopic LAP2α expression enhances proliferation. However, simultaneous expression of LAP2α and wild-type lamin A or an assembly-deficient lamin A mutant restored the nucleoplasmic lamin A pool in these cells and abolished the growth-promoting effect of LAP2α. Our data show that LAP2α promotes or inhibits proliferation of progeria cells depending on the level of A-type lamins in the nuclear interior.This article has an associated First Person interview with the first author of the paper.


Subject(s)
DNA-Binding Proteins/metabolism , Lamins/metabolism , Membrane Proteins/metabolism , Progeria/metabolism , Progeria/pathology , Cell Cycle , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lamin Type A/metabolism
3.
J Cell Sci ; 130(13): 2087-2096, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28668931

ABSTRACT

Nuclear lamins are components of the peripheral lamina that define the mechanical properties of nuclei and tether heterochromatin to the periphery. A-type lamins localize also to the nuclear interior, but the regulation and specific functions of this nucleoplasmic lamin pool are poorly understood. In this Commentary, we summarize known pathways that are potentially involved in the localization and dynamic behavior of intranuclear lamins, including their post-translational modifications and interactions with nucleoplasmic proteins, such as lamina-associated polypeptide 2α (LAP2α; encoded by TMPO). In addition, new data suggest that lamins in the nuclear interior have an important role in chromatin regulation and gene expression through dynamic binding to both hetero- and euchromatic genomic regions and promoter subdomains, thereby affecting epigenetic pathways and chromatin accessibility. Nucleoplasmic lamins also have a role in spatial chromatin organization and may be involved in mechanosignaling. In view of this newly emerging concept, we propose that the previously reported cellular phenotypes in lamin-linked diseases are, at least in part, rooted in an impaired regulation and/or function of the nucleoplasmic lamin A/C pool.


Subject(s)
Cell Nucleus/genetics , DNA-Binding Proteins/genetics , Lamin Type A/genetics , Lamins/genetics , Membrane Proteins/genetics , Heterochromatin/genetics , Humans , Lamin Type A/metabolism , Lamins/metabolism , Nuclear Lamina/genetics , Nuclear Lamina/metabolism , Protein Processing, Post-Translational/genetics , Signal Transduction
4.
J Virol ; 89(5): 2857-65, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25540383

ABSTRACT

UNLABELLED: Many of the small DNA tumor viruses encode transforming proteins that function by targeting critical cellular pathways involved in cell proliferation and survival. In this study, we have examined whether some of the functions of the polyomavirus small T antigens (ST) are shared by the E6 and E7 oncoproteins of two oncogenic papillomaviruses. Using three different assays, we have found that E7 can provide some simian virus 40 (SV40) or murine polyomavirus (PyV) ST functions. Both human papillomavirus 16 (HPV16) and bovine papillomavirus (BPV1) E7 proteins are capable of partially substituting for SV40 ST in a transformation assay that also includes SV40 large T antigen, the catalytic subunit of cellular telomerase, and oncogenic Ras. Like SV40 ST, HPV16 E7 has the ability to override a quiescence block induced by mitogen deprivation. Like PyV ST, it also has the ability to inhibit myoblast differentiation. At least two of these activities are dependent upon the interaction of HPV16 E7 with retinoblastoma protein family members. For small T antigens, interaction with PP2A is needed for each of these functions. Even though there is no strong evidence that E6 or E7 share the ability of small T to interact with PP2A, E7 provides these functions related to cellular transformation. IMPORTANCE: DNA tumor viruses have provided major insights into how cancers develop. Some viruses, like the human papillomaviruses, can cause cancer directly. Both the papillomaviruses and the polyomaviruses have served as tools for understanding pathways that are often perturbed in cancer. Here, we have compared the functions of transforming proteins from several DNA tumor viruses, including two papillomaviruses and two polyomaviruses. We tested the papillomavirus E6 and E7 oncoproteins in three functional assays and found that E7 can provide some or all of the functions of the SV40 small T antigen, another well-characterized oncoprotein, in two of these assays. In a third assay, papillomavirus E7 has the same effect as the murine polyomavirus small T protein. In summary, we report several new functions for the papillomavirus E7 proteins, which will contribute new insights into the roles of viruses in cancer and the cellular pathways they perturb in carcinogenesis.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Transformation, Viral , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Repressor Proteins/metabolism , Genetic Complementation Test , Human papillomavirus 16/genetics , Human papillomavirus 16/physiology , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/physiology , Simian virus 40/genetics , Simian virus 40/physiology
5.
Mol Cell ; 54(6): 932-945, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24857551

ABSTRACT

Quiescence (G0) allows cycling cells to reversibly cease proliferation. A decision to enter quiescence is suspected of occurring early in G1, before the restriction point (R). Surprisingly, we have identified G2 as an interval during which inhibition of the protein phosphatase PP2A results in failure to exhibit stable quiescence. This effect is accompanied by shortening of the ensuing G1. The PP2A subcomplex required for stable G0 contains the B56γ B subunit. After PP2A inhibition in G2, aberrant overexpression of cyclin E occurs during mitosis and is responsible for overriding quiescence. Strikingly, suppression of Ras signaling re-establishes normal cyclin E levels during M and restores G0. These data point to PP2A-B56γ-driven Ras signaling modulation in G2 as essential for suppressing aberrant cyclin E expression during mitosis and thereby achieving normal G0 control. Thus, G2 is an interval during which the length and growth factor dependence of the next G1 interval are established.


Subject(s)
G1 Phase/genetics , G2 Phase/genetics , Oncogene Protein p21(ras)/genetics , Protein Phosphatase 2/genetics , Resting Phase, Cell Cycle/physiology , Cell Line , Cyclin E/biosynthesis , Humans , MCF-7 Cells , Mitosis/genetics , Protein Subunits/genetics , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics
6.
Genes Dev ; 27(20): 2274-91, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-24142877

ABSTRACT

Endogenous BRCA1 p220 expression peaks in S and G2 when it is activated, and the protein participates in certain key DNA damage responses. In contrast, its expression is markedly reduced in G0/G1. While variations in transcription represent a significant part of p220 expression control, there is at least one other relevant process. We found that a microRNA, miR-545, that is expressed throughout the cell cycle down-modulates endogenous p220 mRNA and protein abundance directly in both G0/G1 and S/G2. When miR-545 function was inhibited by a specific antagomir, endogenous p220 expression increased in G0/G1, and aberrant p220-associated DNA damage responses and de novo DNA strand breaks accumulated. Analogous results were observed upon inhibition of miR-545 function in S/G2. Both sets of antagomir effects were mimicked by infecting cells with a p220 cDNA-encoding adenoviral vector. Thus, strand breaks were a product of p220 overexpression, and their prevention by miR-545 depends on its modulation of p220 expression. Breaks were also dependent on aberrant, overexpressed p220-driven recruitment of RAD51 to either spontaneously arising or mutagen-based DNA damage sites. Hence, when its level is not physiologically maintained, endogenous p220 aberrantly directs at least one DNA repair protein, RAD51, to damage sites, where their action contributes to the development of de novo DNA damage. Thus, like its loss, a surfeit of endogenous p220 function represents a threat to genome integrity.


Subject(s)
BRCA1 Protein/genetics , Cell Cycle/physiology , DNA Damage/genetics , Gene Expression Regulation , BRCA1 Protein/metabolism , Binding Sites , Cell Line, Tumor , DNA Damage/radiation effects , DNA Repair , HEK293 Cells , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Binding , RNA, Messenger/metabolism , Rad51 Recombinase/metabolism , Ultraviolet Rays
7.
Cell Cycle ; 8(10): 1488-93, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19377295

ABSTRACT

Lamins are components of the peripheral nuclear lamina that has important mechanical roles in nuclear architecture. Lamins have increasingly been implicated in nuclear processes, including DNA replication, chromatin organization and transcription. Recent data highlight the specific functions of a small pool of lamina-independent A-type lamins, located throughout the nucleoplasm, in the regulation of cell cycle progression in early tissue progenitor cells. This novel role of nucleoplasmic lamins is likely mediated by their regulatory activity towards the tumor suppressor Retinoblastoma protein. We describe how an interaction partner of A-type lamins, lamina-associated polypeptide 2alpha (LAP2alpha) affects targeting of lamins A and C to the nuclear interior and how this complex may affect cell cycle progression and tissue homeostasis. Finally, we discuss implications of these novel findings for understanding the pathogenesis of laminopathies, a group of human diseases caused by mutations in lamins.


Subject(s)
Cell Nucleus/metabolism , Homeostasis , Lamins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Proliferation , DNA-Binding Proteins/metabolism , Humans , Membrane Proteins/metabolism , Models, Biological , Protein Binding , Retinoblastoma/metabolism
8.
Nat Cell Biol ; 10(11): 1341-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18849980

ABSTRACT

Lamina-associated polypeptide (LAP) 2alpha is a chromatin-associated protein that binds A-type lamins. Mutations in both LAP2alpha and A-type lamins are linked to human diseases called laminopathies, but the molecular mechanisms are poorly understood. The A-type lamin-LAP2alpha complex interacts with and regulates retinoblastoma protein (pRb), but the significance of this interaction in vivo is unknown. Here we address the function of the A-type lamin-LAP2alpha complex with the use of LAP2alpha-deficient mice. We show that LAP2alpha loss causes relocalization of nucleoplasmic A-type lamins to the nuclear envelope and impairs pRb function. This causes inefficient cell-cycle arrest in dense fibroblast cultures and hyperproliferation of epidermal and erythroid progenitor cells in vivo, leading to tissue hyperplasia. Our results support a disease-relevant model in which LAP2alpha defines A-type lamin localization in the nucleoplasm, which in turn affects pRb-mediated regulation of progenitor cell proliferation and differentiation in highly regenerative tissues.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/metabolism , Erythroid Precursor Cells/physiology , Lamin Type A/metabolism , Membrane Proteins/metabolism , Stem Cells/physiology , Animals , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Disease Models, Animal , Epidermal Cells , Lamin Type A/deficiency , Lamin Type A/genetics , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Stem Cells/cytology
9.
J Cell Sci ; 120(Pt 5): 737-47, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17284516

ABSTRACT

Lamina-associated polypeptide 2alpha (LAP2alpha) is a nuclear protein dynamically associating with chromatin during the cell cycle. In addition, LAP2alpha interacts with A-type lamins and retinoblastoma protein and regulates cell cycle progression via the E2F-Rb pathway. Using yeast two-hybrid analysis and three independent in vitro binding assays we identified a new LAP2alpha interaction partner of hitherto unknown functions, which we termed LINT-25. LINT-25 protein levels were upregulated during G1 phase in proliferating cells and upon cell cycle exit in quiescence, senescence and differentiation. Upon cell cycle exit LINT-25 accumulated in heterochromatin foci, and LAP2alpha protein levels were downregulated by proteasomal degradation. Although LAP2alpha was not required for the upregulation and reorganization of LINT-25 during cell cycle exit, transient expression of LINT-25 in proliferating cells caused loss of LAP2alpha and subsequent cell death. Our data show a role of LINT-25 and LAP2alpha during cell cycle exit, in which LINT-25 acts upstream of LAP2alpha.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Lamins/metabolism , Membrane Proteins/genetics , Mice , Microscopy, Fluorescence , Nuclear Proteins/genetics , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...