Lamin


DEVELOPMENTAL BIOLOGY

Lamin-B in systemic inflammation, tissue homeostasis, and aging

Gradual loss of tissue function (or homeostasis) is a natural process of aging and is believed to cause many age-associated diseases. In human epidemiology studies, the low-grade and chronic systemic inflammation in elderly has been correlated with the development of aging related pathologies. Although it is suspected that tissue decline is related to systemic inflammation, the cause and consequence of these aging phenomena are poorly understood. By studying the Drosophila fat body and gut, this study has uncovered a mechanism by which lamin-B loss in the fat body upon aging induces age-associated systemic inflammation. This chronic inflammation results in the repression of gut local immune response, which in turn leads to the over-proliferation and mis-differentiation of the intestinal stem cells, thereby resulting in gut hyperplasia. Implications and remaining questions are discussed in light of these new observations (Chen, 2015).

Confocal analysis of nuclear lamina behavior during male meiosis and spermatogenesis in Drosophila melanogaster

Lamin family proteins are structural components of a filamentous framework, the nuclear lamina (NL), underlying the inner membrane of nuclear envelope. The NL not only plays a role in nucleus mechanical support and nuclear shaping, but is also involved in many cellular processes including DNA replication, gene expression and chromatin positioning. Spermatogenesis is a very complex differentiation process in which each stage is characterized by nuclear architecture dramatic changes, from the early mitotic stage to the sperm differentiation final stage. Nevertheless, very few data are present in the literature on the NL behavior during this process. This study shows the first and complete description of NL behavior during meiosis and spermatogenesis in Drosophila melanogaster. By confocal imaging, the NL modifications were characterized from mitotic stages, through meiotic divisions to sperm differentiation with an anti-laminDm0 antibody against the major component of the Drosophila NL. It was observed that continuous changes in the NL structure occurred in parallel with chromatin reorganization throughout the whole process and that meiotic divisions occurred in a closed context. Finally, NL was examined in solofuso meiotic mutant, where chromatin segregation is severely affected, and a strict correlation was found between the presence of chromatin and that of NL (Fabbretti, 2016).

Age-associated de-repression of retrotransposons in the Drosophila fat body, its potential cause and consequence

Eukaryotic genomes contain transposable elements (TE) that can move into new locations upon activation. Since uncontrolled transposition of TEs, including the retrotransposons and DNA transposons, can lead to DNA breaks and genomic instability, multiple mechanisms, including heterochromatin-mediated repression, have evolved to repress TE activation. Studies in model organisms have shown that TEs become activated upon aging as a result of age-associated deregulation of heterochromatin. Considering that different organisms or cell types may undergo distinct heterochromatin changes upon aging, it is important to identify pathways that lead to TE activation in specific tissues and cell types. Through deep sequencing of isolated RNAs, this study report an increased expression of many retrotransposons in the old Drosophila fat body, an organ equivalent to the mammalian liver and adipose tissue. This de-repression correlates with an increased number of DNA damage foci and decreased level of Drosophila lamin-B in the old fat body cells. Depletion of the Drosophila lamin-B in the young or larval fat body results in a reduction of heterochromatin and a corresponding increase in retrotransposon expression and DNA damage. Further manipulations of lamin-B and retrotransposon expression suggest a role of the nuclear lamina in maintaining the genome integrity of the Drosophila fat body by repressing retrotransposons (Chen, 2016).

Effects of Mutation or Deletion

A Drosophila Lamin mutant is described resulting from a P element insertion into the first intron of the Lamin gene. Homozygous mutant animals showed a severe phenotype including retardation in development, reduced viability, sterility, and impaired locomotion. Reduced Lamin expression causes an enrichment of nuclear pore complexes in cytoplasmic annulate lamellae and in nuclear envelope clusters. In several cells, particularly the densely packed somata of the central nervous system, defective nuclear envelopes are also observed. All aspects of the mutant phenotype are rescued upon P element-mediated germline transformation with a Lamin transgene. These data constitute the first genetic proof that lamins are essential for the structural organization of the cell nucleus (Lenz-Bohme, 1997).

The functions of Klarsicht and nuclear lamin in developmentally regulated nuclear migrations of photoreceptor cells in the Drosophila eye

The Klarsicht (Klar) protein is a crucial factor in the regulation of bidirectional transport of lipid droplets. Lipid droplets in early embryos move bidirectionally along microtubules, and the balance of plus- and minus-end travel distances changes twice over a 2-h period, resulting in switches in the direction of net transport. In the absence of Klar, travel distances, travel velocities, and stall forces are greatly reduced, for both plus- and minus-end travel. Without Klar the motors for plus- and minus-end motion are active indiscriminately, engaging in a tug-of-war. Thus, Klar seems to be central for understanding how the activity of opposite-polarity motors is coordinated during bidirectional transport. In this system, Klar controls the minus-end motor cytoplasmic dynein and an as yet unknown plus-end motor (Patterson, 2004; Guo, 2005 and references therein).

In its role in photoreceptor nuclear migration, Klarsicht is required for connecting the microtubule organizing center (MTOC) to the nucleus. In addition, in a genetic screen for klarsicht-interacting genes, Lam Dm0, which encodes nuclear lamin, was found. Like Klarsicht, lamin is required for photoreceptor nuclear migration and for nuclear attachment to the MTOC. Moreover, perinuclear localization of Klarsicht requires lamin. It is proposed that nuclear migration requires linkage of the MTOC to the nucleus through an interaction between microtubules, Klarsicht, and lamin (Patterson, 2004).

Nuclear migration in the developing eye is critical for shaping each individual cell and thus for normal morphology of the entire compound eye. The Drosophila compound eye develops within the larval eye imaginal disc, an epithelial monolayer. Within the eye disc, the morphogenetic furrow marks the initiation of eye assembly. Rows of identical facets, or ommatidia, assemble posterior to the furrow, starting with the eight photoreceptors (R-cells), followed by the lens-secreting cone cells, and finally the pigment cells. Anterior to the furrow, cells are undifferentiated and their nuclei are positioned randomly within the monolayer. The nuclei dive basally within the furrow and posterior to the furrow, migrate apically as they are recruited into ommatidia (Patterson, 2004 and references therein).

Two Drosophila genes, klarsicht (previously known as marbles) and Glued, are essential for the apical migration of nuclei in differentiating R-cells (Fischer-Vize, 1994; Fan, 1997). Glued encodes the large subunit of dynactin, a protein complex that regulates the minus-end-directed microtubule motor dynein. The requirement for dynactin suggests that R-cell nuclear migration is a dynein- and microtubule-dependent process. Consistent with this idea, two other Drosophila genes, Bicaudal-D< and Lis1, both of which may regulate dynein, are implicated in R-cell nuclear migration, although their mutant phenotypes are weak compared with klarsicht and Glued. Lis-1, a WD40 repeat protein, is the homolog of the human disease gene Lissencephaly-1. Lissencephaly, or smooth brain, is a disorder resulting from defects in neuronal migrations essential for normal human brain development. Neuronal migration requires nuclear migration, and the involvement of Lis-1 in Drosophila R-cell nuclear migration suggests that the two processes may be in part analogous. It is now clear that a connection between the MTOC and the nucleus is necessary for nuclear migration and that this connection is mediated by Klar and nuclear lamin. In addition to suggesting a specific role for Klar in nuclear migration, the results propose a general mechanistic explanation for the cytoplasmic effects of nuclear lamin, including human laminopathies (Patterson, 2004).

To understand the role of Klar in R-cell nuclear migration, Klar subcellular localization and the position of the MTOC was investigated in klar mutant eye discs. In addition, genetics was used to identify nuclear lamin, which functions in the same pathway with Klar. Klar was found to be perinuclear and associated with microtubules apical to the nucleus. In addition, in klar and Lam mutant discs, MTOCs form normally in R-cells, but are often not associated with the nucleus as they are in wild-type eyes. Finally, Lam+ was found to be required for Klar localization to the nuclear membrane. These observations, taken together with previous results, suggest a model for the function of Klar in nuclear migration where Klar, held in the nuclear envelope by nuclear lamin, links the nucleus to the MTOC (Patterson, 2004).

The interaction between Klar and lamin may be indirect, but it is likely to be specific, rather than a generalized failure of nuclear envelope assembly in Lam mutants. Although most R-cell nuclei fail to migrate apically even in weak, viable Lam mutants, >90% of nuclear envelopes are intact even in stronger, lethal Lam mutants (Patterson, 2004 and references therein).

It is proposed that one or more proteins may form a bridge between the KASH domain of Klar, present in the outer nuclear membrane, and nuclear lamin, in the inner nuclear envelope. The observation that in addition to its perinuclear localization, Klar is cytoplasmic (on apical microtubules) supports the idea that Klar is in the outer, as opposed to the inner, nuclear membrane. Similarly, C. elegans Anc-1 is present in the cytoplasm as well as the nuclear membrane, and a model has been proposed where the Anc-1 KASH domain is held in the outer nuclear membrane by an inner nuclear membrane protein, Unc-84. Although nuclear lamin has not been shown directly to be required for Anc-1 nuclear membrane localization, nuclear envelope localization of Unc-84 requires lamin. For Syne-1, the vertebrate homolog of Anc-1, experiments where the detergent digitonin was used to allow antibody access to the outer but not the inner nuclear membrane provide direct evidence that the KASH domain is in the outer nuclear membrane (Zhen, 2002). There is, however, some conflicting data (Patterson, 2004 and references therein).

It is speculated that the N-terminal portion of Klar is linked to microtubules by dynein. At present, it is not possible to test for colocalization of Klar and dynein because there are no available reagents that allow detection of dynein or dynactin in the eye disc. Nevertheless, there is much evidence to support an essential role for dynein in R-cell nuclear migration and Klar function. Dynactin, a regulator of dynein, is essential for R-cell nuclear migration in the eye; mutants in the p150 dynactin subunit (Glued) have a phenotype similar to that of klar mutants in the eye disc. In addition, dynein linkage could explain why Klar is localized to microtubules only apical to the nucleus; Klar that escapes the hold of the nuclear envelope, still attached to dynein, could walk along microtubules to the MTOC. Finally, Klar has been implicated as a regulator of dynein in Drosophila embryos. In addition to its role in R-cell nuclear migration, Klar is required for developmentally regulated migration of lipid storage vesicles during embryogenesis. Lipid droplets at the center of the cellular blastoderm embryo normally migrate cortically during gastrulation. In embryos from klar mutant mothers, the lipid droplets fail to migrate. A variety of data support a model where dynein transports the lipid droplets along microtubules, whose minus ends are at the cell periphery. The results of biophysical experiments has led to a model where Klar may attach the appropriate types of motor to lipid droplets, control the number of actively engaged motors on a droplet, or coordinate the activities of kinesins and dyneins bound simultaneously to the same droplet. Notably, dynein is required for nuclear attachment to centrosomes (the MTOCs) during mitosis in the Drosophila embryo. Klar, however, is not essential for this process (Patterson, 2004 and references therein).

The observation that the MTOC is normally apical to the R-cell nuclei, at the leading edge of nuclear movement, suggests that a force pulls on the MTOC from above. It is speculated that the mechanism for this force could be analogous to the means by which the nucleus of budding yeast are pulled into the bud neck before cell division. One pathway for migration of the nucleus into the bud neck involves dynein, anchored at the cell cortex to which the nucleus is moving. Cortically tethered dynein 'reels in' the nucleus by walking along microtubules whose plus ends are at the cortex, toward the MTOC, which is anchored to the nucleus. In support of this idea, microtubule plus-ends are present apically in R-cells, and dynactin is essential for R-cell nuclear migration (Patterson, 2004 and references therein).

Whether a force emanating from the apical membrane pulling on the MTOC would drive nuclear migration or serve as an anchor after the nucleus has migrated depends on where the MTOC initially forms. The gamma-tubulin antibody detects MTOCs only apically in differentiating cells. Transiently basal MTOCs associated with nuclei that are about to rise could have escaped detection. However, if the MTOC does form apically, then the force that drives nuclear migration would come from below the nucleus, that is, dynein, linked to the nuclear membrane by Klar and lamin, walking on microtubules up toward the MTOC (Patterson, 2004).

The model proposed whereby Klar forms a bridge between nuclear lamin in the inner nuclear membrane and cytoplasmic microtubules provides a general framework for explaining how nuclear lamin affects cytoplasmic events. Drosophila Lam mutations result in D/V polarity defects in eggs, and tracheal branching defects in embryos. Moreover, a variety of human diseases are the result of mutations in the LMNA gene, which encodes lamin A. The Drosophila Lam Dm0 gene encodes type B lamin, whereas the Drosophila LamC gene encodes lamin C, which is most similar to human lamin A. The A/C- and B-type lamins are similar proteins, with some different structural features, and some expression pattern differences. LMNA-associated human diseases affect the heart, skeletal muscles, and the nervous system (Emery-Dreifuss muscular dystrophy, limb-girdle muscular dystrophy, cardiomyopathy, and Charcot-Marie-Tooth disorder), and metabolism (Dunnigan-type lipodystrophy). The two main hypotheses as to how nuclear lamin defects can result in these disease phenotypes are that the mutations either result in nuclear envelope fragility or result in changes in gene expression. An alternative hypothesis is that the inner nuclear envelope interacts with the cytoplasm through proteins like Klar or Anc-1/Syne-1, which connect the inner nuclear envelope to the microtubule, or actin cytoskeletons, respectively (Patterson, 2004 and references therein).


REFERENCES

Amati, B. and Gasser, S. M. (1990a). Drosophila scaffold-attached regions bind nuclear scaffolds and can function as ARS elements in both budding and fission yeasts. Mol. Cell. Biol. (10): 5442-5454. PubMed Citation: 2118998

Amati, B., et al. (1990b). Nuclear scaffold attachment stimulates, but is not essential for ARS activity in Saccharomyces cerevisiae: analysis of the Drosophila ftz SAR. EMBO J. (12): 4007-4016. PubMed Citation: 2123454

Ashery-Padan, R., Weiss, A. M., Feinstein, N. and Gruenbaum, Y. (1997a). Distinct regions specify the targeting of otefin to the nucleoplasmic side of the nuclear envelope. J. Biol. Chem. 272(4): 2493-9. PubMed Citation: 8999964

Ashery-Padan, R., et al. (1997b). Localization and posttranslational modifications of otefin, a protein required for vesicle attachment to chromatin, during Drosophila melanogaster development. Mol. Cell Biol. 17(7): 4114-23. PubMed Citation: 9199347

Bailer, S. M., et al. (1991). Characterization of A 54-kD protein of the inner nuclear membrane: evidence for cell cycle-dependent interaction with the nuclear lamina. J. Cell Biol. 114(3): 389-400. PubMed Citation: 1650369

Baricheva, E. A., et al. (1996). DNA from Drosophila melanogaster beta-heterochromatin binds specifically to nuclear lamins in vitro and the nuclear envelope in situ. Gene 171(2): 171-176. PubMed Citation: 8666268

Bao, X., et al. (2005). The JIL-1 kinase interacts with lamin Dm0 and regulates nuclear lamina morphology of Drosophila nurse cells. J. Cell Sci. 118(Pt 21): 5079-87. 16254246

Bogachev, S. S., et al. (1996). Beta-heterochromatin in Drosophila: molecular organization and function. Molecular biological analysis of a MAR/SAR DNA sequence in centromere heterochromatin of Drosophila melanogaster. Genetika 32(2): 240-251 [Article in Russian]. PubMed Citation: 8713624

Bonne, G., et al. (1999). Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy. Nat. Genet. 21(3): 285-8. PubMed Citation: 10080180

Bossie, C. A. and Sanders, M. M. (1993). A cDNA from Drosophila melanogaster encodes a lamin C-like intermediate filament protein. J. Cell Sci. 104 ( Pt 4): 1263-72. PubMed Citation:

Buendia, B., Santa-Maria, A. and Courvalin, J. C. (1999). Caspase-dependent proteolysis of integral and peripheral proteins of nuclear membranes and nuclear pore complex proteins during apoptosis. J. Cell Sci. 112(Pt 11): 1743-1753. PubMed Citation: 10318766

Chen, H., Zheng, X. and Zheng, Y. (2014). Age-associated loss of lamin-B leads to systemic inflammation and gut hyperplasia. Cell 159: 829-843. PubMed ID: 25417159

Chen, H., Zheng, X. and Zheng, Y. (2015). Lamin-B in systemic inflammation, tissue homeostasis, and aging. Nucleus [Epub ahead of print]. PubMed ID: 25875575

Chen, H., Zheng, X., Xiao, D. and Zheng, Y. (2016). Age-associated de-repression of retrotransposons in the Drosophila fat body, its potential cause and consequence. Aging Cell [Epub ahead of print]. PubMed ID: 27072046

Collas, P., Courvalin, J. C. and Poccia, D. (1996). Targeting of membranes to sea urchin sperm chromatin is mediated by a lamin B receptor-like integral membrane protein. J Cell Biol. 135(6 Pt 2): 1715-25. 8991085

Collas, P. (1999). Sequential PKC- and Cdc2-mediated phosphorylation events elicit zebrafish nuclear envelope disassembly. J. Cell Sci. 112 ( Pt 6): 977-87. PubMed Citation: 10036247

Collas, P., et al. (1997). Protein kinase C-mediated interphase lamin B phosphorylation and solubilization. J. Biol. Chem. 272(34): 21274-21280. PubMed Citation: 9261138

Ellis, D. J., et al. (1997). GST-lamin fusion proteins act as dominant negative mutants in Xenopus egg extract and reveal the function of the lamina in DNA replication. J. Cell Sci. 110( Pt 20): 2507-2518. PubMed Citation: 9372440

Fabbretti, F., Iannetti, I., Guglielmi, L., Perconti, S., Evangelistella, C., Proietti De Santis, L., Bongiorni, S. and Prantera, G. (2016). Confocal analysis of nuclear lamina behavior during male meiosis and spermatogenesis in Drosophila melanogaster. PLoS One 11: e0151231. PubMed ID: 26963718

Foisner, R. and Gerace, L. (1993). Integral membrane proteins of the nuclear envelope interact with lamins and chromosomes, and binding is modulated by mitotic phosphorylation. Cell 73(7): 1267-1279. 8324822

Fuchs, E. and Weber, K. (1994). Intermediate filaments: structure, dynamics, function and disease. Annu. Rev. Biochem. 63: 345-382

Furukawa, K., et al. (1995). Cloning of a cDNA for lamina-associated polypeptide 2 (LAP2) and identification of regions that specify targeting to the nuclear envelope. EMBO J. 14(8): 1626-1636

Georgatos, S. D., Pyrpasopoulou, A., Theodoropoulos, P. A. (1997). Nuclear envelope breakdown in mammalian cells involves stepwise lamina disassembly and microtubule-drive deformation of the nuclear membrane. J. Cell Sci. 110: 2129-2140

Goldberg, M., et al. (1995). Xenopus lamin B3 has a direct role in the assembly of a replication competent nucleus: evidence from cell-free egg extracts. Cell Sci. 108( Pt 11): 3451-3461

Goldberg, M., et al. (1998). Interactions among Drosophila nuclear envelope proteins lamin, otefin, and YA. Mol. Cell Biol. 18(7): 4315-23. PubMed Citation: 9632815

Goldberg, M., et al. (1999). The tail domain of lamin Dm0 binds histones H2A and H2B. Proc. Natl. Acad. Sci. 96(6): 2852-7

Gruenbaum, Y., et al. (1988). Drosophila nuclear lamin precursor Dm0 is translated from either of two developmentally regulated mRNA species apparently encoded by a single gene. J. Cell Biol. 106(3): 585-596. 88153889

Guo, Y., Jangi, S. and Welte, M. A. (2005). Organelle-specific control of intracellular transport: distinctly targeted isoforms of the regulator Klar. Mol. Biol. Cell. 16(3): 1406-16. 15647372

Hocevar, B. A., Burns, D. J. and Fields, A. P. (1993). Identification of protein kinase C (PKC) phosphorylation sites on human lamin B. Potential role of PKC in nuclear lamina structural dynamics. J. Biol. Chem. 268(10): 7545-7552

Hozak, P., et al. (1995). Lamin proteins form an internal nucleoskeleton as well as a peripheral lamina in human cells. J. Cell Sci. 108( Pt 2): 635-644

Kawahire, S., et al. (1997). cDNA cloning of nuclear localization signal binding protein NBP60, a rat homologue of lamin B receptor, and identification of binding sites of human lamin B receptor for nuclear localization signals and chromatin. J. Biochem. (Tokyo) 121(5): 881-889

Lenz-Böhme, B., et al. (1997). Insertional mutation of the Drosophila nuclear Lamin Dm0 gene results in defective nuclear envelopes, clustering of nuclear pore complexes, and accumulation of annulate lamellae. J.Cell Biol 137: 1001-16

Lin, F. and Worman, H. J. (1997). Expression of nuclear lamins in human tissues and cancer cell lines and transcription from the promoters of the lamin A/C and B1 genes. Exp. Cell Res. 236(2): 378-384

Liu, J., Song, K. and Wolfner, M. F. (1995). Mutational analyses of fs(1)Ya, an essential, developmentally regulated, nuclear envelope protein in Drosophila. Genetics 141(4): 1473-1481

Liu, J., et al. (1997). Formation of the male pronuclear lamina in Drosophila melanogaster. Dev. Biol. 184(2): 187-196

Liu, J. and Wolfner, M. F. (1998). Functional dissection of YA, an essential, developmentally regulated nuclear lamina protein in Drosophila melanogaster. Mol. Cell. Biol. 18(1): 188-197

Lopez, J. M. and Wolfner, M. F. (1997). The developmentally regulated Drosophila embryonic nuclear lamina protein 'Young Arrest' (fs(1)Ya) is capable of associating with chromatin. J Cell Sci 1997 Mar;110( Pt 5): 643-651

Lourim, D., Kempf, A., and Krohne, G. (1996). Characterization and quantitation of three B-type lamins in Xenopus oocytes and eggs: increase of lamin LI protein synthesis during meiotic maturation. J. Cell Sci. 109( Pt 7): 1775-1785

Lourim, D. and Krohne, G. (1998). Chromatin binding and polymerization of the endogenous Xenopus egg lamins: the opposing effects of glycogen and ATP. J. Cell Sci. 111 (Pt 24): 3675-86

Luderus, M. E., et al. (1994). Binding of matrix attachment regions to lamin polymers involves single-stranded regions and the minor groove. Mol. Cell. Biol. (9): 6297-6305

Maison C., et al. (1997). The inner nuclear membrane protein LAP1 forms a native complex with B-type lamins and partitions with spindle-associated mitotic vesicles. EMBO J. 16(16): 4839-4850

Mani, S. S., et al. (2003). A hydrophilic lamin-binding domain from the Drosophila YA protein can target proteins to the nuclear envelope. J. Cell Sci. 116: 2067-2072. 12679383

Maus, N., Stuurman, N., and Fisher, P. A. (1995). Disassembly of the Drosophila nuclear lamina in a homologous cell-free system. J. Cell Sci. 108( Pt 5): 2027-2035

McCall, K. and Steller, H. (1998). Requirement for DCP-1 caspase during Drosophila oogenesis. Science 279(5348): 230-234

Milon, B. C., et al. (2012). Role of histone deacetylases in gene regulation at nuclear lamina. PLoS One 7(11): e49692. PubMed Citation: 23226217

Moir, R. D., et al. (2000). Nuclear lamins A and B1. Different pathways of assembly during nuclear envelope formation in living cells. J. Cell Biol. 151(6): 1155-68. 11121432

Mounkes, L. C., et al. (2003). A progeroid syndrome in mice is caused by defects in A-type lamins. Nature. 423(6937): 298-301. 12748643

Nazer, E., Dale, R. K., Chinen, M., Radmanesh, B. and Lei, E. P. (2018). Argonaute2 and LaminB modulate gene expression by controlling chromatin topology. PLoS Genet 14(3): e1007276. PubMed ID: 29529026

Nikolakaki, E., et al. (1997). Mitotic phosphorylation of the lamin B receptor by a serine/arginine kinase and p34(cdc2). J. Biol. Chem. 272(10): 6208-6213

Osman, M., et al. (1990). Molecular analysis of the Drosophila nuclear lamin gene. Genomics 1990 Oct;8(2): 217-224

Paddy, M. R., et al. (1996). Time-resolved, in vivo studies of mitotic spindle formation and nuclear lamina breakdown in Drosophila early embryos. J. Cell Sci. 109( Pt 3): 591-607

Padiath, Q. S., et al. (2006). Lamin B1 duplications cause autosomal dominant leukodystrophy. Nat. Genet. 38(10): 1114-23. Medline abstract: 16951681

Patterson, K., et al. (2004). The functions of Klarsicht and nuclear lamin in developmentally regulated nuclear migrations of photoreceptor cells in the Drosophila eye. Mol. Biol. Cell. 15(2): 600-10. 14617811

Pickersgill, H., et al. (2006). Characterization of the Drosophila melanogaster genome at the nuclear lamina. Nat. Genet. 38(9): 1005-14. Medline abstract: 16878134

Pyrpasopoulou, A., et al. (1996). The lamin B receptor (LBR) provides essential chromatin docking sites at the nuclear envelope. EMBO J. 15(24): 7108-7119

Riemer, D., et al. (1995). Expression of Drosophila lamin C is developmentally regulated: analogies with vertebrate A-type lamins. J. Cell Sci. 108( Pt 10): 3189-3198

Rzepecki, R., et al. (1998). In vivo association of lamins with nucleic acids in Drosophila melanogaster. J. Cell Sci. 111(1):121-129

Sasseville, A. M. and Raymond, Y. (1995). Lamin A precursor is localized to intranuclear foci. J. Cell Sci. 108(Pt 1): 273-285

Schmidt, M. and Krohne, G. (1996). In vivo assembly kinetics of fluorescently labeled Xenopus lamin A mutants. Eur. J. Cell Biol. 68(4): 345-354

Sharakhov, I. V., Bogachev, S. S. and Fisher, P. A. (1997). Chromosomal localization of M/SAR from DNA of the clone lambda20p1.4 in Drosophila melanogaster salivary glands. Genetika (2): 277-280 [Article in Russian]

Shevelyov, Y. Y., Lavrov, S. A., Mikhaylova, L. M., Nurminsky, I. D., Kulathinal, R. J., Egorova, K. S., Rozovsky, Y. M. and Nurminsky, D. I. (2009). The B-type lamin is required for somatic repression of testis-specific gene clusters. Proc Natl Acad Sci U S A 106: 3282-3287. PubMed ID: 19218438

Simos, G., et al. (1996). Characterization of p18, a component of the lamin B receptor complex and a new integral membrane protein of the avian erythrocyte nuclear envelope. J. Biol. Chem. 271(21): 12617-12625

Smith, D. E., et al. (1987). Biosynthesis and interconversion of Drosophila nuclear lamin isoforms during normal growth and in response to heat shock. J. Cell Biol. 105(2): 771-790. 87308383

Smith, D. E. and Fisher, P. A. (1989). Interconversion of Drosophila nuclear lamin isoforms during oogenesis, early embryogenesis, and upon entry of cultured cells into mitosis. J. Cell Biol. (2): 255-265. 89139549

Speese, S. D., Ashley, J., Jokhi, V., Nunnari, J., Barria, R., Li, Y., Ataman, B., Koon, A., Chang, Y. T., Li, Q., Moore, M. J. and Budnik, V. (2012). Nuclear envelope budding enables large ribonucleoprotein particle export during synaptic Wnt signaling. Cell 149(4): 832-846. PubMed ID: 22579286

Stuurman, N., Maus, N., and Fisher, P. A. (1995). Interphase phosphorylation of the Drosophila nuclear lamin: site-mapping using a monoclonal antibody. J. Cell Sci. 108(Pt 9): 3137-3144

Stuurman, N., Sasse, B., and Fisher, P. A. (1996). Intermediate filament protein polymerization: molecular analysis of Drosophila nuclear lamin head-to-tail binding. J. Struct. Biol. 117(1): 1-15

Stuurman, N. (1997). Identification of a conserved phosphorylation site modulating nuclear lamin polymerization. FEBS Lett. 401(2-3): 171-174

Stuurman, N., et al. (1999). Ectopic overexpression of Drosophila lamin C is stage-specific lethal. Exp. Cell Res. 248(2): 350-7

Stuurman, N., et al. (2000). Interactions between coiled-coil proteins: Drosophila lamin Dm0 binds to the bicaudal-D protein. Eur. J. Cell Biol. 78(4): 278-87

Takahashi, A., et al. (1996). CrmA/SPI-2 inhibition of an endogenous ICE-related protease responsible for lamin A cleavage and apoptotic nuclear fragmentation. J. Biol. Chem. 271(51): 32487-32490

Taniura, H., Glass, C. and Gerace, L. (1995). A chromatin binding site in the tail domain of nuclear lamins that interacts with core histones. J. Cell Biol. 131(1): 33-44

Uchino, R., Sugiyama, S., Katagiri, M., Chuman, Y. and Furukawa, K. (2016). Non-farnesylated B-type lamin can tether chromatin inside the nucleus and its chromatin interaction requires the Ig-fold region. Chromosoma [Epub ahead of print]. PubMed ID: 26892013

Ulitzur, N., et al. (1992). Lamin activity is essential for nuclear envelope assembly in a Drosophila embryo cell-free extract. J. Cell Biol. 119(1): 17-25

Verboon, J. M., Rincon-Arano, H., Werwie, T. R., Delrow, J. J., Scalzo, D., Nandakumar, V., Groudine, M. and Parkhurst, S. M. (2015a). Wash interacts with lamin and affects global nuclear organization. Curr Biol 25(6): 804-810. PubMed ID: 25754639

Verboon, J. M., Rahe, T. K., Rodriguez-Mesa, E. and Parkhurst, S. M. (2015b). Wash functions downstream of Rho1 GTPase in a subset of Drosophila immune cell developmental migrations. Mol Biol Cell 26(9): 1665-1674. PubMed ID: 25739458

Verboon, J. M., Nakamura, M., Davidson, K. A., Decker, J. R., Nandakumar, V. and Parkhurst, S. M. (2020). Drosophila Wash and the Wash regulatory complex function in nuclear envelope budding. J Cell Sci 133(13). PubMed ID: 32503943

Wagner, N., Weber, D., Seitz, S. and Krohne, G. (2004). The lamin B receptor of Drosophila melanogaster. J Cell Sci 117(Pt 10): 2015-2028. PubMed ID: 15054108

Weaver, V.M., et al. (1996). Degradation of nuclear matrix and DNA cleavage in apoptotic thymocytes. J. Cell Sci. 109( Pt 1): 45-56

Xie, W., Chojnowski, A., Boudier, T., Lim, J. S., Ahmed, S., Ser, Z., Stewart, C. and Burke, B. (2016). A-type lamins form distinct filamentous networks with differential nuclear pore complex associations. Curr Biol 26(19):2651-2658. PubMed ID: 27641764

Yang, L., Guan, T. and Gerace, L. (1997a). Integral membrane proteins of the nuclear envelope are dispersed throughout the endoplasmic reticulum during mitosis. J. Cell Biol. 137(6): 1199-1210

Yang, L., Guan, T. and Gerace, L. (1997b). Lamin-binding fragment of LAP2 inhibits increase in nuclear volume during the cell cycle and progression into S phase. J. Cell Biol. 139(5): 1077-1087. 98044214

Ye, Q. and Worman, H. J. (1994). Primary structure analysis and lamin B and DNA binding of human LBR, an integral protein of the nuclear envelope inner membrane. J. Biol. Chem. 269(15): 11306-11311

Ye, Q. and Worman, H. J. (1996). Interaction between an integral protein of the nuclear envelope inner membrane and human chromodomain proteins homologous to Drosophila HP1. J. Biol. Chem. 271(25): 14653-14656

Yu, J., et al. (1999). Nuclear entry of the Drosophila melanogaster nuclear lamina protein YA correlates with developmentally regulated changes in its phosphorylation state. Dev. Biol. 210(1): 124-34

Zhao, K., et al. (1996). Binding of matrix attachment regions to nuclear lamin is mediated by the rod domain and depends on the lamin polymerization state. FEBS Lett. 380(1-2): 161-164

Zhivotovsky, B., Gahm, A. and Orrenius, S. (1997). Two different proteases are involved in the proteolysis of lamin during apoptosis. Biochem. Biophys. Res. Commun. 233(1): 96-101


lamin Dm0: Biological Overview | Evolutionary Homologs | Regulation | Developmental Biology | Effects of Mutation

date revised: 20 April 2021 

Home page: The Interactive Fly © 1997 Thomas B. Brody, Ph.D.

The Interactive Fly resides on the
Society for Developmental Biology's Web server.