1
|
Ryan SM, Almassey M, Burch AM, Ngo G, Martin JM, Myers D, Compton D, Archie S, Cross M, Naeger L, Salzman A, Virola‐Iarussi A, Barbee SA, Mortimer NT, Sanyal S, Vrailas‐Mortimer AD. Drosophila p38 MAPK interacts with BAG-3/starvin to regulate age-dependent protein homeostasis. Aging Cell 2021; 20:e13481. [PMID: 34674371 PMCID: PMC8590102 DOI: 10.1111/acel.13481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 08/23/2021] [Accepted: 09/09/2021] [Indexed: 12/25/2022] Open
Abstract
As organisms age, they often accumulate protein aggregates that are thought to be toxic, potentially leading to age‐related diseases. This accumulation of protein aggregates is partially attributed to a failure to maintain protein homeostasis. A variety of genetic factors have been linked to longevity, but how these factors also contribute to protein homeostasis is not completely understood. In order to understand the relationship between aging and protein aggregation, we tested how a gene that regulates lifespan and age‐dependent locomotor behaviors, p38 MAPK (p38Kb), influences protein homeostasis as an organism ages. We find that p38Kb regulates age‐dependent protein aggregation through an interaction with starvin, a regulator of muscle protein homeostasis. Furthermore, we have identified Lamin as an age‐dependent target of p38Kb and starvin.
Collapse
Affiliation(s)
- Sarah M. Ryan
- Department of Biological Sciences University of Denver Denver CO USA
| | - Michael Almassey
- School of Biological Sciences Illinois State University Normal IL USA
| | | | - Gia Ngo
- Department of Biological Sciences University of Denver Denver CO USA
| | - Julia M. Martin
- School of Biological Sciences Illinois State University Normal IL USA
| | - David Myers
- School of Biological Sciences Illinois State University Normal IL USA
| | - Devin Compton
- School of Biological Sciences Illinois State University Normal IL USA
| | - Shira Archie
- School of Biological Sciences Illinois State University Normal IL USA
| | - Megan Cross
- School of Biological Sciences Illinois State University Normal IL USA
| | - Lauren Naeger
- School of Biological Sciences Illinois State University Normal IL USA
| | - Ashley Salzman
- School of Biological Sciences Illinois State University Normal IL USA
| | | | - Scott A. Barbee
- Department of Biological Sciences University of Denver Denver CO USA
| | | | - Subhabrata Sanyal
- Department of Cell Biology Emory University Atlanta GA USA
- Calico San Francisco CA USA
| | - Alysia D. Vrailas‐Mortimer
- Department of Biological Sciences University of Denver Denver CO USA
- School of Biological Sciences Illinois State University Normal IL USA
- Department of Cell Biology Emory University Atlanta GA USA
| |
Collapse
|
2
|
Lamin A/C Mechanotransduction in Laminopathies. Cells 2020; 9:cells9051306. [PMID: 32456328 PMCID: PMC7291067 DOI: 10.3390/cells9051306] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/14/2022] Open
Abstract
Mechanotransduction translates forces into biological responses and regulates cell functionalities. It is implicated in several diseases, including laminopathies which are pathologies associated with mutations in lamins and lamin-associated proteins. These pathologies affect muscle, adipose, bone, nerve, and skin cells and range from muscular dystrophies to accelerated aging. Although the exact mechanisms governing laminopathies and gene expression are still not clear, a strong correlation has been found between cell functionality and nuclear behavior. New theories base on the direct effect of external force on the genome, which is indeed sensitive to the force transduced by the nuclear lamina. Nuclear lamina performs two essential functions in mechanotransduction pathway modulating the nuclear stiffness and governing the chromatin remodeling. Indeed, A-type lamin mutation and deregulation has been found to affect the nuclear response, altering several downstream cellular processes such as mitosis, chromatin organization, DNA replication-transcription, and nuclear structural integrity. In this review, we summarize the recent findings on the molecular composition and architecture of the nuclear lamina, its role in healthy cells and disease regulation. We focus on A-type lamins since this protein family is the most involved in mechanotransduction and laminopathies.
Collapse
|
3
|
Abstract
Active meiotic chromosome movements are a universally conserved feature. They occur at the early stages of prophase of the first meiotic division and support the chromosome pairing process by (1) efficiently installing the synaptonemal complex between homologous chromosomes, (2) discouraging inadvertent chromosome interactions and (3) bringing homologous chromosomes into proximity. Chromosome movements are driven by forces in the cytoplasm, which are passed on to chromosome ends attached to the nuclear periphery by nuclear-membrane-spanning protein modules. In this extra view, we highlight our recent studies into the role of the nuclear lamina during this process to emphasize that it is a highly conserved structure in metazoans. The nuclear lamina forms a rigid proteinaceous network that underlies the inner nuclear membrane to provide stability to the nucleus. Misdemeanors of the nuclear lamina during meiosis has deleterious consequences for the viability and health of the offspring, highlighting the importance of a functional nuclear lamina during this cell cycle stage. Abbreviations: DSB: DNA double strand break; LEM: LAP2, Emerin, MAN1; LINC: LInker of the Nucleoskeleton and Cytoskeleton; RPM: rapid prophase movement; SUN/KASH: Sad1p, UNC-84/Klarsicht, ANC-1, Syne Homology
Collapse
Affiliation(s)
- Dimitra Paouneskou
- a Department of Chromosome Biology, Max F. Perutz Laboratories , University of Vienna , Vienna , Austria
| | - Verena Jantsch
- a Department of Chromosome Biology, Max F. Perutz Laboratories , University of Vienna , Vienna , Austria
| |
Collapse
|
4
|
Perovanovic J, Hoffman EP. Mechanisms of allelic and clinical heterogeneity of lamin A/C phenotypes. Physiol Genomics 2018; 50:694-704. [PMID: 29750601 PMCID: PMC6335092 DOI: 10.1152/physiolgenomics.00128.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mutations in the lamin A/C (LMNA) gene cause a broad range of clinical syndromes that show tissue-restricted abnormalities of post mitotic tissues, such as muscle, nerve, heart, and adipose tissue. Mutations in other nuclear envelope proteins cause clinically overlapping disorders. The majority of mutations are dominant single amino acid changes (toxic protein produced by the single mutant gene), and patients are heterozygous with both normal and abnormal proteins. Experimental support has been provided for different models of cellular pathogenesis in nuclear envelope diseases, including changes in heterochromatin formation at the nuclear membrane (epigenomics), changes in the timing of steps during terminal differentiation of cells, and structural abnormalities of the nuclear membrane. These models are not mutually exclusive and may be important in different cells at different times of development. Recent experiments using fusion proteins of normal and mutant lamin A/C proteins fused to a bacterial adenine methyltransferase (DamID) provided compelling evidence of mutation-specific perturbation of epigenomic imprinting during terminal differentiation. These gain-of-function properties include lineage-specific ineffective genomic silencing during exit from the cell cycle (heterochromatinization), as well as promiscuous initiation of silencing at incorrect places in the genome. To date, these findings have been limited to a few muscular dystrophy and lipodystrophy LMNA mutations but seem shared with a distinct nuclear envelope disease, emerin-deficient muscular dystrophy. The dominant-negative structural model and gain-of-function epigenomic models for distinct LMNA mutations are not mutually exclusive, and it is likely that both models contribute to aspects of the many complex clinical phenotypes observed.
Collapse
Affiliation(s)
- Jelena Perovanovic
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis, Musculoskeletal, and Skin Diseases, National Institutes of Health , Bethesda, Maryland
| | - Eric P Hoffman
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, State University of New York , Binghamton New York
| |
Collapse
|
5
|
Machowska M, Piekarowicz K, Rzepecki R. Regulation of lamin properties and functions: does phosphorylation do it all? Open Biol 2016; 5:rsob.150094. [PMID: 26581574 PMCID: PMC4680568 DOI: 10.1098/rsob.150094] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The main functions of lamins are their mechanical and structural roles as major building blocks of the karyoskeleton. They are also involved in chromatin structure regulation, gene expression, intracellular signalling pathway modulation and development. All essential lamin functions seem to depend on their capacity for assembly or disassembly after the receipt of specific signals, and after specific, selective and precisely regulated interactions through their various domains. Reversible phosphorylation of lamins is crucial for their functions, so it is important to understand how lamin polymerization and interactions are modulated, and which sequences may undergo such modifications. This review combines experimental data with results of our in silico analyses focused on lamin phosphorylation in model organisms to show the presence of evolutionarily conserved sequences and to indicate specific in vivo phosphorylations that affect particular functions.
Collapse
Affiliation(s)
- Magdalena Machowska
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, ul. Fryderyka Joliot-Curie 14a, Wrocław 50-383, Poland
| | - Katarzyna Piekarowicz
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, ul. Fryderyka Joliot-Curie 14a, Wrocław 50-383, Poland
| | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wrocław, ul. Fryderyka Joliot-Curie 14a, Wrocław 50-383, Poland
| |
Collapse
|
6
|
Abstract
The intermediate filament proteins, A- and B-type lamins, form the nuclear lamina scaffold adjacent to the inner nuclear membrane. Lamins also contribute to chromatin regulation and various signaling pathways affecting gene expression. In this review, Osmanagic-Myers et al. focus on the role of nuclear lamins in mechanosensing and also discuss how disease-linked lamin mutants may impair the response of cells to mechanical stimuli and influence the properties of the extracellular matrix. The intermediate filament proteins, A- and B-type lamins, form the nuclear lamina scaffold adjacent to the inner nuclear membrane. B-type lamins confer elasticity, while A-type lamins lend viscosity and stiffness to nuclei. Lamins also contribute to chromatin regulation and various signaling pathways affecting gene expression. The mechanical roles of lamins and their functions in gene regulation are often viewed as independent activities, but recent findings suggest a highly cross-linked and interdependent regulation of these different functions, particularly in mechanosignaling. In this newly emerging concept, lamins act as a “mechanostat” that senses forces from outside and responds to tension by reinforcing the cytoskeleton and the extracellular matrix. A-type lamins, emerin, and the linker of the nucleoskeleton and cytoskeleton (LINC) complex directly transmit forces from the extracellular matrix into the nucleus. These mechanical forces lead to changes in the molecular structure, modification, and assembly state of A-type lamins. This in turn activates a tension-induced “inside-out signaling” through which the nucleus feeds back to the cytoskeleton and the extracellular matrix to balance outside and inside forces. These functions regulate differentiation and may be impaired in lamin-linked diseases, leading to cellular phenotypes, particularly in mechanical load-bearing tissues.
Collapse
|
7
|
Lyakhovetsky R, Gruenbaum Y. Studying lamins in invertebrate models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 773:245-62. [PMID: 24563351 DOI: 10.1007/978-1-4899-8032-8_11] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lamins are nuclear intermediate filament proteins that are conserved in all multicellular animals. Proteins that resemble lamins are also found in unicellular organisms and in plants. Lamins form a proteinaceous meshwork that outlines the nucleoplasmic side of the inner nuclear membrane, while a small fraction of lamin molecules is also present in the nucleoplasm. They provide structural support for the nucleus and help regulate many other nuclear activities. Much of our knowledge on the function of nuclear lamins and their associated proteins comes from studies in invertebrate organisms and specifically in the nematode Caenorhabditis elegans and the fruit fly Drosophila melanogaster. The simpler lamin system and the powerful genetic tools offered by these model organisms greatly promote such studies. Here we provide an overview of recent advances in the biology of invertebrate nuclear lamins, with special emphasis on their assembly, cellular functions and as models for studying the molecular basis underlying the pathology of human heritable diseases caused by mutations in lamins A/C.
Collapse
Affiliation(s)
- Roman Lyakhovetsky
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Givat Ram, Jerusalem, 91904, Israel
| | | |
Collapse
|
8
|
Swift J, Harada T, Buxboim A, Shin JW, Tang HY, Speicher DW, Discher DE. Label-free mass spectrometry exploits dozens of detected peptides to quantify lamins in wildtype and knockdown cells. Nucleus 2013; 4:450-9. [PMID: 24448480 DOI: 10.4161/nucl.27413] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Label-free quantitation and characterization of proteins by mass spectrometry (MS) is now feasible, especially for moderately expressed structural proteins such as lamins that typically yield dozens of tryptic peptides from tissue cells. Using standard cell culture samples, we describe general algorithms for quantitative analysis of peptides identified in liquid chromatography tandem mass spectrometry (LC-MS/MS). The algorithms were foundational to the discovery that the absolute stoichiometry of A-type to B-type lamins scales with tissue stiffness (Swift et al., Science 2013). Isoform dominance helps make sense of why mutations and changes with age of mechanosensitive lamin-A,C only affect "stiff" tissues such as heart, muscle, bone, or even fat, but not brain. A Peak Ratio Fingerprinting (PRF) algorithm is elaborated here through its application to lamin-A,C knockdown. After demonstrating the large dynamic range of PRF using calibrated mixtures of human and mouse lysates, we validate measurements of partial knockdown with standard cell biology analyses using quantitative immunofluorescence and immunoblotting. Optimal sets of MS-detected peptides as determined by PRF demonstrate that the strongest peptide signals are not necessarily the most reliable for quantitation. After lamin-A,C knockdown, PRF computes an invariant set of "housekeeping" proteins as part of a broader proteomic analysis that also shows the proteome of mesenchymal stem cells (MSCs) is more broadly perturbed than that of a human epithelial cancer line (A549s), with particular variation in nuclear and cytoskeletal proteins. These methods offer exciting prospects for basic and clinical studies of lamin-A,C as well as other MS-detectable proteins.
Collapse
Affiliation(s)
- Joe Swift
- Molecular & Cell Biophysics Laboratory, University of Pennsylvania; Philadelphia PA USA; Center for Systems and Computational Biology; Wistar Institute; Philadelphia PA USA
| | - Takamasa Harada
- Molecular & Cell Biophysics Laboratory, University of Pennsylvania; Philadelphia PA USA
| | - Amnon Buxboim
- Molecular & Cell Biophysics Laboratory, University of Pennsylvania; Philadelphia PA USA
| | - Jae-Won Shin
- Molecular & Cell Biophysics Laboratory, University of Pennsylvania; Philadelphia PA USA
| | - Hsin-Yao Tang
- Center for Systems and Computational Biology; Wistar Institute; Philadelphia PA USA
| | - David W Speicher
- Center for Systems and Computational Biology; Wistar Institute; Philadelphia PA USA
| | - Dennis E Discher
- Molecular & Cell Biophysics Laboratory, University of Pennsylvania; Philadelphia PA USA; Center for Systems and Computational Biology; Wistar Institute; Philadelphia PA USA
| |
Collapse
|
9
|
Yamaguchi A, Iwatani M, Ogawa M, Kitano H, Matsuyama M. In vitro characterization of the RS motif in N-terminal head domain of goldfish germinal vesicle lamin B3 necessary for phosphorylation of the p34cdc2 target serine by SRPK1. FEBS Open Bio 2013; 3:165-76. [PMID: 23772390 PMCID: PMC3668540 DOI: 10.1016/j.fob.2013.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Revised: 03/20/2013] [Accepted: 03/21/2013] [Indexed: 11/28/2022] Open
Abstract
The nuclear envelopes surrounding the oocyte germinal vesicles of lower vertebrates (fish and frog) are supported by the lamina, which consists of the protein lamin B3 encoded by a gene found also in birds but lost in the lineage leading to mammals. Like other members of the lamin family, goldfish lamin B3 (gfLB3) contains two putative consensus phosphoacceptor p34cdc2 sites (Ser-28 and Ser-398) for the M-phase kinase to regulate lamin polymerization on the N- and C-terminal regions flanking a central rod domain. Partial phosphorylation of gfLB3 occurs on Ser-28 in the N-terminal head domain in immature oocytes prior to germinal vesicle breakdown, which suggests continual rearrangement of lamins by a novel lamin kinase in fish oocytes. We applied the expression-screening method to isolate lamin kinases by using phosphorylation site Ser-28-specific monoclonal antibody and a vector encoding substrate peptides from a goldfish ovarian cDNA library. As a result, SRPK1 was screened as a prominent lamin kinase candidate. The gfLB3 has a short stretch of the RS repeats (9-SRASTVRSSRRS-20) upstream of the Ser-28, within the N-terminal head. This stretch of repeats is conserved among fish lamin B3 but is not found in other lamins. In vitro phosphorylation studies and GST-pull down assay revealed that SRPK1 bound to the region of sequential RS repeats (9–20) with affinity and recruited serine into the active site by a grab-and-pull manner. These results indicate SRPK1 may phosphorylate the p34cdc2 site in the N-terminal head of GV-lamin B3 at the RS motifs, which have the general property of aggregation. SRPK1 was screened as a prominent lamin kinase candidate from goldfish ovary. The goldfish lamin B3 (LB3) has RS repeats upstream of the cdc2 target site. The RS repeats are conserved among fish LB3s but are not found in other lamins. SRPK1 binds to the RS repeats with affinity and phosphorylates cdc2 site by a grab-and-pull manner.
Collapse
Affiliation(s)
- Akihiko Yamaguchi
- Laboratory of Marine Biology, Department of Bioresource Sciences, Faculty of Agriculture, Kyushu University, Fukuoka 812-8581, Japan
| | | | | | | | | |
Collapse
|
10
|
Simon DN, Wilson KL. Partners and post-translational modifications of nuclear lamins. Chromosoma 2013; 122:13-31. [PMID: 23475188 DOI: 10.1007/s00412-013-0399-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 12/16/2022]
Abstract
Nuclear intermediate filament networks formed by A- and B-type lamins are major components of the nucleoskeleton that are required for nuclear structure and function, with many links to human physiology. Mutations in lamins cause diverse human diseases ('laminopathies'). At least 54 partners interact with human A-type lamins directly or indirectly. The less studied human lamins B1 and B2 have 23 and seven reported partners, respectively. These interactions are likely to be regulated at least in part by lamin post-translational modifications. This review summarizes the binding partners and post-translational modifications of human lamins and discusses their known or potential implications for lamin function.
Collapse
Affiliation(s)
- Dan N Simon
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
11
|
Zaremba-Czogalla M, Piekarowicz K, Wachowicz K, Kozioł K, Dubińska-Magiera M, Rzepecki R. The different function of single phosphorylation sites of Drosophila melanogaster lamin Dm and lamin C. PLoS One 2012; 7:e32649. [PMID: 22393432 PMCID: PMC3290585 DOI: 10.1371/journal.pone.0032649] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 01/28/2012] [Indexed: 11/18/2022] Open
Abstract
Lamins' functions are regulated by phosphorylation at specific sites but our understanding of the role of such modifications is practically limited to the function of cdc 2 (cdk1) kinase sites in depolymerization of the nuclear lamina during mitosis. In our study we used Drosophila lamin Dm (B-type) to examine the function of particular phosphorylation sites using pseudophosphorylated mutants mimicking single phosphorylation at experimentally confirmed in vivo phosphosites (S(25)E, S(45)E, T(435)E, S(595)E). We also analyzed lamin C (A-type) and its mutant S(37)E representing the N-terminal cdc2 (mitotic) site as well as lamin Dm R(64)H mutant as a control, non-polymerizing lamin. In the polymerization assay we could observe different effects of N-terminal cdc2 site pseudophosphorylation on A- and B-type lamins: lamin Dm S(45)E mutant was insoluble, in contrast to lamin C S(37)E. Lamin Dm T(435)E (C-terminal cdc2 site) and R(64)H were soluble in vitro. We also confirmed that none of the single phosphorylation site modifications affected the chromatin binding of lamin Dm, in contrast to the lamin C N-terminal cdc2 site. In vivo, all lamin Dm mutants were incorporated efficiently into the nuclear lamina in transfected Drosophila S2 and HeLa cells, although significant amounts of S(45)E and T(435)E were also located in cytoplasm. When farnesylation incompetent mutants were expressed in HeLa cells, lamin Dm T(435)E was cytoplasmic and showed higher mobility in FRAP assay.
Collapse
Affiliation(s)
| | | | | | | | | | - Ryszard Rzepecki
- Laboratory of Nuclear Proteins, Faculty of Biotechnology, University of Wroclaw, Wroclaw, Poland
| |
Collapse
|
12
|
Prokocimer M, Davidovich M, Nissim-Rafinia M, Wiesel-Motiuk N, Bar DZ, Barkan R, Meshorer E, Gruenbaum Y. Nuclear lamins: key regulators of nuclear structure and activities. J Cell Mol Med 2009; 13:1059-85. [PMID: 19210577 PMCID: PMC4496104 DOI: 10.1111/j.1582-4934.2008.00676.x] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2008] [Accepted: 01/19/2009] [Indexed: 11/27/2022] Open
Abstract
The nuclear lamina is a proteinaceous structure located underneath the inner nuclear membrane (INM), where it associates with the peripheral chromatin. It contains lamins and lamin-associated proteins, including many integral proteins of the INM, chromatin modifying proteins, transcriptional repressors and structural proteins. A fraction of lamins is also present in the nucleoplasm, where it forms stable complexes and is associated with specific nucleoplasmic proteins. The lamins and their associated proteins are required for most nuclear activities, mitosis and for linking the nucleoplasm to all major cytoskeletal networks in the cytoplasm. Mutations in nuclear lamins and their associated proteins cause about 20 different diseases that are collectively called laminopathies'. This review concentrates mainly on lamins, their structure and their roles in DNA replication, chromatin organization, adult stem cell differentiation, aging, tumorogenesis and the lamin mutations leading to laminopathic diseases.
Collapse
Affiliation(s)
- Miron Prokocimer
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Maya Davidovich
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Malka Nissim-Rafinia
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Naama Wiesel-Motiuk
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Daniel Z Bar
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Rachel Barkan
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Eran Meshorer
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| | - Yosef Gruenbaum
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of JerusalemJerusalem, Israel
| |
Collapse
|
13
|
Melcer S, Gruenbaum Y, Krohne G. Invertebrate lamins. Exp Cell Res 2007; 313:2157-66. [PMID: 17451683 DOI: 10.1016/j.yexcr.2007.03.004] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2007] [Revised: 02/25/2007] [Accepted: 03/06/2007] [Indexed: 01/11/2023]
Abstract
Lamins are the main component of the nuclear lamina and considered to be the ancestors of all intermediate filament proteins. They are localized mainly at the nuclear periphery where they form protein complexes with integral proteins of the nuclear inner membrane, transcriptional regulators, histones and chromatin modifiers. Studying lamins in invertebrate species has unique advantages including the smaller number of lamin genes in the invertebrate genomes and powerful genetic analyses in Caenorhabditis elegans and Drosophila melanogaster. These simpler nuclear lamina systems allow direct analyses of their structure and functions. Here we give an overview of recent advances in the field of invertebrate nuclear lamins with special emphasis on their evolution, assembly and functions.
Collapse
Affiliation(s)
- Shai Melcer
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | | | | |
Collapse
|
14
|
Mattout A, Goldberg M, Tzur Y, Margalit A, Gruenbaum Y. Specific and conserved sequences in D. melanogaster and C. elegans lamins and histone H2A mediate the attachment of lamins to chromosomes. J Cell Sci 2006; 120:77-85. [PMID: 17148572 DOI: 10.1242/jcs.03325] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The intimate association between nuclear lamins and chromatin is thought to regulate higher order chromatin organization. Previous studies have mapped a region between the rod domain and the Ig fold in the tail domain of Drosophila melanogaster lamin Dm0, which binds chromatin in vitro via the histone H2A/H2B dimer. This region contains an evolutionarily conserved nuclear localization signal (NLS) KRKR, and a sequence composed of the amino acids TRAT. Here we show that binding of lamin Dm0 to chromatin requires both NLS and TRAT sequences. Substituting either of the threonine residues in the TRAT sequence with negatively charged residues decreases the binding of lamin Dm0 to chromatin, indicating that this binding could be regulated by phosphorylation. Both lamin Dm0 and C. elegans Ce-lamin bind directly to histone H2A in vitro and this binding requires the NLS. The amino and carboxyl tail domains of histone H2A are each essential, but not sufficient, for binding to lamin Dm0; only a polypeptide containing both histone H2A tail domains binds efficiently to lamin Dm0. Taken together, these results suggest that specific residues in lamin Dm0 and histone H2A mediate the attachment of the nuclear lamina to chromosomes in vivo, which could have implications on the understanding of laminopathic diseases.
Collapse
Affiliation(s)
- Anna Mattout
- Department of Genetics, The Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem 91904 Israel
| | | | | | | | | |
Collapse
|
15
|
Yamaguchi A, Katsu Y, Matsuyama M, Yoshikuni M, Nagahama Y. Phosphorylation of the p34(cdc2) target site on goldfish germinal vesicle lamin B3 before oocyte maturation. Eur J Cell Biol 2006; 85:501-17. [PMID: 16600424 DOI: 10.1016/j.ejcb.2006.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2005] [Revised: 02/07/2006] [Accepted: 02/08/2006] [Indexed: 10/24/2022] Open
Abstract
The nuclear membranes surrounding fish and frog oocyte germinal vesicles (GVs) are supported by the lamina, an internal, mesh-like structure that consists of the protein lamin B3. The mechanisms by which lamin B3 is transported into GVs and is assembled to form the nuclear lamina are not well understood. In this study, we developed a heterogeneous microinjection system in which wild-type or mutated goldfish GV lamin B3 (GFLB3) was expressed in Escherichia coli, biotinylated, and microinjected into Xenopus oocytes. The localization of the biotinylated GFLB3 was visualized by fluorescence confocal microscopy. The results of these experiments indicated that the N-terminal domain plays important roles in both nuclear transport and assembly of lamin B3 to form the nuclear lamina. The N-terminal domain includes a major consensus phosphoacceptor site for the p34(cdc2) kinase at amino acid residue Ser-28. To investigate nuclear lamin phosphorylation, we generated a monoclonal antibody (C7B8D) against Ser-28-phosphorylated GFLB3. Two-dimensional (2-D) electrophoresis of GV protein revealed two major spots of lamin B3 with different isoelectric points (5.9 and 6.1). The C7B8D antibody recognized the pI-5.9 spot but not the pI-6.1 spot. The former spot disappeared when the native lamina was incubated with lambda phage protein phosphatase (lambda-PP), indicating that a portion of the lamin protein was already phosphorylated in the goldfish GV-stage oocytes. GFLB3 that had been microinjected into Xenopus oocytes was also phosphorylated in Xenopus GV lamina, as judged by Western blotting with C7B8D. Thus, lamin phosphorylation appears to occur prior to oocyte maturation in vivo in both these species. Taken together, our results suggest that the balance between phosphorylation by interphase lamin kinases and dephosphorylation by phosphatases regulates the conformational changes in the lamin B3 N-terminal head domain that in turn regulates the continual in vivo rearrangement and remodeling of the oocyte lamina.
Collapse
Affiliation(s)
- Akihiko Yamaguchi
- Laboratory of Marine Biology, Department of Animal and Marine Bioresource Sciences, Faculty of Agriculture, Kyushu University, Hakozaki 6-10-1, Higashi-Ku, Fukuoka 812-8581, Japan.
| | | | | | | | | |
Collapse
|
16
|
Pang M, McConnell M, Fisher PA. The Drosophila mus 308 gene product, implicated in tolerance of DNA interstrand crosslinks, is a nuclear protein found in both ovaries and embryos. DNA Repair (Amst) 2005; 4:971-82. [PMID: 15961355 DOI: 10.1016/j.dnarep.2005.04.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2004] [Revised: 04/08/2005] [Accepted: 04/11/2005] [Indexed: 10/25/2022]
Abstract
mus 308 designates one of over 30 mutagen sensitivity loci found in Drosophila. It is predicted to code for a 229-kDa polypeptide. Published sequence analyses of others indicate that this polypeptide would have helicase motifs near its N-terminus, and similarities to bacterial DNA polymerase I-like enzymes near its C-terminus. In our studies, two different and highly specific antibodies were prepared and used for identification as well as characterization of the mus 308 gene product. Western blot analyses reveal a single reactive polypeptide in both ovaries and embryos as well as in two Drosophila embryo tissue culture cell lines; it is nearly absent in homozygous mus 308 mutants. This polypeptide is about 229 kDa in size, and indirect immunofluorescence shows that the mus 308 gene product localizes throughout nuclei in wild-type cells but appears to be absent in a mus 308 mutant. Immunoblot analyses throughout development suggest greatest abundance at the end of embryogenesis, immediately before hatching of first instar larvae. They also showed a smaller ( approximately 100 kDa) antigenically and genetically related polypeptide found only in adult males. Immunoprecipitation, a highly effective method of specific purification, suggests that the mus 308 protein has DNA polymerase activity that is NEM-sensitive but largely aphidicolin-resistant. In addition, the immunoprecipitated material has DNA-dependent ATPase but lacks detectable helicase.
Collapse
Affiliation(s)
- Maria Pang
- Department of Pharmacological Sciences, State University of New York at Stony Brook, Stony Brook, NY 11794-8651, USA
| | | | | |
Collapse
|
17
|
Summers MK, Bothos J, Halazonetis TD. The CHFR mitotic checkpoint protein delays cell cycle progression by excluding Cyclin B1 from the nucleus. Oncogene 2005; 24:2589-98. [PMID: 15674323 DOI: 10.1038/sj.onc.1208428] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CHFR, a novel checkpoint gene inactivated in human cancer, delays chromosome condensation in cells treated with microtubule poisons. To understand the molecular mechanism for this delay, we characterized cells with inactivated CHFR and stably transfected derivatives expressing the wild-type gene. After exposure to microtubule poisons, the CHFR-expressing cells arrested transiently in early prophase with a characteristic ruffled morphology of the nuclear envelope and no signs of chromosome condensation. Several markers suggested that Cyclin A/Cdc2 had been activated, whereas Aurora-A and -B and Cyclin B1/Cdc2 were inactive. Further, Cyclin B1 was excluded from the nucleus. Ectopic expression of Cyclin B1 with a mutant nuclear export sequence induced chromosome condensation, and thus overcame the CHFR checkpoint. We conclude that the mechanism by which CHFR delays chromosome condensation involves inhibition of accumulation of Cyclin B1 in the nucleus.
Collapse
Affiliation(s)
- Matthew K Summers
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104-4268, USA
| | | | | |
Collapse
|
18
|
Loseva O, Engström Y. Analysis of Signal-dependent Changes in the Proteome of Drosophila Blood Cells During an Immune Response. Mol Cell Proteomics 2004; 3:796-808. [PMID: 15148344 DOI: 10.1074/mcp.m400028-mcp200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Innate immunity is based on the recognition of cell-surface molecules of infecting agents. Microbial substances, such as peptidoglycan, lipopolysaccharide, and beta-1,3-glucans, produce functional responses in Drosophila hemocytes that contribute to innate immunity. We have used two-dimensional gel electrophoresis and MS to resolve lipopolysaccharide-induced changes in the protein profile of a Drosophila hemocytic cell line. We identified 24 intracellular proteins that were up- or down-regulated, or modified, in response to immune challenge. Several proteins with predicted immune functions, including lysosomal proteases, actin-binding/remodeling proteins, as well as proteins involved in cellular responses to oxidative stress, were affected by the immune assault. Intriguingly, a number of the proteins identified in this study have recently been implicated in phagocytosis in higher vertebrates. We suggest that phagocytosis is activated in Drosophila hemocytes by the presence of microbial substances, and that this activation constitutes an evolutionarily conserved arm of innate immunity. In addition, a number of proteins involved in calcium-regulated signaling, mRNA processing, and nuclear transport were affected, consistent with a possible role in reprogramming of gene expression. In conclusion, the present proteome analysis identified many proteins previously not linked to innate immunity, demonstrating that differential protein profiling of Drosophila hemocytes is a valuable tool for identification of new players in immune-related cellular processes.
Collapse
Affiliation(s)
- Olga Loseva
- Department of Molecular Biology and Functional Genomics, Arrhenius Laboratories for Natural Sciences, Stockholm University, Stockholm SE-106 91, Sweden
| | | |
Collapse
|
19
|
Organization, Replication, Transposition, and Repair of DNA. Biochemistry 2001. [DOI: 10.1016/b978-012492543-4/50030-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
20
|
Rzepecki R, Fisher PA. During both interphase and mitosis, DNA topoisomerase II interacts with DNA as well as RNA through the protein's C-terminal domain. J Cell Sci 2000; 113 ( Pt 9):1635-47. [PMID: 10751154 DOI: 10.1242/jcs.113.9.1635] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
DNA topoisomerase II (topo II) is thought to be a nuclear enzyme; during interphase most was insoluble and could be recovered in the pellet after centrifugation of cell homogenates at 10,000 g (P-10). Upon entry into mitosis, the majority of topo II did not associate with condensed chromosomes but was apparently solubilized and redistributed throughout the cell. Although two non-chromosomal subfractions of mitotic topo II were defined by centrifugation at 130,000 g, the vast majority (>90%) was recovered in the pellet (P-130). In vivo nucleic acid interactions with topo II were monitored by a recently developed approach of UV-photo-crosslinking, immunoprecipitation and (32)P-labeling. P-10 (interphase) topo II was largely associated with DNA. P-130 (mitotic non-chromosomal) topo II was primarily associated with RNA. These nucleic acid interactions with both interphase and mitotic topo II occurred through the catalytically inert and as yet, poorly understood C-terminal domain of the protein. P-10 topo II was highly active enzymatically. Activity, measured by the ability of topo II to decatenate kDNA minicircles, was reduced by treatment with phosphatase. In contrast, P-130 topo II was relatively inactive but activity could be increased by phosphatase treatment. In vivo, P-130 topo II was more heavily phosphorylated than P-10 topo II; in both, only the C-terminal domain of topo II was detectably modified. Our observations suggest that cell cycle-dependent changes in the distribution, nucleic acid interactions and enzymatic activity of topo II are regulated, at least in part, by phosphorylation/dephosphorylation.
Collapse
Affiliation(s)
- R Rzepecki
- The Department of Pharmacological Sciences, University Medical Center, The State University of New York at Stony Brook, Stony Brook, NY 11794-8651, USA
| | | |
Collapse
|
21
|
Bakhtiar R, Nelson RW. Electrospray ionization and matrix-assisted laser desorption ionization mass spectrometry. Emerging technologies in biomedical sciences. Biochem Pharmacol 2000; 59:891-905. [PMID: 10692554 DOI: 10.1016/s0006-2952(99)00317-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tremendous progress in biomedical sciences has been made possible in part by recent advances in bioanalytical methods, in particular biological mass spectrometry. Since the introduction of electrospray ionization mass spectrometry (ESI-MS) in 1984 and matrix-assisted laser desorption ionization mass spectrometry (MALDI-MS) in 1988, the field of bioanalytical mass spectrometry has seen rapid growth. In concert with separation techniques such as capillary electrophoresis and high performance liquid chromatography, mass spectrometry allows characterization of a large array of small organic molecules, peptides, proteins, oligonucleotides, and RNA fragments. Thus, substantially more expedient and definitive determination of molecular weight is now possible by mass spectrometric analysis. In this commentary, general descriptions of ESI- and MALDI-MS are presented. Furthermore, several recent developments and applications in addressing difficult biological problems are discussed.
Collapse
Affiliation(s)
- R Bakhtiar
- Department of Chemistry, Stevens Institute of Technology, Hoboken, NJ, USA.
| | | |
Collapse
|
22
|
Abstract
Lamins and other nuclear envelope proteins organize nuclear architecture through structural attachments that vary dynamically during the cell cycle and cell differentiation. Genetic studies have now shown that people with mutations in either lamins A/C or emerin, a nuclear membrane protein, develop Emery-Dreifuss muscular dystrophy. A mouse model for this rare disease has been created by knocking out the gene that encodes lamin A/C. This article discusses these and other recent results in the wider context of nuclear envelope function, as a framework for thinking about the possible ways in which defects in nuclear envelope proteins can lead to disease.
Collapse
Affiliation(s)
- K L Wilson
- Dept of Cell Biology and Anatomy, The Johns Hopkins University School of Medicine, 725 N. Wolfe Street, Baltimore, MD 21205, USA.
| |
Collapse
|
23
|
Munch-Petersen B, Knecht W, Lenz C, Søndergaard L, Piskur J. Functional expression of a multisubstrate deoxyribonucleoside kinase from Drosophila melanogaster and its C-terminal deletion mutants. J Biol Chem 2000; 275:6673-9. [PMID: 10692477 DOI: 10.1074/jbc.275.9.6673] [Citation(s) in RCA: 68] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The occurrence of a deoxyribonucleoside kinase in Drosophila melanogaster (Dm-dNK) with remarkably broad substrate specificity has recently been indicated (Munch-Petersen, B., Piskur, J., and Søndergaard, L. (1998) J. Biol. Chem. 273, 3926-3931). To prove that the capacity to phosphorylate all four deoxyribonucleosides is in fact associated to one polypeptide chain, partially sequenced cDNA clones, originating from the Berkeley Drosophila genome sequencing project, were searched for homology with human deoxyribonucleoside kinases. The total sequence of one cDNA clone and the corresponding genomic DNA was determined and expressed in Escherichia coli as a glutathione S-transferase fusion protein. The purified and thrombin cleaved recombinant protein phosphorylated the four deoxyribonucleosides with high turnover and K(m) values similar to those of the native Dm-dNK, as well as the four ribonucleosides and many therapeutical nucleoside analogs. Dm-dNK has apparently the same origin as the mammalian kinases, thymidine kinase 2, deoxycytidine kinase, deoxyguanosine kinase, and the herpes viral thymidine kinases, but it has a unique C terminus that seems to be important for catalytic activity and specificity. The C-terminal 20 amino acids were dispensable for phosphorylation of deoxyribonucleosides but necessary for full activity with purine ribonucleosides. Removal of the C-terminal 20 amino acids increased the specific activity 2-fold, but 99% of the activity was lost after removal of the C-terminal 30 amino acids.
Collapse
Affiliation(s)
- B Munch-Petersen
- Department of Life Sciences and Chemistry, Roskilde University, P. O. Box 260, DK 4000 Roskilde, Denmark.
| | | | | | | | | |
Collapse
|