Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 134
Filter
Add more filters










Publication year range
1.
Cells ; 13(5)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38474395

ABSTRACT

Dystroglycan is a ubiquitously expressed heterodimeric cell-surface laminin receptor with roles in cell adhesion, signalling, and membrane stabilisation. More recently, the transmembrane ß-subunit of dystroglycan has been shown to localise to both the nuclear envelope and the nucleoplasm. This has led to the hypothesis that dystroglycan may have a structural role at the nuclear envelope analogous to its role at the plasma membrane. The biochemical fraction of myoblast cells clearly supports the presence of dystroglycan in the nucleus. Deletion of the dystroglycan protein by disruption of the DAG1 locus using CRISPR/Cas9 leads to changes in nuclear size but not overall morphology; moreover, the Young's modulus of dystroglycan-deleted nuclei, as determined by atomic force microscopy, is unaltered. Dystroglycan-disrupted myoblasts are also no more susceptible to nuclear stresses including chemical and mechanical, than normal myoblasts. Re-expression of dystroglycan in DAG1-disrupted myoblasts restores nuclear size without affecting other nuclear parameters.


Subject(s)
Dystroglycans , Laminin , Dystroglycans/metabolism , Laminin/metabolism , Cell Nucleus/metabolism , Cell Membrane/metabolism , Nuclear Envelope/metabolism
2.
PLoS Biol ; 21(5): e3001746, 2023 05.
Article in English | MEDLINE | ID: mdl-37134077

ABSTRACT

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


Subject(s)
Hematologic Diseases , Hematopoiesis, Extramedullary , Neoplasms , Humans , Animals , Mice , Hematopoiesis, Extramedullary/physiology , Leukemia Inhibitory Factor/pharmacology , Interleukin-1alpha/pharmacology , Hematopoiesis
3.
Hum Mol Genet ; 32(2): 177-191, 2023 01 06.
Article in English | MEDLINE | ID: mdl-35925868

ABSTRACT

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


Subject(s)
Laminopathies , Muscle, Striated , Animals , Mice , Microtubule Proteins/metabolism , Nuclear Proteins/metabolism , Membrane Proteins/genetics , Cytoskeleton/genetics , Cytoskeleton/metabolism , Nuclear Matrix/genetics , Microtubules/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Intermediate Filament Proteins/metabolism , Muscle, Striated/metabolism , Laminopathies/metabolism
4.
Hum Mol Genet ; 31(18): 3161-3180, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35567414

ABSTRACT

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


Subject(s)
Pregnancy Proteins , Retroelements , Animals , Anxiety/genetics , Anxiety Disorders/genetics , Female , Genomic Imprinting , Humans , Locus Coeruleus/metabolism , Mice , Neurons/metabolism , Pregnancy , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism , Social Behavior
5.
Stem Cell Reports ; 16(12): 2928-2941, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34767749

ABSTRACT

The immature characteristics and metabolic phenotypes of human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) restrict their applications for disease modeling, drug discovery, and cell-based therapy. Leveraging on the metabolic shifts from glycolysis to fatty acid oxidation as CMs mature, a human hexokinase1-GFP metabolic reporter cell line (H7 HK1-GFP) was generated to facilitate the isolation of fetal or more matured hPSC-CMs. RNA sequencing of fetal versus more matured CMs uncovered a potential role of interferon-signaling pathway in regulating CM maturation. Indeed, IFN-γ-treated CMs resulted in an upregulation of the JAK-STAT pathway, which was found to be associated with increased expression of CM maturation genes, shift from MYH6 to MYH7 expression, and improved sarcomeric structure. Functionally, IFN-γ-treated CMs exhibited a more matured electrophysiological profile, such as increased calcium dynamics and action potential upstroke velocity, demonstrated through calcium imaging and MEA. Expectedly, the functional improvements were nullified with a JAK-STAT inhibitor, ruxolitinib.


Subject(s)
Cell Differentiation , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Janus Kinases/metabolism , Myocytes, Cardiac/cytology , STAT Transcription Factors/metabolism , Signal Transduction , Up-Regulation , CRISPR-Cas Systems/genetics , Cell Differentiation/drug effects , Cell Line , Electrophysiological Phenomena/drug effects , Genes, Reporter , Green Fluorescent Proteins/metabolism , Human Embryonic Stem Cells/drug effects , Humans , Interferon-gamma/metabolism , Interferon-gamma/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects
6.
PLoS Genet ; 17(8): e1009757, 2021 08.
Article in English | MEDLINE | ID: mdl-34449766

ABSTRACT

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


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Mitosis/physiology , Adaptor Proteins, Signal Transducing/physiology , Apoptosis Regulatory Proteins/physiology , Cell Cycle Proteins/metabolism , Cytokinesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/genetics , HeLa Cells , Humans , Protein Transport , Spindle Apparatus/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Nat Commun ; 12(1): 4722, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34354059

ABSTRACT

Mutations in the LaminA gene are a common cause of monogenic dilated cardiomyopathy. Here we show that mice with a cardiomyocyte-specific Lmna deletion develop cardiac failure and die within 3-4 weeks after inducing the mutation. When the same Lmna mutations are induced in mice genetically deficient in the LINC complex protein SUN1, life is extended to more than one year. Disruption of SUN1's function is also accomplished by transducing and expressing a dominant-negative SUN1 miniprotein in Lmna deficient cardiomyocytes, using the cardiotrophic Adeno Associated Viral Vector 9. The SUN1 miniprotein disrupts binding between the endogenous LINC complex SUN and KASH domains, displacing the cardiomyocyte KASH complexes from the nuclear periphery, resulting in at least a fivefold extension in lifespan. Cardiomyocyte-specific expression of the SUN1 miniprotein prevents cardiomyopathy progression, potentially avoiding the necessity of developing a specific therapeutic tailored to treating each different LMNA cardiomyopathy-inducing mutation of which there are more than 450.


Subject(s)
Cardiomyopathy, Dilated/genetics , Lamin Type A/genetics , Lamin Type A/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Animals , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Dependovirus/genetics , Female , Humans , Lamin Type A/deficiency , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Transduction, Genetic
8.
Curr Opin Genet Dev ; 67: 130-141, 2021 04.
Article in English | MEDLINE | ID: mdl-33524904

ABSTRACT

The regulation of genomic function is in part mediated through the physical organization and architecture of the nucleus. Disruption to nuclear organization and architecture is increasingly being recognized by its contribution to many diseases. The LINC complexes - protein structures traversing the nuclear envelope, that physically connect the nuclear interior, and hence the genome, to cytoplasmic cytoskeletal networks are an important component in the physical organization of the genome and its function. This connection, potentially allows for the constant detection of environmental mechanical stimuli, resulting in altered regulation of nuclear architecture and genome function, either directly or via the process of mechanotransduction. Here, we review the influences LINC complexes exert on genome functions and their impact on cellular/organismal health.


Subject(s)
Genome/genetics , Mechanotransduction, Cellular/genetics , Nuclear Proteins/genetics , RNA, Long Noncoding/genetics , Cell Nucleus/genetics , Cell Nucleus/ultrastructure , Cytoplasm/genetics , Cytoplasm/ultrastructure , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Humans , Nuclear Envelope/genetics , Nuclear Envelope/ultrastructure , Nuclear Proteins/ultrastructure , RNA, Long Noncoding/ultrastructure
9.
Annu Rev Genomics Hum Genet ; 21: 263-288, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32428417

ABSTRACT

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


Subject(s)
Cell Nucleus/genetics , Chromatin/chemistry , Chromatin/genetics , Laminopathies/pathology , Animals , Humans , Laminopathies/genetics
10.
Cells ; 9(4)2020 03 31.
Article in English | MEDLINE | ID: mdl-32244403

ABSTRACT

LMNA encodes for Lamin A/C, type V intermediate filaments that polymerize under the inner nuclear membrane to form the nuclear lamina. A small fraction of Lamin A/C, less polymerized, is also found in the nucleoplasm. Lamin A/C functions include roles in nuclear resistance to mechanical stress and gene regulation. LMNA mutations are responsible for a wide variety of pathologies, including Emery-Dreifuss (EDMD) and LMNA-related congenital muscular dystrophies (L-CMD) without clear genotype-phenotype correlations. Both diseases presented with striated muscle disorders although L-CMD symptoms appear much earlier and are more severe. Seeking for pathomechanical differences to explain the severity of L-CMD mutations, we performed an in silico analysis of the UMD-LMNA database and found that L-CMD mutations mainly affect residues involved in Lamin dimer and tetramer stability. In line with this, we found increased nucleoplasmic Lamin A/C in L-CMD patient fibroblasts and mouse myoblasts compared to the control and EDMD. L-CMD myoblasts show differentiation defects linked to their inability to upregulate muscle specific nuclear envelope (NE) proteins expression. NE proteins were mislocalized, leading to misshapen nuclei. We conclude that these defects are due to both the absence of Lamin A/C from the nuclear lamina and its maintenance in the nucleoplasm of myotubes.


Subject(s)
Lamin Type A/deficiency , Lamin Type A/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophy, Emery-Dreifuss/pathology , Severity of Illness Index , Animals , Cells, Cultured , Computer Simulation , Databases, Genetic , Disease Models, Animal , Humans , Lamin Type A/genetics , Mice , Muscle Fibers, Skeletal/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation/genetics , Myoblasts/metabolism , Phenotype
11.
Aging Cell ; 19(3): e13108, 2020 03.
Article in English | MEDLINE | ID: mdl-32087607

ABSTRACT

Hutchinson-Gilford progeria is a premature aging syndrome caused by a truncated form of lamin A called progerin. Progerin expression results in a variety of cellular defects including heterochromatin loss, DNA damage, impaired proliferation and premature senescence. It remains unclear how these different progerin-induced phenotypes are temporally and mechanistically linked. To address these questions, we use a doxycycline-inducible system to restrict progerin expression to different stages of the cell cycle. We find that progerin expression leads to rapid and widespread loss of heterochromatin in G1-arrested cells, without causing DNA damage. In contrast, progerin triggers DNA damage exclusively during late stages of DNA replication, when heterochromatin is normally replicated, and preferentially in cells that have lost heterochromatin. Importantly, removal of progerin from G1-arrested cells restores heterochromatin levels and results in no permanent proliferative impediment. Taken together, these results delineate the chain of events that starts with progerin expression and ultimately results in premature senescence. Moreover, they provide a proof of principle that removal of progerin from quiescent cells restores heterochromatin levels and their proliferative capacity to normal levels.


Subject(s)
DNA Damage/genetics , Heterochromatin/metabolism , Lamin Type A/metabolism , Progeria/metabolism , Signal Transduction/genetics , Aging, Premature/genetics , Aging, Premature/metabolism , Cell Proliferation/genetics , Cells, Cultured , Cellular Senescence/genetics , DNA Damage/drug effects , DNA Replication/genetics , Doxorubicin/pharmacology , Fibroblasts/metabolism , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression , Humans , Lamin Type A/genetics , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Progeria/genetics
12.
Elife ; 82019 11 05.
Article in English | MEDLINE | ID: mdl-31686651

ABSTRACT

Here we show that a major muscle specific isoform of the murine LINC complex protein SUN1 is required for efficient muscle regeneration. The nucleoplasmic domain of the isoform specifically binds to and inhibits Drosha, a key component of the microprocessor complex required for miRNA synthesis. Comparison of the miRNA profiles between wildtype and SUN1 null myotubes identified a cluster of miRNAs encoded by a non-translated retrotransposon-like one antisense (Rtl1as) transcript that are decreased in the WT myoblasts due to SUN1 inhibition of Drosha. One of these miRNAs miR-127 inhibits the translation of the Rtl1 sense transcript, that encodes the retrotransposon-like one protein (RTL1), which is also required for muscle regeneration and is expressed in regenerating/dystrophic muscle. The LINC complex may therefore regulate gene expression during muscle regeneration by controlling miRNA processing. This provides new insights into the molecular pathology underlying muscular dystrophies and how the LINC complex may regulate mechanosignaling.


Subject(s)
Microtubule-Associated Proteins/physiology , Muscles/physiology , RNA, Long Noncoding/genetics , Regeneration , Ribonuclease III/metabolism , Animals , Mice
13.
Curr Protoc Cell Biol ; 84(1): e96, 2019 09.
Article in English | MEDLINE | ID: mdl-31483108

ABSTRACT

Protein-protein interactions (PPIs) add an essential layer of complexity to the information encoded by the genome. Modulation of such interactions is a key feature of most, if not all, cellular activities and allows cells to respond rapidly to both internal and external signals and stimuli. In this respect, the development of the BioID assay to interrogate PPIs within a cellular context represents an important adjunct to the range of tools currently at researchers' disposal. To address some of its current limitations, we devised 2C-BioID, in which biotin ligase and the protein of interest remain as separate entities until induced to associate. This is accomplished using the well-established FKBP-FRB dimerization system (based on the rapamycin-induced binding of FK506 binding protein and FKBP12-rapamycin binding domain.). The design of 2C-BioID ensures that biotin ligase association with the protein of interest occurs only after addition of the rapamycin analogue AP21967. As such, 2C-BioID alleviates potential targeting issues and improves the ability to exclude false positives, thereby refining the specificity of BioID-generated interactomes. © 2019 by John Wiley & Sons, Inc.


Subject(s)
Dimerization , Protein Binding , Protein Interaction Mapping/methods , Tacrolimus Binding Protein 1A/metabolism , Biotinylation , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/metabolism , Genome , Humans , Repressor Proteins/metabolism , Sirolimus/analogs & derivatives , Sirolimus/metabolism , Tacrolimus Binding Protein 1A/genetics
14.
Hum Mol Genet ; 28(15): 2486-2500, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31009944

ABSTRACT

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


Subject(s)
Carrier Proteins/genetics , Heart/physiopathology , Lamin Type A/genetics , Membrane Proteins/genetics , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Emery-Dreifuss/genetics , Mutation , Nuclear Proteins/genetics , Animals , Animals, Newborn , Disease Models, Animal , Female , Haploinsufficiency , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophy, Emery-Dreifuss/physiopathology , Myocardium/metabolism , Myocardium/pathology
15.
iScience ; 10: 40-52, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30500481

ABSTRACT

The modulation of protein-protein interactions (PPIs) is an essential regulatory activity defining diverse cell functions in development and disease. BioID is an unbiased proximity-dependent biotinylation method making use of a biotin-protein ligase fused to a protein of interest and has become an important tool for mapping of PPIs within cellular contexts. We devised an advanced method, 2C-BioID, in which the biotin-protein ligase is kept separate from the protein of interest, until the two are induced to associate by the addition of a dimerizing agent. As proof of principle, we compared the interactomes of lamina-associated polypeptide 2ß (LAP2ß) with those of lamins A and C, using 2C- and conventional BioID. 2C-BioID greatly enhanced data robustness by facilitating the in silico elimination of non-specific interactors as well as overcoming the problems associated with aberrant protein localization. 2C-BioID therefore significantly strengthens the specificity and reliability of BioID-based interactome analysis, by the more stringent exclusion of false-positives and more efficient intracellular targeting.

16.
Gastroenterology ; 154(6): 1625-1629.e8, 2018 05.
Article in English | MEDLINE | ID: mdl-29366840

ABSTRACT

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


Subject(s)
E2F Transcription Factors/physiology , Homeostasis/genetics , Lamin Type A/physiology , Pancreas, Exocrine/metabolism , Retinoblastoma Protein/physiology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics
17.
Cell Mol Gastroenterol Hepatol ; 4(3): 365-383, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28913408

ABSTRACT

BACKGROUND & AIMS: Lamins are nuclear intermediate filament proteins that comprise the major components of the nuclear lamina. Mutations in LMNA, which encodes lamins A/C, cause laminopathies, including lipodystrophy, cardiomyopathy, and premature aging syndromes. However, the role of lamins in the liver is unknown, and it is unclear whether laminopathy-associated liver disease is caused by primary hepatocyte defects or systemic alterations. METHODS: To address these questions, we generated mice carrying a hepatocyte-specific deletion of Lmna (knockout [KO] mice) and characterized the KO liver and primary hepatocyte phenotypes by immunoblotting, immunohistochemistry, microarray analysis, quantitative real-time polymerase chain reaction, and Oil Red O and Picrosirius red staining. RESULTS: KO hepatocytes manifested abnormal nuclear morphology, and KO mice showed reduced body mass. KO mice developed spontaneous male-selective hepatosteatosis with increased susceptibility to high-fat diet-induced steatohepatitis and fibrosis. The hepatosteatosis was associated with up-regulated transcription of genes encoding lipid transporters, lipid biosynthetic enzymes, lipid droplet-associated proteins, and interferon-regulated genes. Hepatic Lmna deficiency led to enhanced signal transducer and activator of transcription 1 (Stat1) expression and blocked growth hormone-mediated Janus kinase 2 (Jak2), signal transducer and activator of transcription 5 (Stat5), and extracellular signal-regulated kinase (Erk) signaling. CONCLUSIONS: Lamin A/C acts cell-autonomously to maintain hepatocyte homeostasis and nuclear shape and buffers against male-selective steatohepatitis by positively regulating growth hormone signaling and negatively regulating Stat1 expression. Lamins are potential genetic modifiers for predisposition to steatohepatitis and liver fibrosis. The microarray data can be found in the Gene Expression Omnibus repository (accession number: GSE93643).

18.
Nat Genet ; 49(9): 1354-1363, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28740264

ABSTRACT

The transcriptional network acting downstream of LIF, WNT and MAPK-ERK to stabilize mouse embryonic stem cells (ESCs) in their naive state has been extensively characterized. However, the upstream factors regulating these three signaling pathways remain largely uncharted. PR-domain-containing proteins (PRDMs) are zinc-finger sequence-specific chromatin factors that have essential roles in embryonic development and cell fate decisions. Here we characterize the transcriptional regulator PRDM15, which acts independently of PRDM14 to regulate the naive state of mouse ESCs. Mechanistically, PRDM15 modulates WNT and MAPK-ERK signaling by directly promoting the expression of Rspo1 (R-spondin1) and Spry1 (Sprouty1). Consistent with these findings, CRISPR-Cas9-mediated disruption of PRDM15-binding sites in the Rspo1 and Spry1 promoters recapitulates PRDM15 depletion, both in terms of local chromatin organization and the transcriptional modulation of these genes. Collectively, our findings uncover an essential role for PRDM15 as a chromatin factor that modulates the transcription of upstream regulators of WNT and MAPK-ERK signaling to safeguard naive pluripotency.


Subject(s)
DNA-Binding Proteins/genetics , Embryonic Stem Cells/metabolism , Gene Expression Regulation , MAP Kinase Signaling System/genetics , Transcription Factors/genetics , Wnt Signaling Pathway/genetics , Animals , Blotting, Western , Cell Line , Cell Self Renewal/genetics , Cells, Cultured , Cellular Reprogramming/genetics , DNA-Binding Proteins/metabolism , Fluorescent Antibody Technique , Gene Expression Profiling/methods , Humans , Induced Pluripotent Stem Cells/metabolism , Mice, Knockout , Mice, Nude , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism
19.
Am J Physiol Lung Cell Mol Physiol ; 313(3): L548-L558, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28522567

ABSTRACT

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


Subject(s)
Epithelial Cells/metabolism , Leukemia Inhibitory Factor/biosynthesis , Myeloid Cells/metabolism , Pneumonia, Bacterial/metabolism , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/pathology , Animals , Bronchoalveolar Lavage Fluid , Cell Line , Escherichia coli/physiology , Female , Hematopoiesis , Leukemia Inhibitory Factor/genetics , Lung/metabolism , Lung/pathology , Macrophages, Alveolar/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor RelA/metabolism
20.
Endocrinology ; 158(6): 1916-1928, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28368537

ABSTRACT

The cytokine leukemia inhibitory factor (LIF) is essential for rendering the uterus receptive for blastocyst implantation. In mice, LIF receptor expression (LIFR) is largely restricted to the uterine luminal epithelium (LE). LIF, secreted from the endometrial glands (GEs), binds to the LIFR, activating the Janus kinase-signal transducer and activation of transcription (STAT) 3 (Jak-Stat3) signaling pathway in the LE. JAK-STAT activation converts the LE to a receptive state so that juxtaposed blastocysts begin to implant. To specifically delete the LIFR in the LE, we derived a line of mice in which Cre recombinase was inserted into the endogenous lactoferrin gene (Ltf-Cre). Lactoferrin expression in the LE is induced by E2, and we demonstrate that Cre recombinase activity is restricted to the LE and GE. To determine the requirement of the LIFR in implantation, we derived an additional mouse line carrying a conditional (floxed) Lifrflx/flx gene. Crossing Ltf-Cre mice with Lifrflx/flx mice generated Lifrflx/Δ:LtfCre/+ females that were overtly normal but infertile. Many of these females, despite repeated matings, did not become pregnant. Unimplanted blastocysts were recovered from the Lifrflx/Δ:LtfCre/+ uteri and, when transferred to wild-type recipients, implanted normally, indicating that uterine receptivity rather than the embryo's competency is compromised. The loss of Lifr results in both the failure for STAT3 to translocate to the LE nuclei and a reduction in the expression of the LIF regulated gene Msx1 that regulates uterine receptivity. These results reveal that uterine expression of the LIFR is essential for embryo implantation and further define the components of the LIF signaling pathway necessary for effective implantation.


Subject(s)
Embryo Implantation/genetics , Embryo Loss/genetics , Leukemia Inhibitory Factor Receptor alpha Subunit/genetics , Uterus/metabolism , Animals , Embryo Loss/metabolism , Embryo Loss/pathology , Embryo, Mammalian , Epithelium/metabolism , Female , Leukemia Inhibitory Factor Receptor alpha Subunit/metabolism , Mice , Mice, Knockout , Organ Specificity/genetics , Pregnancy , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...