Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 126(4): 1592-602, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26999604

ABSTRACT

The alternatively spliced products of LMNA, lamin C and prelamin A (the precursor to lamin A), are produced in similar amounts in most tissues and have largely redundant functions. This redundancy suggests that diseases, such as Hutchinson-Gilford progeria syndrome (HGPS), that are caused by prelamin A-specific mutations could be treated by shifting the output of LMNA more toward lamin C. Here, we investigated mechanisms that regulate LMNA mRNA alternative splicing and assessed the feasibility of reducing prelamin A expression in vivo. We identified an exon 11 antisense oligonucleotide (ASO) that increased lamin C production at the expense of prelamin A when transfected into mouse and human fibroblasts. The same ASO also reduced the expression of progerin, the mutant prelamin A protein in HGPS, in fibroblasts derived from patients with HGPS. Mechanistic studies revealed that the exon 11 sequences contain binding sites for serine/arginine-rich splicing factor 2 (SRSF2), and SRSF2 knockdown lowered lamin A production in cells and in murine tissues. Moreover, administration of the exon 11 ASO reduced lamin A expression in wild-type mice and progerin expression in an HGPS mouse model. Together, these studies identify ASO-mediated reduction of prelamin A as a potential strategy to treat prelamin A-specific diseases.


Subject(s)
Alternative Splicing/drug effects , Lamin Type A/biosynthesis , Oligodeoxyribonucleotides, Antisense/pharmacology , Progeria/drug therapy , Progeria/metabolism , RNA, Messenger/metabolism , Animals , Disease Models, Animal , Exons , Gene Knockdown Techniques , Humans , Lamin Type A/genetics , Mice , Mice, Transgenic , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligodeoxyribonucleotides, Antisense/genetics , Progeria/genetics , RNA, Messenger/genetics , Ribonucleoproteins/genetics , Ribonucleoproteins/metabolism , Serine-Arginine Splicing Factors
2.
Hum Mol Genet ; 24(10): 2826-40, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25652409

ABSTRACT

Neurons in the brain produce lamin C but almost no lamin A, a consequence of the removal of prelamin A transcripts by miR-9, a brain-specific microRNA. We have proposed that miR-9-mediated regulation of prelamin A in the brain could explain the absence of primary neurological disease in Hutchinson-Gilford progeria syndrome, a genetic disease caused by the synthesis of an internally truncated form of farnesyl-prelamin A (progerin). This explanation makes sense, but it is not entirely satisfying because it is unclear whether progerin-even if were expressed in neurons-would be capable of eliciting neuropathology. To address that issue, we created a new Lmna knock-in allele, Lmna(HG-C), which produces progerin transcripts lacking an miR-9 binding site. Mice harboring the Lmna(HG-C) allele produced progerin in neurons, but they had no pathology in the central nervous system. However, these mice invariably developed esophageal achalasia, and the enteric neurons and nerve fibers in gastrointestinal tract were markedly abnormal. The same disorder, achalasia, was observed in genetically modified mice that express full-length farnesyl-prelamin A in neurons (Zmpste24-deficient mice carrying two copies of a Lmna knock-in allele yielding full-length prelamin A transcripts lacking a miR-9 binding site). Our findings indicate that progerin and full-length farnesyl-prelamin A are toxic to neurons of the enteric nervous system.


Subject(s)
Enteric Nervous System/pathology , Esophageal Achalasia/genetics , Lamin Type A/genetics , Neurons/metabolism , Protein Prenylation , Animals , Esophageal Achalasia/pathology , Female , Gene Knock-In Techniques , Lamin Type A/metabolism , Male , Mice , Mice, Transgenic , MicroRNAs/metabolism , Mutation , Neurons/pathology , RNA Interference
3.
Mol Cell Biol ; 34(24): 4534-44, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25312645

ABSTRACT

B-type lamins (lamins B1 and B2) have been considered to be essential for many crucial functions in the cell nucleus (e.g., DNA replication and mitotic spindle formation). However, this view has been challenged by the observation that an absence of both B-type lamins in keratinocytes had no effect on cell proliferation or the development of skin and hair. The latter findings raised the possibility that the functions of B-type lamins are subserved by lamins A and C. To explore that idea, we created mice lacking all nuclear lamins in keratinocytes. Those mice developed ichthyosis and a skin barrier defect, which led to death from dehydration within a few days after birth. Microscopy of nuclear-lamin-deficient skin revealed hyperkeratosis and a disordered stratum corneum with an accumulation of neutral lipid droplets; however, BrdU incorporation into keratinocytes was normal. Skin grafting experiments confirmed the stratum corneum abnormalities and normal BrdU uptake. Interestingly, the absence of nuclear lamins in keratinocytes resulted in an interspersion of nuclear/endoplasmic reticulum membranes with the chromatin. Thus, a key function of the nuclear lamina is to serve as a "fence" and prevent the incursion of cytoplasmic organelles into the nuclear chromatin.


Subject(s)
Ichthyosis/genetics , Keratinocytes/transplantation , Lamins/genetics , Lamins/metabolism , Skin/pathology , Animals , Animals, Newborn , Bromodeoxyuridine , Cell Proliferation , Chromatin/metabolism , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Female , Ichthyosis/mortality , Ichthyosis/pathology , Keratinocytes/metabolism , Mice , Mice, Nude , Skin/metabolism
4.
Cell Metab ; 19(5): 849-60, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24726386

ABSTRACT

Triglyceride-rich lipoproteins (TRLs) undergo lipolysis by lipoprotein lipase (LPL), an enzyme that is transported to the capillary lumen by an endothelial cell protein, GPIHBP1. For LPL-mediated lipolysis to occur, TRLs must bind to the lumen of capillaries. This process is often assumed to involve heparan sulfate proteoglycans (HSPGs), but we suspected that TRL margination might instead require GPIHBP1. Indeed, TRLs marginate along the heart capillaries of wild-type but not Gpihbp1⁻/⁻ mice, as judged by fluorescence microscopy, quantitative assays with infrared-dye-labeled lipoproteins, and EM tomography. Both cell-culture and in vivo studies showed that TRL margination depends on LPL bound to GPIHBP1. Notably, the expression of LPL by endothelial cells in Gpihbp1⁻/⁻ mice did not restore defective TRL margination, implying that the binding of LPL to HSPGs is ineffective in promoting TRL margination. Our studies show that GPIHBP1-bound LPL is the main determinant of TRL margination.


Subject(s)
Capillaries/metabolism , Lipoprotein Lipase/metabolism , Lipoproteins/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/metabolism , Animals , Cell Line , Endothelial Cells/metabolism , Heart/physiology , Mice
5.
Mol Biol Cell ; 25(10): 1666-75, 2014 May.
Article in English | MEDLINE | ID: mdl-24672053

ABSTRACT

Lamins B1 and B2 (B-type lamins) have very similar sequences and are expressed ubiquitously. In addition, both Lmnb1- and Lmnb2-deficient mice die soon after birth with neuronal layering abnormalities in the cerebral cortex, a consequence of defective neuronal migration. The similarities in amino acid sequences, expression patterns, and knockout phenotypes raise the question of whether the two proteins have redundant functions. To investigate this topic, we generated "reciprocal knock-in mice"-mice that make lamin B2 from the Lmnb1 locus (Lmnb1(B2/B2)) and mice that make lamin B1 from the Lmnb2 locus (Lmnb2(B1/B1)). Lmnb1(B2/B2) mice produced increased amounts of lamin B2 but no lamin B1; they died soon after birth with neuronal layering abnormalities in the cerebral cortex. However, the defects in Lmnb1(B2/B2) mice were less severe than those in Lmnb1-knockout mice, indicating that increased amounts of lamin B2 partially ameliorate the abnormalities associated with lamin B1 deficiency. Similarly, increased amounts of lamin B1 in Lmnb2(B1/B1) mice did not prevent the neurodevelopmental defects elicited by lamin B2 deficiency. We conclude that lamins B1 and B2 have unique roles in the developing brain and that increased production of one B-type lamin does not fully complement loss of the other.


Subject(s)
Cerebral Cortex/abnormalities , Lamin Type B/genetics , Neurogenesis/genetics , Amino Acid Sequence , Animals , Cerebral Cortex/embryology , Gene Knock-In Techniques , Lamin Type B/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Tube Defects/genetics , Nuclear Lamina/metabolism , Sequence Analysis, DNA
6.
Hum Mol Genet ; 23(6): 1506-15, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24203701

ABSTRACT

Lamins A and C (products of the LMNA gene) are found in roughly equal amounts in peripheral tissues, but the brain produces mainly lamin C and little lamin A. In HeLa cells and fibroblasts, the expression of prelamin A (the precursor to lamin A) can be reduced by miR-9, but the relevance of those cell culture studies to lamin A regulation in the brain was unclear. To address this issue, we created two new Lmna knock-in alleles, one (Lmna(PLAO-5NT)) with a 5-bp mutation in a predicted miR-9 binding site in prelamin A's 3' UTR, and a second (Lmna(PLAO-UTR)) in which prelamin A's 3' UTR was replaced with lamin C's 3' UTR. Neither allele had significant effects on lamin A levels in peripheral tissues; however, both substantially increased prelamin A transcript levels and lamin A protein levels in the cerebral cortex and the cerebellum. The increase in lamin A expression in the brain was more pronounced with the Lmna(PLAO-UTR) allele than with the Lmna(PLAO-5NT) allele. With both alleles, the increased expression of prelamin A transcripts and lamin A protein was greater in the cerebral cortex than in the cerebellum. Our studies demonstrate the in vivo importance of prelamin A's 3' UTR and its miR-9 binding site in regulating lamin A expression in the brain. The reduced expression of prelamin A in the brain likely explains why children with Hutchinson-Gilford progeria syndrome (a progeroid syndrome caused by a mutant form of prelamin A) are spared from neurodegenerative disease.


Subject(s)
Cerebellum/metabolism , Cerebral Cortex/metabolism , Lamin Type A/metabolism , MicroRNAs/metabolism , Nuclear Proteins/genetics , Progeria/genetics , Protein Precursors/genetics , 3' Untranslated Regions , Alleles , Animals , Disease Models, Animal , Female , Gene Expression Regulation , Gene Knock-In Techniques , Lamin Type A/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Mutation , Progeria/metabolism , Progeria/pathology
7.
Proc Natl Acad Sci U S A ; 110(21): E1923-32, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23650370

ABSTRACT

The role of protein farnesylation in lamin A biogenesis and the pathogenesis of progeria has been studied in considerable detail, but the importance of farnesylation for the B-type lamins, lamin B1 and lamin B2, has received little attention. Lamins B1 and B2 are expressed in nearly every cell type from the earliest stages of development, and they have been implicated in a variety of functions within the cell nucleus. To assess the importance of protein farnesylation for B-type lamins, we created knock-in mice expressing nonfarnesylated versions of lamin B1 and lamin B2. Mice expressing nonfarnesylated lamin B2 developed normally and were free of disease. In contrast, mice expressing nonfarnesylated lamin B1 died soon after birth, with severe neurodevelopmental defects and striking nuclear abnormalities in neurons. The nuclear lamina in migrating neurons was pulled away from the chromatin so that the chromatin was left "naked" (free from the nuclear lamina). Thus, farnesylation of lamin B1--but not lamin B2--is crucial for brain development and for retaining chromatin within the bounds of the nuclear lamina during neuronal migration.


Subject(s)
Brain/embryology , Cell Movement/physiology , Chromatin/metabolism , Lamin Type B/metabolism , Nuclear Lamina/metabolism , Protein Prenylation/physiology , Animals , Chromatin/genetics , Lamin Type B/genetics , Mice , Mice, Transgenic , Nuclear Lamina/genetics
8.
Mol Biol Cell ; 22(23): 4683-93, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21976703

ABSTRACT

Neuronal migration is essential for the development of the mammalian brain. Here, we document severe defects in neuronal migration and reduced numbers of neurons in lamin B1-deficient mice. Lamin B1 deficiency resulted in striking abnormalities in the nuclear shape of cortical neurons; many neurons contained a solitary nuclear bleb and exhibited an asymmetric distribution of lamin B2. In contrast, lamin B2 deficiency led to increased numbers of neurons with elongated nuclei. We used conditional alleles for Lmnb1 and Lmnb2 to create forebrain-specific knockout mice. The forebrain-specific Lmnb1- and Lmnb2-knockout models had a small forebrain with disorganized layering of neurons and nuclear shape abnormalities, similar to abnormalities identified in the conventional knockout mice. A more severe phenotype, complete atrophy of the cortex, was observed in forebrain-specific Lmnb1/Lmnb2 double-knockout mice. This study demonstrates that both lamin B1 and lamin B2 are essential for brain development, with lamin B1 being required for the integrity of the nuclear lamina, and lamin B2 being important for resistance to nuclear elongation in neurons.


Subject(s)
Brain/embryology , Brain/growth & development , Lamin Type B/metabolism , Neurons/pathology , Prosencephalon/abnormalities , Animals , Brain/abnormalities , Lamin Type B/genetics , Mice , Mice, Knockout , Neurons/metabolism , Nuclear Lamina , Prosencephalon/growth & development
9.
J Biol Chem ; 285(50): 39239-48, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-20889497

ABSTRACT

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), a GPI-anchored endothelial cell protein, binds lipoprotein lipase (LPL) and transports it into the lumen of capillaries where it hydrolyzes triglycerides in lipoproteins. GPIHBP1 is assumed to be expressed mainly within the heart, skeletal muscle, and adipose tissue, the sites where most lipolysis occurs, but the tissue pattern of GPIHBP1 expression has never been evaluated systematically. Because GPIHBP1 is found on the luminal face of capillaries, we predicted that it would be possible to define GPIHBP1 expression patterns with radiolabeled GPIHBP1-specific antibodies and positron emission tomography (PET) scanning. In Gpihbp1(-/-) mice, GPIHBP1-specific antibodies were cleared slowly from the blood, and PET imaging showed retention of the antibodies in the blood pools (heart and great vessels). In Gpihbp1(+/+) mice, the antibodies were cleared extremely rapidly from the blood and, to our surprise, were taken up mainly by lung and liver. Immunofluorescence microscopy confirmed the presence of GPIHBP1 in the capillary endothelium of both lung and liver. In most tissues with high levels of Gpihbp1 expression, Lpl expression was also high, but the lung was an exception (very high Gpihbp1 expression and extremely low Lpl expression). Despite low Lpl transcript levels, however, LPL protein was readily detectable in the lung, suggesting that some of that LPL originates elsewhere and then is captured by GPIHBP1 in the lung. In support of this concept, lung LPL levels were significantly lower in Gpihbp1(-/-) mice than in Gpihbp1(+/+) mice. In addition, Lpl(-/-) mice expressing human LPL exclusively in muscle contained high levels of human LPL in the lung.


Subject(s)
Gene Expression Regulation , Glycosylphosphatidylinositols/metabolism , Receptors, Lipoprotein/chemistry , Animals , Binding Sites , Capillaries/metabolism , Cell Membrane/metabolism , Endothelium/metabolism , Kinetics , Lung/metabolism , Mice , Mice, Transgenic , Models, Biological , Positron-Emission Tomography/methods
10.
Cell Metab ; 12(1): 42-52, 2010 Jul 07.
Article in English | MEDLINE | ID: mdl-20620994

ABSTRACT

The lipolytic processing of triglyceride-rich lipoproteins by lipoprotein lipase (LPL) is the central event in plasma lipid metabolism, providing lipids for storage in adipose tissue and fuel for vital organs such as the heart. LPL is synthesized and secreted by myocytes and adipocytes, but then finds its way into the lumen of capillaries, where it hydrolyzes lipoprotein triglycerides. The mechanism by which LPL reaches the lumen of capillaries has remained an unresolved problem of plasma lipid metabolism. Here, we show that GPIHBP1 is responsible for the transport of LPL into capillaries. In Gpihbp1-deficient mice, LPL is mislocalized to the interstitial spaces surrounding myocytes and adipocytes. Also, we show that GPIHBP1 is located at the basolateral surface of capillary endothelial cells and actively transports LPL across endothelial cells. Our experiments define the function of GPIHBP1 in triglyceride metabolism and provide a mechanism for the transport of LPL into capillaries.


Subject(s)
Capillaries/enzymology , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Adipose Tissue/blood supply , Animals , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Lipid Metabolism , Lipoprotein Lipase/analysis , Lipoproteins/metabolism , Mice , Mice, Knockout , Receptors, Lipoprotein/analysis , Receptors, Lipoprotein/genetics , Triglycerides/metabolism
11.
J Biol Chem ; 285(27): 20818-26, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20439468

ABSTRACT

Lamin A, a key component of the nuclear lamina, is generated from prelamin A by four post-translational processing steps: farnesylation, endoproteolytic release of the last three amino acids of the protein, methylation of the C-terminal farnesylcysteine, and finally, endoproteolytic release of the last 15 amino acids of the protein (including the farnesylcysteine methyl ester). The last cleavage step, mediated by ZMPSTE24, releases mature lamin A. This processing scheme has been conserved through vertebrate evolution and is widely assumed to be crucial for targeting lamin A to the nuclear envelope. However, its physiologic importance has never been tested. To address this issue, we created mice with a "mature lamin A-only" allele (Lmna(LAO)), which contains a stop codon immediately after the last codon of mature lamin A. Thus, Lmna(LAO/LAO) mice synthesize mature lamin A directly, bypassing prelamin A synthesis and processing. The levels of mature lamin A in Lmna(LAO/LAO) mice were indistinguishable from those in "prelamin A-only" mice (Lmna(PLAO/PLAO)), where all of the lamin A is produced from prelamin A. Lmna(LAO/LAO) exhibited normal body weights and had no detectable disease phenotypes. A higher frequency of nuclear blebs was observed in Lmna(LAO/LAO) embryonic fibroblasts; however, the mature lamin A in the tissues of Lmna(LAO/LAO) mice was positioned normally at the nuclear rim. We conclude that prelamin A processing is dispensable in mice and that direct synthesis of mature lamin A has little if any effect on the targeting of lamin A to the nuclear rim in mouse tissues.


Subject(s)
Cell Nucleus/pathology , Fibroblasts/pathology , Lamin Type A/biosynthesis , Animals , Blotting, Western , Conserved Sequence , Crosses, Genetic , Embryo, Mammalian , Fibroblasts/cytology , Fibroblasts/metabolism , Introns , Lamin Type A/genetics , Methylation , Mice , Mice, Knockout , Microscopy, Fluorescence , Mutagenesis, Site-Directed , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Protein Modification, Translational , Protein Precursors/genetics , Protein Precursors/metabolism , Vertebrates
12.
Proc Natl Acad Sci U S A ; 107(11): 5076-81, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20145110

ABSTRACT

Nuclear lamins are components of the nuclear lamina, a structural scaffolding for the cell nucleus. Defects in lamins A and C cause an array of human diseases, including muscular dystrophy, lipodystrophy, and progeria, but no diseases have been linked to the loss of lamins B1 or B2. To explore the functional relevance of lamin B2, we generated lamin B2-deficient mice and found that they have severe brain abnormalities resembling lissencephaly, with abnormal layering of neurons in the cerebral cortex and cerebellum. This neuronal layering abnormality is due to defective neuronal migration, a process that is dependent on the organized movement of the nucleus within the cell. These studies establish an essential function for lamin B2 in neuronal migration and brain development.


Subject(s)
Cerebellum/abnormalities , Cerebellum/embryology , Cerebral Cortex/abnormalities , Cerebral Cortex/embryology , Lamin Type B/deficiency , Animals , Cell Movement , Cerebellum/pathology , Cerebral Cortex/pathology , Gene Silencing , Lamin Type B/metabolism , Mice , Neurons/pathology
13.
J Biol Chem ; 283(15): 9797-804, 2008 Apr 11.
Article in English | MEDLINE | ID: mdl-18230615

ABSTRACT

HIV protease inhibitors (HIV-PIs) are key components of highly active antiretroviral therapy, but they have been associated with adverse side effects, including partial lipodystrophy and metabolic syndrome. We recently demonstrated that a commonly used HIV-PI, lopinavir, inhibits ZMPSTE24, thereby blocking lamin A biogenesis and leading to an accumulation of prelamin A. ZMPSTE24 deficiency in humans causes an accumulation of prelamin A and leads to lipodystrophy and other disease phenotypes. Thus, an accumulation of prelamin A in the setting of HIV-PIs represents a plausible mechanism for some drug side effects. Here we show, with metabolic labeling studies, that lopinavir leads to the accumulation of the farnesylated form of prelamin A. We also tested whether a new and chemically distinct HIV-PI, darunavir, inhibits ZMPSTE24. We found that darunavir does not inhibit the biochemical activity of ZMPSTE24, nor does it lead to an accumulation of farnesyl-prelamin A in cells. This property of darunavir is potentially attractive. However, all HIV-PIs, including darunavir, are generally administered with ritonavir, an HIV-PI that is used to block the metabolism of other HIV-PIs. Ritonavir, like lopinavir, inhibits ZMPSTE24 and leads to an accumulation of prelamin A.


Subject(s)
HIV Infections/diet therapy , HIV Protease Inhibitors/pharmacology , Membrane Proteins/metabolism , Metalloendopeptidases/metabolism , Nuclear Proteins/metabolism , Protein Precursors/metabolism , Protein Processing, Post-Translational/drug effects , Sulfonamides/pharmacology , Animals , Darunavir , HIV Infections/enzymology , HIV Infections/genetics , HIV Protease Inhibitors/adverse effects , HIV Protease Inhibitors/therapeutic use , Humans , Lamin Type A , Lipodystrophy/chemically induced , Lipodystrophy/enzymology , Lipodystrophy/genetics , Lopinavir , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Metabolic Syndrome/chemically induced , Metabolic Syndrome/enzymology , Metabolic Syndrome/genetics , Metalloendopeptidases/antagonists & inhibitors , Metalloendopeptidases/genetics , Mice , Mice, Knockout , Pyrimidinones/adverse effects , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Ritonavir/adverse effects , Ritonavir/pharmacology , Ritonavir/therapeutic use , Sulfonamides/adverse effects , Sulfonamides/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...