Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
PLoS Genet ; 16(8): e1008752, 2020 08.
Article in English | MEDLINE | ID: mdl-32790717

ABSTRACT

Myelin is composed of plasma membrane spirally wrapped around axons and compacted into dense sheaths by myelin-associated proteins. Myelin is elaborated by neuroepithelial derived oligodendrocytes in the central nervous system (CNS) and by neural crest derived Schwann cells in the peripheral nervous system (PNS). While some myelin proteins accumulate in only one lineage, myelin basic protein (Mbp) is expressed in both. Overlapping the Mbp gene is Golli, a transcriptional unit that is expressed widely both within and beyond the nervous system. A super-enhancer domain within the Golli/Mbp locus contains multiple enhancers shown previously to drive reporter construct expression specifically in oligodendrocytes or Schwann cells. In order to determine the contribution of each enhancer to the Golli/Mbp expression program, and to reveal if functional interactions occur among them, we derived mouse lines in which they were deleted, either singly or in different combinations, and relative mRNA accumulation was measured at key stages of early development and at maturity. Although super-enhancers have been shown previously to facilitate interaction among their component enhancers, the enhancers investigated here demonstrated largely additive relationships. However, enhancers demonstrating autonomous activity strictly in one lineage, when missing, were found to significantly reduce output in the other, thus revealing cryptic "stealth" activity. Further, in the absence of a key oligodendrocyte enhancer, Golli accumulation was markedly and uniformly attenuated in all cell types investigated. Our observations suggest a model in which enhancer-mediated DNA-looping and potential super-enhancer properties underlie Golli/Mbp regulatory organization.


Subject(s)
Enhancer Elements, Genetic , Myelin Basic Protein/genetics , Animals , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Neurogenesis , Oligodendroglia/cytology , Oligodendroglia/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Schwann Cells/cytology , Schwann Cells/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism
2.
Sci Rep ; 6: 18701, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26726832

ABSTRACT

Fortilin, a pro-survival molecule, inhibits p53-induced apoptosis by binding to the sequence-specific DNA-binding domain of the tumor suppressor protein and preventing it from transcriptionally activating Bax. Intriguingly, fortilin protects cells against ROS-induced cell death, independent of p53. The signaling pathway through which fortilin protects cells against ROS-induced cell death, however, is unknown. Here we report that fortilin physically interacts with the antioxidant enzyme peroxiredoxin-1 (PRX1), protects it from proteasome-mediated degradation, and keeps it enzymatically active by blocking its deactivating phosphorylation by Mst1, a serine/threonine kinase. At the whole animal level, the liver-specific overexpression of fortilin reduced PRX1 phosphorylation in the liver, enhanced PRX1 activity, and protected the transgenic animals against alcohol-induced, ROS-mediated, liver damage. These data suggest the presence of a novel oxidative-stress-handling pathway where the anti-p53 molecule fortilin augments the peroxidase PRX1 by protecting it against degradation and inactivation of the enzyme. Fortilin-PRX1 interaction in the liver could be clinically exploited further to prevent acute alcohol-induced liver damage in humans.


Subject(s)
Alcohols/adverse effects , Biomarkers, Tumor/metabolism , Liver Diseases/etiology , Liver Diseases/metabolism , Peroxiredoxins/metabolism , Animals , Apoptosis , Biomarkers, Tumor/chemistry , Disease Models, Animal , Enzyme Activation , Gene Expression , Hepatocyte Growth Factor/metabolism , Homeodomain Proteins/metabolism , Liver Diseases/pathology , Mice , Oxidation-Reduction , Peroxiredoxins/chemistry , Peroxiredoxins/genetics , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Proteolysis , Proto-Oncogene Proteins/metabolism , Reactive Oxygen Species/metabolism , Tumor Protein, Translationally-Controlled 1 , Tumor Suppressor Protein p53/metabolism
3.
Hum Mol Genet ; 25(21): 4686-4702, 2016 11 01.
Article in English | MEDLINE | ID: mdl-28173160

ABSTRACT

Progressive forms of multiple sclerosis lead to chronic disability, substantial decline in quality of life and reduced longevity. It is often suggested that they occur independently of inflammation. Here we investigated the disease progression in mouse models carrying PLP1 point mutations previously found in patients displaying clinical features of multiple sclerosis. These mouse models show loss-of-function of PLP1 associated with neuroinflammation; the latter leading to clinically relevant axonal degeneration, neuronal loss and brain atrophy as demonstrated by inactivation of the recombination activating gene 1. Moreover, these pathological hallmarks were substantially amplified when we attenuated immune regulation by inactivation of the programmed cell death-1 gene. Our observations support the view that primary oligodendroglial abnormalities can evoke pathogenically relevant neuroinflammation that drives neurodegeneration, as observed in some forms of multiple sclerosis but also in other, genetically-mediated neurodegenerative disorders of the human nervous system. As many potent immunomodulatory drugs have emerged during the last years, it is tempting to consider immunomodulation as a treatment option not only for multiple sclerosis, but also for so far non-treatable, genetically-mediated disorders of the nervous system accompanied by pathogenic neuroinflammation.


Subject(s)
Multiple Sclerosis/genetics , Mutation , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Animals , Disease Models, Animal , Disease Progression , Female , Humans , Immunologic Factors/genetics , Immunologic Factors/immunology , Inflammation/genetics , Inflammation/metabolism , Male , Mice , Mice, Transgenic , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/immunology , Neurodegenerative Diseases/metabolism
4.
Glia ; 64(1): 175-94, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26507463

ABSTRACT

In mammals, large caliber axons are ensheathed by myelin, a glial specialization supporting axon integrity and conferring accelerated and energy-efficient action potential conduction. Myelin basic protein (MBP) is required for normal myelin elaboration with maximal mbp transcription in oligodendrocytes requiring the upstream M3 enhancer. To further characterize the mechanism regulating mbp transcription, we defined M3 structure/function relationships by evaluating its evolutionary conservation, DNA footprints and the developmental programing conferred in mice by M3 derivatives. Multiple M3 regulatory element combinations were found to drive expression in oligodendrocytes and Schwann cells with a minimal 129 bp sequence conferring expression in oligodendrocytes throughout myelin elaboration, maintenance and repair. Unexpectedly, M3 derivatives conferred markedly different spatial and temporal expression programs thus illuminating striking transcriptional heterogeneity within post-mitotic oligodendrocytes. Finally, one M3 derivative engaged only during primary myelination, not during adult remyelination, demonstrating that transcriptional regulation in the two states is not equivalent.


Subject(s)
Gene Regulatory Networks , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Animals , Avian Proteins/genetics , Avian Proteins/metabolism , Base Sequence , Brain/growth & development , Brain/metabolism , Chickens , Conserved Sequence , Immunohistochemistry , In Situ Hybridization , Male , Mice, Transgenic , Molecular Sequence Data , Mutation , Optic Nerve/growth & development , Optic Nerve/metabolism , Sciatic Nerve/growth & development , Sciatic Nerve/metabolism , Sequence Alignment , Spinal Cord/growth & development , Spinal Cord/metabolism , beta-Galactosidase/metabolism
5.
Mol Ther ; 20(7): 1367-77, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22491214

ABSTRACT

Despite aggressive treatment regimes, glioma remains a largely fatal disease. Current treatment limitations are attributed to the precarious locations within the brain where such tumors grow, their highly infiltrative nature precluding complete resection and lack of specificity among agents capable of attenuating their growth. Here, we show that in vitro, glioma cells of diverse origins internalize a peptide encompassing a tubulin-binding site (TBS) on the neurofilament light protein. The internalized peptide disrupts the microtubule network, inhibits migration and proliferation, and leads to apoptosis. Using an intracerebral transplant model, we show that most, if not all, of these responses to peptide exposure also occur in vivo. Notably, a single intratumor injection significantly attenuates tumor growth, while neither peptide uptake nor downstream consequences are observed elsewhere in the host nervous system. Such preferential uptake suggests that the peptide may have potential as a primary or supplementary glioblastoma treatment modality by exploiting its autonomous microtubule-disrupting activity or engaging its capacity to selectively target glioma cells with other cell-disrupting cargos.


Subject(s)
Apoptosis/drug effects , Glioma/drug therapy , Microtubules/drug effects , Neurofilament Proteins/metabolism , Neurofilament Proteins/pharmacology , Tubulin/metabolism , Animals , Brain/metabolism , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Neurofilament Proteins/therapeutic use , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Protein Binding , Random Allocation , Rats , Rats, Inbred F344
6.
Nat Genet ; 43(9): 902-7, 2011 Aug 14.
Article in English | MEDLINE | ID: mdl-21841778

ABSTRACT

A variant of the PTPN22-encoded Lyp phosphatase (Lyp620W) confers risk for autoimmune disease, but the mechanisms underlying this association remain unclear. We show here that mice expressing the Lyp variant homolog Pep619W manifest thymic and splenic enlargement accompanied by increases in T-cell number, activation and positive selection and in dendritic- and B-cell activation. Although Ptpn22 (Pep) transcript levels were comparable in Pep619W and wild-type Pep619R mice, Pep protein levels were dramatically reduced in the mutant mice, with Pep619W protein being more rapidly degraded and showing greater association with and in vitro cleavage by calpain 1 than Pep619R. Similarly, levels of the Lyp620W variant were decreased in human T and B cells, and its calpain binding and cleavage were increased relative to wild-type Lyp620R. Thus, calpain-mediated degradation with consequently reduced Lyp/Pep expression and lymphocyte and dendritic cell hyperresponsiveness represents a mechanism whereby Lyp620W may increase risk for autoimmune disease.


Subject(s)
Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Dendritic Cells/immunology , Lymphocyte Activation , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Animals , Autoimmune Diseases/pathology , B-Lymphocytes/immunology , Calpain/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Mice , Mice, Mutant Strains , Organ Size , Spleen/immunology , Spleen/pathology , T-Lymphocytes/immunology , Thymus Gland/immunology , Thymus Gland/pathology
7.
Nucleic Acids Res ; 39(18): 7974-91, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21729871

ABSTRACT

In the central nervous system (CNS), myelin is produced from spirally-wrapped oligodendrocyte plasma membrane and, as exemplified by the debilitating effects of inherited or acquired myelin abnormalities in diseases such as multiple sclerosis, it plays a critical role in nervous system function. Myelin sheath production coincides with rapid up-regulation of numerous genes. The complexity of their subsequent expression patterns, along with recently recognized heterogeneity within the oligodendrocyte lineage, suggest that the regulatory networks controlling such genes drive multiple context-specific transcriptional programs. Conferring this nuanced level of control likely involves a large repertoire of interacting transcription factors (TFs). Here, we combined novel strategies of computational sequence analyses with in vivo functional analysis to establish a TF network model of coordinate myelin-associated gene transcription. Notably, the network model captures regulatory DNA elements and TFs known to regulate oligodendrocyte myelin gene transcription and/or oligodendrocyte development, thereby validating our approach. Further, it links to numerous TFs with previously unsuspected roles in CNS myelination and suggests collaborative relationships amongst both known and novel TFs, thus providing deeper insight into the myelin gene transcriptional network.


Subject(s)
Gene Regulatory Networks , Myelin Sheath/genetics , Transcription Factors/metabolism , Animals , Base Sequence , Conserved Sequence , Enhancer Elements, Genetic , Gene Expression Profiling , Gene Expression Regulation , Genes, Reporter , Mice , Myelin Sheath/metabolism , Neuroglia/metabolism , Oligodendroglia/metabolism , Optic Nerve/metabolism , Promoter Regions, Genetic , Prosencephalon/metabolism
8.
Mamm Genome ; 22(11-12): 635-47, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21769670

ABSTRACT

Methylenetetrahydrofolate reductase (MTHFR), a key enzyme in folate metabolism, synthesizes 5-methyltetrahydrofolate, the main circulatory form of folate which is required for maintaining nontoxic levels of homocysteine and providing one-carbon units for methylation. A common 677C â†’ T variant in MTHFR confers mild MTHFR deficiency and has been associated with a number of human disorders, including neural tube defects and vascular disease. Two promoters of Mthfr, designated as upstream and downstream promoters, are located upstream of a transcription start site cluster and have previously demonstrated cell-specific activities. In this study we used a unique approach for targeted, single-copy transgene insertion to generate transgenic mice carrying a Mthfr upstream or Mthfr downstream promoter-reporter construct located 5' to the endogenous Hprt (hypoxanthine-guanine phosphoribosyltransferase) locus. The Mthfr downstream promoter demonstrated activity in the neural tube, neural crest cells, dorsal root ganglia, heart, and endothelial cells of blood vessels in 10.5-days post coitum embryos and placentas. Upstream promoter activity was absent at this developmental stage. Postnatally, both promoters demonstrated activity in the brain stem, hippocampus, and thalamus of 1-week-old brain that became stronger in the adult. The Mthfr upstream promoter also showed activity in the cerebellum and cerebral cortex. Both promoters were active in male reproductive tissues, including 1-week-old epididymides, and there was upstream promoter-specific activity in the adult testis. Our investigation of Mthfr regulation in an in vivo mouse model revealed temporal- and tissue-specific regulation that supports important roles for MTHFR in the developing embryo, and in postnatal brain and male reproductive tissues.


Subject(s)
Embryo, Mammalian/metabolism , Homocystinuria/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Muscle Spasticity/metabolism , Promoter Regions, Genetic , Animals , Blood Vessels/metabolism , Central Nervous System/metabolism , Female , Genotype , Male , Methylenetetrahydrofolate Reductase (NADPH2)/deficiency , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Placenta/metabolism , Polymorphism, Single Nucleotide , Pregnancy , Psychotic Disorders/metabolism , Testis/metabolism
9.
Development ; 138(7): 1329-37, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21385763

ABSTRACT

Schwann cells elaborate myelin sheaths around axons by spirally wrapping and compacting their plasma membranes. Although actin remodeling plays a crucial role in this process, the effectors that modulate the Schwann cell cytoskeleton are poorly defined. Here, we show that the actin cytoskeletal regulator, neural Wiskott-Aldrich syndrome protein (N-WASp), is upregulated in myelinating Schwann cells coincident with myelin elaboration. When N-WASp is conditionally deleted in Schwann cells at the onset of myelination, the cells continue to ensheath axons but fail to extend processes circumferentially to elaborate myelin. Myelin-related gene expression is also severely reduced in the N-WASp-deficient cells and in vitro process and lamellipodia formation are disrupted. Although affected mice demonstrate obvious motor deficits these do not appear to progress, the mutant animals achieving normal body weights and living to advanced age. Our observations demonstrate that N-WASp plays an essential role in Schwann cell maturation and myelin formation.


Subject(s)
Cytoskeleton/metabolism , Myelin Sheath/metabolism , Peripheral Nerves/metabolism , Schwann Cells/metabolism , Wiskott-Aldrich Syndrome Protein, Neuronal/metabolism , Animals , Axons/metabolism , Blotting, Western , Cells, Cultured , Cytoskeleton/genetics , Fluorescent Antibody Technique , Gait/genetics , Gene Expression , Mice , Mice, Knockout , Myelin Sheath/genetics , Reverse Transcriptase Polymerase Chain Reaction , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
10.
Nucleic Acids Res ; 39(7): 2548-58, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21131280

ABSTRACT

Multiple regulatory modules contribute to the complex expression programs realized by many loci. Although long thought of as isolated components, recent studies demonstrate that such regulatory sequences can physically associate with promoters and with each other and may localize to specific sub-nuclear transcription factories. These associations provide a substrate for putative interactions and have led to the suggested existence of a transcriptional interactome. Here, using a controlled strategy of transgenesis, we analyzed the functional consequences of regulatory sequence interaction within the myelin basic protein (mbp) locus. Interactions were revealed through comparisons of the qualitative and quantitative expression programs conferred by an allelic series of 11 different enhancer/inter-enhancer combinations ligated to a common promoter/reporter gene. In a developmentally contextual manner, the regulatory output of all modules changed markedly in the presence of other sequences. Predicted by transgene expression programs, deletion of one such module from the endogenous locus reduced oligodendrocyte expression levels but unexpectedly, also attenuated expression of the overlapping golli transcriptional unit. These observations support a regulatory architecture that extends beyond a combinatorial model to include frequent interactions capable of significantly modulating the functions conferred through regulatory modules in isolation.


Subject(s)
Enhancer Elements, Genetic , Myelin Basic Protein/genetics , Transcription Factors/genetics , Animals , Gene Silencing , Genetic Loci , Mice , Mice, Knockout , Mice, Transgenic , Myelin Basic Protein/metabolism , Myelin Sheath/physiology , Oligodendroglia/metabolism , Promoter Regions, Genetic , Schwann Cells/metabolism , Transcription Factors/metabolism , Transcription, Genetic
11.
Glia ; 58(1): 29-42, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19455714

ABSTRACT

Axons are linked to induction of myelination during development and to the maintenance of myelin and myelinated tracts in the adult CNS. Currently, it is unknown whether and how axonal plasticity in adult CNS impacts the myelinating cells and their precursors. In this article, we report that newly formed axonal sprouts are able to induce a protracted myelination response in adult CNS. We show that newly formed axonal sprouts, induced by lesion of the entorhino-hippocampal perforant pathway, have the ability to induce a myelination response in stratum radiatum and lucidum CA3. The lesion resulted in significant recruitment of newly formed myelinating cells, documented by incorporation of the proliferation marker bromodeoxyuridine into chondroitin sulphate NG2 expressing cells in stratum radiatum and lucidum CA3 early after lesion, and the occurrence of a 28% increase in the number of oligodendrocytes, of which some had incorporated bromodeoxyuridine, 9 weeks post-lesion. Additionally, a marked increase (41%) in myelinated fibres was detected in silver stained sections. Interestingly, these apparently new fibres achieved the same axon diameter as unlesioned mice but myelin thickness remained thinner than normal, suggesting that the sprouting axons in stratum radiatum and lucidum CA3 were not fully myelinated 9 weeks after lesion. Our combined results show that sprouting axons provide a strong stimulus to oligodendrocyte lineage cells to engage actively in the myelination processes in the adult CNS.


Subject(s)
Axons/physiology , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/metabolism , Neuronal Plasticity/physiology , Oligodendroglia/metabolism , Animals , Antigens/metabolism , Axons/ultrastructure , Axotomy/methods , CD11b Antigen/metabolism , Female , Hippocampus/injuries , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Myelin Basic Protein/genetics , Myelin Sheath/ultrastructure , Nerve Fibers, Myelinated/ultrastructure , Nerve Regeneration/genetics , Neuronal Plasticity/genetics , Oligodendroglia/ultrastructure , Perforant Pathway/injuries , Perforant Pathway/metabolism , Proteoglycans/metabolism , Silver Staining/methods , Statistics, Nonparametric , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
12.
J Neurosci ; 29(35): 11043-54, 2009 Sep 02.
Article in English | MEDLINE | ID: mdl-19726663

ABSTRACT

Neurofilaments assemble from three intermediate-filament proteins, contribute to the radial growth of axons, and are exceptionally stable. Microtubules are dynamic structures that assemble from tubulin dimers to support intracellular transport of molecules and organelles. We show here that neurofilaments, and other intermediate-filament proteins, contain motifs in their N-terminal domains that bind unassembled tubulin. Peptides containing such motifs inhibit the in vitro polymerization of microtubules and can be taken up by cultured cells in which they disrupt microtubules leading to altered cell shapes and an arrest of division. In transgenic mice in which neurofilaments are withheld from the axonal compartment, axonal tubulin accumulation is normal but microtubules assemble in excessive numbers. These observations suggest a model in which axonal neurofilaments modulate local microtubule assembly. This capacity also suggests novel mechanisms through which inherited or acquired disruptions in intermediate filaments might contribute to pathogenesis in multiple conditions.


Subject(s)
Neurofilament Proteins/metabolism , Peptide Fragments/metabolism , Tubulin Modulators/metabolism , Tubulin/metabolism , Amino Acid Sequence , Animals , Brain/metabolism , Brain/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , Protein Binding/physiology , Tubulin/physiology
13.
PLoS One ; 3(12): e3940, 2008.
Article in English | MEDLINE | ID: mdl-19081794

ABSTRACT

Loss of the PTEN tumor suppressor is a common occurrence in human prostate cancer, particularly in advanced disease. In keeping with its role as a pivotal upstream regulator of the phosphatidylinositol 3-kinase signaling pathway, experimentally-induced deletion of Pten in the murine prostate invariably results in neoplasia. However, and unlike humans where prostate tumorigenesis likely evolves over decades, disease progression in the constitutively Pten deficient mouse prostate is relatively rapid, culminating in invasive cancer within several weeks post-puberty. Given that the prostate undergoes rapid androgen-dependent growth at puberty, and that Pten excisions during this time might be especially tumorigenic, we hypothesized that delaying prostate-specific Pten deletions until immediately after puberty might alter the pace of tumorigenesis. To this end we generated mice with a tamoxifen-inducible Cre recombinase transgene enabling temporal control over prostate-specific gene alterations. This line was then interbred with mice carrying floxed Pten alleles. Despite evidence of increased Akt/mTOR/S6K axis activity at early time points in Pten-deficient epithelial cells, excisions induced in the post-pubertal (6 wk-old) prostate yielded gradual acquisition of a range of lesions. These progressed from pre-malignant changes (nuclear atypia, focal hyperplasia) and low grade prostatic intraepithelial neoplasia (PIN) at 16-20 wks post-tamoxifen exposure, to overtly malignant lesions by approximately 1 yr of age, characterized by high-grade PIN and microinvasive carcinoma. In contrast, when Pten excisions were triggered in the pre-pubertal (2 week-old) prostate, neoplasia evolved over a more abbreviated time-frame, with a spectrum of premalignant lesions, as well as overt PIN and microinvasive carcinoma by 10-12 wks post-tamoxifen exposure. These results indicate that the developmental stage at which Pten deletions are induced dictates the pace of PIN development.


Subject(s)
Gene Deletion , Genes, Tumor Suppressor , PTEN Phosphohydrolase/genetics , Prostatic Intraepithelial Neoplasia/enzymology , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Androgen-Binding Protein/genetics , Animals , Apoptosis , Arrestins/metabolism , Cell Proliferation , Crosses, Genetic , Disease Progression , Epithelium/enzymology , Epithelium/pathology , Female , Humans , Integrases/metabolism , Male , Mice , Neoplasm Invasiveness , PTEN Phosphohydrolase/deficiency , Phosphatidylinositol 3-Kinases/metabolism , Precancerous Conditions/drug therapy , Precancerous Conditions/enzymology , Precancerous Conditions/genetics , Precancerous Conditions/pathology , Prostatic Intraepithelial Neoplasia/drug therapy , Prostatic Intraepithelial Neoplasia/genetics , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Rats , Ribosomal Protein S6/metabolism , Tamoxifen/analogs & derivatives , Tamoxifen/therapeutic use , Time Factors , Up-Regulation , beta-Arrestins
14.
Pharmacogenet Genomics ; 18(7): 577-89, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18551038

ABSTRACT

OBJECTIVE: Folates provide one-carbon units for nucleotide synthesis and methylation reactions. A common polymorphism (677C-->T) in methylenetetrahydrofolate reductase (MTHFR) encodes an enzyme with reduced activity. Response to the antifolate methotrexate (MTX) may be modified in 677TT individuals because MTHFR converts nonmethylated folates, used for thymidine and purine synthesis, to 5-methyltetrahydrofolate, used in homocysteine remethylation to methionine. To study potential interactions between MTHFR activity and MTX, we examined the impact of decreased and increased MTHFR expression on MTX response in mice. METHODS: Mthfr-deficient (Mthfr and Mthfr) and wild-type (Mthfr) mice were injected with MTX or saline and assessed for hematological parameters (hematocrit, hemoglobin, red, and white blood cell numbers), plasma homocysteine, nephrotoxicity, hepatotoxicity, and splenic 2'-deoxyuridine 5'-triphosphate/2'-deoxythymidine 5'-triphosphate ratios. MTHFR-overexpressing transgenic mice (MTHFR-Tg) were generated, metabolites and folate distributions were measured, and response to MTX was assessed. RESULTS: MTX-treated Mthfr and Mthfr mice displayed hyperhomocysteinemia and decreased hematocrit, hemoglobin, and red blood cell numbers compared with wild-type animals. Mthfr mice also showed increased nephrotoxicity and hepatotoxicity. MTHFR-Tg mice were generated and confirmed to have increased levels of MTHFR with altered distributions of folate and thiols in a tissue-specific manner. After MTX treatment, MTHFR-Tg mice exhibited the same decreases in hematological parameters as Mthfr-deficient mice, and significantly decreased thymidine synthesis (higher 2'-deoxyuridine 5'-triphosphate/2'-deoxythymidine 5'-triphosphate ratios) compared with wild-type mice, but they were protected from MTX-induced hyperhomocysteinemia. CONCLUSION: Underexpression and overexpression of Mthfr/MTHFR increase MTX-induced myelosuppression but have distinct effects on plasma homocysteine and nephrotoxicity. Pharmacogenetic analysis of polymorphisms in folate-dependent enzymes may be useful in optimization of MTX therapy.


Subject(s)
Immunosuppression Therapy , Methotrexate/pharmacology , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Single Nucleotide , Animals , Cells, Cultured , Female , Gene Expression/physiology , Homocysteine/blood , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/pharmacology , Kidney Diseases/chemically induced , Male , Methotrexate/adverse effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Models, Biological
15.
Genesis ; 46(4): 229-34, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18395839

ABSTRACT

Employing the Hprt locus as the site for targeted transgenesis we have developed mice expressing the tamoxifen-inducible Cre-ER(T2) fusion protein under the control of the ARR2-rat probasin promoter. This system enables external control over the timing of prostate epithelial cell-specific gene alterations. Using both the ROSA26-lacZ and ROSA26-EYFP reporter strains to monitor recombinase activity, Cre-ER(T2) was found to be specifically expressed in the prostatic epithelium and was strictly tamoxifen dependent. This strain thus allows precise control over the timing of gene alterations in the mouse prostate, enabling analyses of the phenotypic consequences of gene alterations in mice of any age. It also provides an ideal platform to study the impact of environmental, hormonal, and age-related factors on prostate tumorigenesis. This latter feature will be of particular value given the paucity of murine models that accurately mimic the late onset and prolonged natural history of human prostate cancer.


Subject(s)
Epithelium/metabolism , Gene Transfer Techniques , Prostate/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/genetics , Rats
16.
J Neurosci ; 27(36): 9573-84, 2007 Sep 05.
Article in English | MEDLINE | ID: mdl-17804618

ABSTRACT

In the vertebrate nervous system, axon calibers correlate positively with myelin sheath dimensions and electrophysiological parameters including action potential amplitude and conduction velocity. Neurofilaments, a prominent component of the neuronal cytoskeleton, are required by axons to support their normal radial growth. To distinguish between fiber features that arise in response to absolute axon caliber and those that are under autonomous control, we investigated transgenic mice in which neurofilaments are sequestered in neuronal cell bodies. The neurofilament deficient axons in such mice achieve mature calibers only 50% of normal and have altered conduction properties. We show here that this primary axonal defect also induces multiple changes in myelin sheath composition and radial dimensions. Remarkably, other fundamental fiber features, including internodal spacing and the architecture and composition of nodes of Ranvier, remain unaltered. Thus, many fiber characteristics are controlled through mechanisms operating independently of absolute axon caliber and the neurofilament cytoskeleton.


Subject(s)
Axons/metabolism , Neurofilament Proteins/metabolism , Neurons/metabolism , Ranvier's Nodes/metabolism , Ranvier's Nodes/ultrastructure , Animals , Axons/ultrastructure , Cells, Cultured , Central Nervous System/physiology , Central Nervous System/ultrastructure , Lac Operon , Mice , Mice, Transgenic , Myelin Sheath/chemistry , Myelin Sheath/metabolism , Nerve Fibers, Myelinated/chemistry , Nerve Fibers, Myelinated/physiology , Nerve Fibers, Myelinated/ultrastructure , Neural Conduction/genetics , Neural Conduction/physiology , Neurofilament Proteins/genetics , Neurofilament Proteins/ultrastructure , Neurons/ultrastructure , Peripheral Nervous System/physiology , Peripheral Nervous System/ultrastructure , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Transgenes
17.
Cancer Res ; 66(19): 9492-501, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-17018605

ABSTRACT

The p75 CCAAT-displacement protein/Cut homeobox (CDP/Cux) isoform was previously reported to be overexpressed in human breast cancers. To investigate its oncogenic potential, we engineered two transgenic mouse lines expressing p75 CDP/Cux under the control of the mouse mammary tumor virus-long terminal repeat. The FVB strain of mouse is generally used in the generation of mouse models for breast cancer. The transgene was introduced into the hprt locus of 129/Ola embryonic stem cells and, following germ line passage, was backcrossed onto the FVB and C57BL/6 mouse strains. Here, we describe the phenotype of p75 CDP/Cux transgenic virgin female mice of the first backcross generations. We report that after a long latency period, approximately 33% of mice from two independent transgenic lines and from backcrosses into either the FVB or the C57BL/6 strains succumbed to a similar disease characterized by splenomegaly, hepatomegaly, and frequent infiltration of leukocytes into nonhematopoietic organs like the kidneys and lungs. Although an excess of B or T cells was observed in three diseased mice, in 17 other cases, histologic and flow cytometry analyses revealed the expansion of a population of neutrophils in the blood, spleen, and bone marrow. The increase in neutrophils correlated with signs of anemia and thrombocytopenia, whereas there was no indication of a reactive process. Therefore, p75 CDP/Cux transgenic mice displayed heightened susceptibility to a disease defined as a myeloproliferative disease-like myeloid leukemia. These results indicate that the overexpression of p75 CDP/Cux could alter homeostasis in the hematopoietic compartment.


Subject(s)
Homeodomain Proteins/physiology , Leukemia, Myeloid/genetics , Myeloproliferative Disorders/genetics , Nuclear Proteins/physiology , Repressor Proteins/physiology , Animals , Cell Line , Crosses, Genetic , Disease Models, Animal , Hematopoiesis/physiology , Hepatomegaly/genetics , Hepatomegaly/pathology , Homeodomain Proteins/genetics , Homeostasis , Humans , Leukemia, Myeloid/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Nude , Mice, Transgenic , Myeloproliferative Disorders/pathology , Nuclear Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/physiology , Recombinant Fusion Proteins/physiology , Repressor Proteins/genetics , Splenomegaly/genetics , Splenomegaly/pathology , Terminal Repeat Sequences/genetics , Transcription Factors
18.
J Neurosci ; 26(39): 10006-19, 2006 Sep 27.
Article in English | MEDLINE | ID: mdl-17005864

ABSTRACT

Alpha-internexin, a neuronal intermediate filament protein implicated in neurodegenerative disease, coexists with the neurofilament (NF) triplet proteins (NF-L, NF-M, and NF-H) but has an unknown function. The earlier peak expression of alpha-internexin than the triplet during brain development and its ability to form homopolymers, unlike the triplet, which are obligate heteropolymers, have supported a widely held view that alpha-internexin and neurofilament triplet form separate filament systems. Here, we demonstrate, however, that despite a postnatal decline in expression, alpha-internexin is as abundant as the triplet in the adult CNS and exists in a relatively fixed stoichiometry with these subunits. Alpha-internexin exhibits transport and turnover rates identical to those of triplet proteins in optic axons and colocalizes with NF-M on single neurofilaments by immunogold electron microscopy. Alpha-internexin also coassembles with all three neurofilament proteins into a single network of filaments in quadruple-transfected SW13vim(-) cells. Genetically deleting NF-M alone or together with NF-H in mice dramatically reduces alpha-internexin transport and content in axons throughout the CNS. Moreover, deleting alpha-internexin potentiates the effects of NF-M deletion on NF-H and NF-L transport. Finally, overexpressing a NF-H-LacZ fusion protein in mice induces alpha-internexin and neurofilament triplet to aggregate in neuronal perikarya and greatly reduces their transport and content selectively in axons. Our data show that alpha-internexin and the neurofilament proteins are functionally interdependent. The results strongly support the view that alpha-internexin is a fourth subunit of neurofilaments in the adult CNS, providing a basis for its close relationship with neurofilaments in CNS diseases associated with neurofilament accumulation.


Subject(s)
Axons/chemistry , Intermediate Filament Proteins/physiology , Intermediate Filaments/chemistry , Neurofilament Proteins/physiology , Animals , Axons/ultrastructure , Crosses, Genetic , Female , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/deficiency , Intermediate Filament Proteins/genetics , Intermediate Filament Proteins/ultrastructure , Intermediate Filaments/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microscopy, Fluorescence , Microscopy, Immunoelectron , Multiprotein Complexes , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurofilament Proteins/analysis , Neurofilament Proteins/deficiency , Neurofilament Proteins/genetics , Neurofilament Proteins/ultrastructure , Protein Interaction Mapping , Protein Transport , Rats , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/physiology , Retinal Ganglion Cells/chemistry , Retinal Ganglion Cells/ultrastructure , Spinal Cord/chemistry , Spinal Cord/ultrastructure , Structure-Activity Relationship , Transfection
19.
Glia ; 54(2): 105-15, 2006 Aug 01.
Article in English | MEDLINE | ID: mdl-16718683

ABSTRACT

Proliferation of the adult NG2-expressing oligodendrocyte precursor cells has traditionally been viewed as a remyelination response ensuing from destruction of myelin and oligodendrocytes, and not to the axonal pathology that is also a characteristic of demyelinating disease. To better understand the response of the NG2+ cells to the different components of demyelinating pathology, we investigated the response of adult NG2+ cells to axonal degeneration in the absence of primary myelin or oligodendrocyte pathology. Axonal degeneration was induced in the hippocampal dentate gyrus of adult mice by transection of the entorhino-dentate perforant path projection. The acutely induced degeneration of axons and terminals resulted in a prompt response of NG2+ cells, consisting of morphological transformation, cellular proliferation, and upregulation of NG2 expression days 2-3 after surgery. This was followed by a reduction of cellular NG2 expression to subnormal levels from day 5 to 7 and reappearance of normal appearing NG2+ cells from day 10. Mice that had received repeated injections of bromodeoxyuridine from 24 to 72 h after surgery contained significant numbers of bromodeoxyuridine-incorporating oligodendrocytes in the areas with axonal degeneration at day 7. The results suggest that axonal degeneration induces a unique sequence of changes of NG2+ cells and that a subpopulation of the newly generated NG2+ cells differentiate into oligodendrocytes.


Subject(s)
Antigens/biosynthesis , Axons/metabolism , Axons/pathology , Cell Differentiation/physiology , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Oligodendroglia/metabolism , Proteoglycans/biosynthesis , Animals , Antigens/genetics , Cell Differentiation/genetics , Cell Proliferation , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/genetics , Oligodendroglia/cytology , Proteoglycans/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...