Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
PLoS One ; 17(4): e0261544, 2022.
Article in English | MEDLINE | ID: mdl-35404973

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs; Batten disease) are fatal, mainly childhood, inherited neurodegenerative lysosomal storage diseases. Sheep affected with a CLN6 form display progressive regionally defined glial activation and subsequent neurodegeneration, indicating that neuroinflammation may be causative of pathogenesis. In this study, aggregation chimeras were generated from homozygous unaffected normal and CLN6 affected sheep embryos, resulting in seven chimeric animals with varied proportions of normal to affected cells. These sheep were classified as affected-like, recovering-like or normal-like, based on their cell-genotype ratios and their clinical and neuropathological profiles. Neuropathological examination of the affected-like animals revealed intense glial activation, prominent storage body accumulation and severe neurodegeneration within all cortical brain regions, along with vision loss and decreasing intracranial volumes and cortical thicknesses consistent with ovine CLN6 disease. In contrast, intercellular communication affecting pathology was evident at both the gross and histological level in the normal-like and recovering-like chimeras, resulting in a lack of glial activation and rare storage body accumulation in only a few cells. Initial intracranial volumes of the recovering-like chimeras were below normal but progressively recovered to about normal by two years of age. All had normal cortical thicknesses, and none went blind. Extended neurogenesis was evident in the brains of all the chimeras. This study indicates that although CLN6 is a membrane bound protein, the consequent defect is not cell intrinsic. The lack of glial activation and inflammatory responses in the normal-like and recovering-like chimeras indicate that newly generated cells are borne into a microenvironment conducive to maturation and survival.


Subject(s)
Neuronal Ceroid-Lipofuscinoses , Sheep Diseases , Animals , Brain/metabolism , Chimera/metabolism , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Sheep , Sheep Diseases/pathology
2.
Cell Rep ; 25(7): 1912-1923.e9, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30428357

ABSTRACT

We and others have recently reported that the SMC protein Smchd1 is a regulator of chromosome conformation. Smchd1 is critical for the structure of the inactive X chromosome and at autosomal targets such as the Hox genes. However, it is unknown how Smchd1 is recruited to these sites. Here, we report that Smchd1 localizes to the inactive X via the Xist-HnrnpK-PRC1 (polycomb repressive complex 1) pathway. Contrary to previous reports, Smchd1 does not bind Xist or other RNA molecules with any specificity. Rather, the localization of Smchd1 to the inactive X is H2AK119ub dependent. Following perturbation of this interaction, Smchd1 is destabilized, which has consequences for gene silencing genome-wide. Our work adds Smchd1 to the PRC1 silencing pathway for X chromosome inactivation.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Heterogeneous-Nuclear Ribonucleoprotein K/metabolism , Polycomb Repressive Complex 1/metabolism , RNA, Long Noncoding/metabolism , X Chromosome Inactivation/genetics , Animals , Base Sequence , Cell Differentiation , Female , Genome , Histones/metabolism , Lysine/metabolism , Mice , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Oligonucleotides/metabolism , Protein Transport
3.
Nat Struct Mol Biol ; 25(9): 766-777, 2018 09.
Article in English | MEDLINE | ID: mdl-30127357

ABSTRACT

The regulation of higher-order chromatin structure is complex and dynamic, and a full understanding of the suite of mechanisms governing this architecture is lacking. Here, we reveal the noncanonical SMC protein Smchd1 to be a novel regulator of long-range chromatin interactions in mice, and we add Smchd1 to the canon of epigenetic proteins required for Hox-gene regulation. The effect of losing Smchd1-dependent chromatin interactions has varying outcomes that depend on chromatin context. At autosomal targets transcriptionally sensitive to Smchd1 deletion, we found increased short-range interactions and ectopic enhancer activation. In contrast, the inactive X chromosome was transcriptionally refractive to Smchd1 ablation, despite chromosome-wide increases in short-range interactions. In the inactive X, we observed spreading of trimethylated histone H3 K27 (H3K27me3) domains into regions not normally decorated by this mark. Together, these data suggest that Smchd1 is able to insulate chromatin, thereby limiting access to other chromatin-modifying proteins.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/physiology , Genes, Homeobox , Multigene Family , X Chromosome , Animals , Chromosomal Proteins, Non-Histone/genetics , Enhancer Elements, Genetic , Gene Deletion , Gene Silencing , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Proc Natl Acad Sci U S A ; 112(27): E3535-44, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26091879

ABSTRACT

Structural maintenance of chromosomes flexible hinge domain containing 1 (Smchd1) is an epigenetic repressor with described roles in X inactivation and genomic imprinting, but Smchd1 is also critically involved in the pathogenesis of facioscapulohumeral dystrophy. The underlying molecular mechanism by which Smchd1 functions in these instances remains unknown. Our genome-wide transcriptional and epigenetic analyses show that Smchd1 binds cis-regulatory elements, many of which coincide with CCCTC-binding factor (Ctcf) binding sites, for example, the clustered protocadherin (Pcdh) genes, where we show Smchd1 and Ctcf act in opposing ways. We provide biochemical and biophysical evidence that Smchd1-chromatin interactions are established through the homodimeric hinge domain of Smchd1 and, intriguingly, that the hinge domain also has the capacity to bind DNA and RNA. Our results suggest Smchd1 imparts epigenetic regulation via physical association with chromatin, which may antagonize Ctcf-facilitated chromatin interactions, resulting in coordinated transcriptional control.


Subject(s)
Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Epigenesis, Genetic , Genome , Animals , Binding Sites/genetics , Blotting, Western , Brain/cytology , Brain/embryology , Brain/metabolism , CCCTC-Binding Factor , Cells, Cultured , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Female , Gene Expression Regulation, Developmental , Genomic Imprinting , Histones/metabolism , Male , Methylation , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Neural Stem Cells/metabolism , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptome/genetics
5.
J Neuroinflammation ; 10: 97, 2013 Jul 30.
Article in English | MEDLINE | ID: mdl-23899308

ABSTRACT

BACKGROUND: The neuronal ceroid lipofuscinoses (NCLs; or Batten disease) are fatal inherited human neurodegenerative diseases affecting an estimated 1:12,500 live births worldwide. They are caused by mutations in at least 11 different genes. Currently, there are no effective treatments. Progress into understanding pathogenesis and possible therapies depends on studying animal models. The most studied animals are the CLN6 South Hampshire sheep, in which the course of neuropathology closely follows that in affected children. Neurodegeneration, a hallmark of the disease, has been linked to neuroinflammation and is consequent to it. Activation of astrocytes and microglia begins prenatally, starting from specific foci associated with the later development of progressive cortical atrophy and the development of clinical symptoms, including the occipital cortex and blindness. Both neurodegeneration and neuroinflammation generalize and become more severe with increasing age and increasing clinical severity. The purpose of this study was to determine if chronic administration of an anti-inflammatory drug, minocycline, from an early age would halt or reverse the development of disease. METHOD: Minocycline, a tetracycline family antibiotic with activity against neuroinflammation, was tested by chronic oral administration of 25 mg minocycline/kg/day to presymptomatic lambs affected with CLN6 NCL at 3 months of age to 14 months of age, when clinical symptoms are obvious, to determine if this would suppress neuroinflammation or disease progression. RESULTS: Minocycline was absorbed without significant rumen biotransformation to maintain pharmacological concentrations of 1 µM in plasma and 400 nM in cerebrospinal fluid, but these did not result in inhibition of microglial activation or astrocytosis and did not change the neuronal loss or clinical course of the disease. CONCLUSION: Oral administration is an effective route for drug delivery to the central nervous system in large animals, and model studies in these animals should precede highly speculative procedures in humans. Minocycline does not inhibit a critical step in the neuroinflammatory cascade in this form of Batten disease. Identification of the critical steps in the neuroinflammatory cascade in neurodegenerative diseases, and targeting of specific drugs to them, will greatly increase the likelihood of success.


Subject(s)
Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/pharmacokinetics , Inflammation/pathology , Minocycline/pharmacology , Minocycline/pharmacokinetics , Neuronal Ceroid-Lipofuscinoses/metabolism , Animals , Anti-Bacterial Agents/cerebrospinal fluid , Atrophy , Brain/metabolism , Brain/pathology , Chromatography, High Pressure Liquid , Disease Progression , Female , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Growth/drug effects , Image Processing, Computer-Assisted , Inflammation/chemically induced , Liver Function Tests , Macrophage Activation/drug effects , Male , Minocycline/cerebrospinal fluid , Neurodegenerative Diseases/pathology , Neuroglia/drug effects , Neuronal Ceroid-Lipofuscinoses/pathology , Sheep
6.
Epigenetics Chromatin ; 6(1): 19, 2013 Jul 02.
Article in English | MEDLINE | ID: mdl-23819640

ABSTRACT

BACKGROUND: Smchd1 is an epigenetic modifier essential for X chromosome inactivation: female embryos lacking Smchd1 fail during midgestational development. Male mice are less affected by Smchd1-loss, with some (but not all) surviving to become fertile adults on the FVB/n genetic background. On other genetic backgrounds, all males lacking Smchd1 die perinatally. This suggests that, in addition to being critical for X inactivation, Smchd1 functions to control the expression of essential autosomal genes. RESULTS: Using genome-wide microarray expression profiling and RNA-seq, we have identified additional genes that fail X inactivation in female Smchd1 mutants and have identified autosomal genes in male mice where the normal expression pattern depends upon Smchd1. A subset of genes in the Snrpn imprinted gene cluster show an epigenetic signature and biallelic expression consistent with loss of imprinting in the absence of Smchd1. In addition, single nucleotide polymorphism analysis of expressed genes in the placenta shows that the Igf2r imprinted gene cluster is also disrupted, with Slc22a3 showing biallelic expression in the absence of Smchd1. In both cases, the disruption was not due to loss of the differential methylation that marks the imprint control region, but affected genes remote from this primary imprint controlling element. The clustered protocadherins (Pcdhα, Pcdhß, and Pcdhγ) also show altered expression levels, suggesting that their unique pattern of random combinatorial monoallelic expression might also be disrupted. CONCLUSIONS: Smchd1 has a role in the expression of several autosomal gene clusters that are subject to monoallelic expression, rather than being restricted to functioning uniquely in X inactivation. Our findings, combined with the recent report implicating heterozygous mutations of SMCHD1 as a causal factor in the digenically inherited muscular weakness syndrome facioscapulohumeral muscular dystrophy-2, highlight the potential importance of Smchd1 in the etiology of diverse human diseases.

7.
Mamm Genome ; 24(5-6): 206-17, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23636699

ABSTRACT

Observations of inherited phenotypes that cannot be explained solely through genetic inheritance are increasing. Evidence points to transmission of non-DNA molecules in the gamete as mediators of the phenotypes. However, in most cases it is unclear what the molecules are, with DNA methylation, chromatin proteins, and small RNAs being the most prominent candidates. From a screen to generate novel mouse mutants of genes involved in epigenetic reprogramming, we produced a DNA methyltransferase 3b allele that is missing exon 13. Mice that are homozygous for the mutant allele have smaller stature and reduced viability, with particularly high levels of female post-natal death. Reduced DNA methylation was also detected at telocentric repeats and the X-linked Hprt gene. However, none of the abnormal phenotypes or DNA methylation changes worsened with multiple generations of homozygous mutant inbreeding. This suggests that in our model the abnormalities are reset each generation and the processes of transgenerational epigenetic reprogramming are effective in preventing their inheritance.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , Mice/genetics , Alleles , Animals , Base Sequence , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Epigenesis, Genetic , Exons , Female , Homozygote , Male , Mice/growth & development , Mice/metabolism , Mice, Transgenic , Molecular Sequence Data , Pedigree , DNA Methyltransferase 3B
8.
BMC Cancer ; 12: 252, 2012 Jun 18.
Article in English | MEDLINE | ID: mdl-22708734

ABSTRACT

BACKGROUND: While it is now more than a decade since the first description of the gene mutation underlying the tumour predisposition syndrome multiple endocrine neoplasia type 1 (MEN1), the mechanism by which its protein product menin acts to prevent development of tumours is still poorly understood. METHODS: We undertook a genetic experiment to assess whether menin synergises with p53. Mice carrying various combinations of Men1 and Trp53 mutations were generated then survival and pathology assessed. RESULTS: While homozygous loss of Trp53 in mice resulted in early onset, aggressive tumours and profoundly reduced lifespan, heterozygous loss of either Trp53 or Men1 caused later onset disease, with a spectrum of tumours characteristic of each tumour suppressor gene. Loss of one copy of Men1 in animals also lacking both alleles of Trp53 did not exacerbate phenotype, based on survival, animal weight or sites of pathology, compared to Trp53 deletion alone. Dual heterozygous deletion of Men1 and Trp53 resulted in a small reduction in lifespan compared to the individual mutations, without new tumour sites. In the adrenal, we observed development of cortical tumours in dual heterozygous animals, as we have previously seen in Men1+/- animals, and there was loss of heterozygosity at the Men1 allele in these tumours. Median number of pathology observations per animal was increased in dual heterozygous animals compared with heterozygous loss of Trp53 alone. CONCLUSIONS: Simultaneous heterozygous deletion of Men1 in animals with either heterozygous or homozygous deletion of Trp53 did not result in formation of tumours at any new sites, implying additive rather than synergistic effects of these pathways. Mice that were Men1+/- in addition to Trp53+/- had tumours in endocrine as well as other sites, implying that increase in total tumour burden, at sites typically associated with either Men1 or Trp53 loss, contributed to the slight decrease in survival in Men1+/-: Trp53+/- animals in comparison with their littermates.


Subject(s)
Cell Transformation, Neoplastic/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Tumor Suppressor Protein p53/genetics , Adrenal Glands/metabolism , Adrenal Glands/pathology , Animals , Body Weight , Cell Transformation, Neoplastic/metabolism , Genotype , Mice , Mice, Knockout , Multiple Endocrine Neoplasia Type 1/metabolism , Mutation , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Pancreas/metabolism , Pancreas/pathology , Tumor Suppressor Protein p53/metabolism
10.
Hum Gene Ther ; 22(8): 1011-20, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21595499

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs; Batten disease) are inherited neurodegenerative lysosomal storage diseases with common clinical features of blindness and seizures culminating in premature death. Gene-therapy strategies for these diseases depend on whether the missing activity is a secreted lysosomal protein taken up by neighboring cells, or an intramembrane protein that requires careful targeting. Therapies are best developed in animal models with large complex human-like brains. Lentiviral-mediated gene delivery to neural cell cultures from normal sheep and sheep affected with an NCL resulted in green fluorescent protein (GFP) expression in neurons and neuroblasts, more efficiently than in astrocytes. Similar transgene expression was obtained from two constitutive promoters, the viral MND promoter and the human EF1α promoter. In vivo studies showed stable and persistent GFP expression throughout the cell bodies, axons, and dendrites from intracortical injections and indicated ependymal and subependymal transduction. The sheep showed no ill effects from the injections. These data support continuing gene-therapy trials in the sheep models of Batten disease.


Subject(s)
Brain/metabolism , Lentivirus/genetics , Neuronal Ceroid-Lipofuscinoses/therapy , Animals , Gene Transfer Techniques , Genetic Therapy , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Neuronal Ceroid-Lipofuscinoses/genetics , Neurons/metabolism , Sheep , Transgenes
11.
Neurobiol Dis ; 41(3): 614-23, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21111820

ABSTRACT

The neuronal ceroid lipofuscinoses (NCLs, Batten disease) are characterized by progressive neurodegeneration resulting in widespread brain atrophy. Each form is assumed to be the consequence of some universal intracellular event; however, time course studies on the cerebral cortex of a sheep model of the CLN6 form revealed distinct regional neurodegeneration preceded by regional glial activation, spreading from quite localized foci. Previous neurological investigations have concentrated on obviously affected cortical functions. This study investigated the impact of ovine CLN6 NCL on a subcortical structure and function, the discrete gonadotrophin-releasing hormone (GnRH) secreting neurons of the hypothalamus, and the effect of changes in the neuroendocrine system on reproductive efficiency and embryonic development. The number of immunopositive GnRH neurons in the hypothalamus and median eminence of affected sheep was reduced by 80%, but the rest of the hypothalamus showed no changes or atrophy. This specific loss of neuron type was not accompanied by either microglial or astrocyte activation, which was absent from the hypothalamus and was not associated with cell-type-specific storage body accumulation. Ovarian responsiveness to follicle stimulating hormone, ovulation rates, sperm production, fertilization rates, embryonic development, and reproductive efficiency were sub-par but reproduction was still functional. This remains when the sheep are profoundly blind. We conclude that physiological functionality and connectivity, not genotype, determine neuron fate in CLN6 NCL.


Subject(s)
Gonadotropin-Releasing Hormone/deficiency , Hypothalamus/metabolism , Membrane Proteins/genetics , Neuroglia/metabolism , Neuronal Ceroid-Lipofuscinoses/metabolism , Reproduction/physiology , Animals , Female , Gonadotropin-Releasing Hormone/genetics , Hypothalamus/pathology , Male , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neuroglia/pathology , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Neurons/metabolism , Neurons/pathology , Sheep
12.
Nat Struct Mol Biol ; 17(8): 948-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20657585

ABSTRACT

One X chromosome, selected at random, is silenced in each female mammalian cell. Xist encodes a noncoding RNA that influences the probability that the cis-linked X chromosome will be silenced. We found that the A-repeat, a highly conserved element within Xist, is required for the accumulation of spliced Xist RNA. In addition, the A-repeat is necessary for X-inactivation to occur randomly. In combination, our data suggest that normal Xist RNA processing is important in the regulation of random X-inactivation. We propose that modulation of Xist RNA processing may be part of the stochastic process that determines which X chromosome will be inactivated.


Subject(s)
Nuclear Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Untranslated/genetics , RNA-Binding Proteins/metabolism , Repetitive Sequences, Nucleic Acid/genetics , X Chromosome Inactivation/genetics , Alleles , Animals , Base Sequence , Chromosomes, Mammalian/metabolism , Female , HeLa Cells , Histones/metabolism , Humans , Male , Mice , Models, Biological , Molecular Sequence Data , Nucleic Acid Conformation , Protein Binding , Protein Processing, Post-Translational , RNA, Long Noncoding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Untranslated/chemistry , Sequence Deletion/genetics , Serine-Arginine Splicing Factors
13.
Pancreas ; 39(8): 1140-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20531244

ABSTRACT

OBJECTIVES: To identify gene expression alterations associated with insulinoma formation and progression in 2 mouse models of multiple endocrine neoplasia type 1. METHODS: Mice were killed at 12 or 16 months, and pancreatic islets were isolated by enzymatic and physical disruption. Islets were separated by size representing control, normal, hyperplastic, and adenomous islets. RNA was isolated from these islets and profiled on Sentrix Mouse-6 Expression version 1 BeadChips. Array data were analyzed in GeneSpring. RESULTS: One hundred and one genes that were significantly (P ≤ 0.05) altered in hyperplastic islets and insulinomas compared with normal islets were identified. Of these, 64 gene elements showed reduced messenger RNA levels and 37 gene elements had increased gene expression compared with control islets. Altered expression of 3 genes, namely, Gata6, Tspan8, and s100a8, was confirmed by quantitative reverse transcription-polymerase chain reaction, and aberrant levels of Tspan8 and Lmo2 protein measured by Western blot correlated with the changes in messenger RNA levels. CONCLUSIONS: These results suggest that alterations in gene expression of Gata6, Tspan8, S100a8, and Lmo2 may act via novel pathways that play functionally important roles in Men1-associated tumor progression.


Subject(s)
Gene Expression Profiling , Insulinoma/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Pancreatic Neoplasms/genetics , Adaptor Proteins, Signal Transducing , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Blotting, Western , Calgranulin A/genetics , Calgranulin A/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disease Progression , Female , GATA6 Transcription Factor/genetics , GATA6 Transcription Factor/metabolism , Humans , Insulinoma/etiology , Insulinoma/metabolism , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , LIM Domain Proteins , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Metalloproteins/genetics , Metalloproteins/metabolism , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Multiple Endocrine Neoplasia Type 1/complications , Multiple Endocrine Neoplasia Type 1/metabolism , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms/etiology , Pancreatic Neoplasms/metabolism , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tetraspanins
14.
Pigment Cell Melanoma Res ; 23(4): 564-70, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20518858

ABSTRACT

There has been uncertainty regarding the precise role that the pocket protein Rb1 plays in murine melanocyte homeostasis. It has been reported that the TAT-Cre mediated loss of exon 19 from a floxed Rb1 allele causes melanocyte apoptosis in vivo and in vitro. This is at variance with other findings showing, either directly or indirectly, that Rb1 loss in melanocytes has no noticeable effect in vivo, but in vitro leads to a semi-transformed phenotype. In this study, we show that Rb1-null melanocytes lacking exon 19 do not undergo apoptosis and survive both in vitro and in vivo, irrespective of the developmental stage at which Cre-mediated ablation of the exon occurs. Further, Rb1 loss has no serious long-term ramifications on melanocyte homeostasis in vivo, with Rb1-null melanocytes being detected in the skin after numerous hair cycles, inferring that the melanocyte stem cell population carrying the Cre-mediated deletion is maintained. Consequently, whilst Rb1 loss in the melanocyte is able to alter cellular behaviour in vitro, it appears inconsequential with respect to melanocyte homeostasis in the mouse skin.


Subject(s)
Hair/metabolism , Homeostasis , Melanocytes/metabolism , Retinoblastoma Protein/deficiency , Skin/metabolism , Animals , Hair/pathology , Mice , Mice, Knockout , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Skin/pathology
15.
Vet Immunol Immunopathol ; 136(3-4): 324-9, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20409592

ABSTRACT

The aim of this study is to compare the immune responses of sheep stimulated by the intramuscular injection of a liposome formulated-DNA plasmid encoding the Toxoplasma gondii MAG1 antigen only or co-expressed with ovine IL-6. Forty-five, 2-year-old sheep were divided into four groups. Group 1 received an empty pVAXIg plasmid, group 2 no treatment, group 3 liposome formulated plasmid pVAXIgMAG1 and group 4, pVAXIgMAG1 plus pVAXovIL-6 plasmids. All the animals were inoculated at weeks 0 and 4. The injection of sheep with a plasmid encoding for MAG1 only or a MAG1 plasmid co-expressed with a plasmid encoding for ovine IL-6 produced humoral immune responses. The plasmids containing MAG1 elevated significantly serum IgG1 and IgG2 levels 2 weeks and onwards after the first injection of the plasmids. Co-expression of IL-6 with MAG1 had no effect on IG1 or IG2 levels illustrating that IL-6 in the formulation used had no modulating effect on any measured immune response.


Subject(s)
Antigens, Protozoan/immunology , Immunization/veterinary , Interleukin-6/immunology , Sheep Diseases/parasitology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Antibodies, Protozoan/blood , Antigens, Protozoan/genetics , Female , Immunization/methods , Injections, Intramuscular/veterinary , Interferon-gamma/blood , Interleukin-6/genetics , Plasmids/pharmacology , Protozoan Vaccines/immunology , Protozoan Vaccines/standards , Sheep , Sheep Diseases/immunology , Sheep Diseases/prevention & control , Toxoplasma/genetics , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/prevention & control , Vaccines, DNA/immunology , Vaccines, DNA/standards
16.
Neoplasia ; 12(3): 235-43, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20234817

ABSTRACT

Using a Cre/loxP system, we have determined the phenotypic consequences attributable to in vivo deletion of both Rb1 and Trp53 in the mouse adrenal medulla. The coablation of these two tumor suppressor genes during embryogenesis did not disrupt adrenal gland development but resulted in the neoplastic transformation of the neural crest-derived adrenal medulla, yielding pheochromocytomas (PCCs) that developed with complete penetrance and were inevitably bilateral. Despite their typically benign status, these PCCs had profound ramifications on mouse vitality, with effected mice having a median survival of only 121 days. Evaluation of these PCCs by both immunohistochemistry and electron microscopy revealed that most Rb1(-/-):Trp53(-/-) chromaffin cells possessed atypical chromagenic vesicles that did not seem capable of appropriately storing synthesized catecholamines. The structural remodeling of the heart in mice harboring Rb1(-/-):Trp53(-/-) PCCs suggests that the mortality of these mice may be attributable to the inappropriate release of catecholamines from the mutated adrenal chromaffin cells. On the basis of the collective data from Rb1 and Trp53 knockout mouse models, it seems that the conversion of Rb1 loss-driven adrenal medulla hyperplasia to PCC can be greatly enhanced by the compound loss of Trp53, whereas the loss of Trp53 alone is generally ineffectual on adrenal chromaffin cell homeostasis. Consequently, the Trp53 tumor suppressor gene is an efficient genetic modifier of Rb1 loss in the development of PCC, and their compound loss in the adrenal medulla has a profound impact on both cellular homeostasis and animal vitality.


Subject(s)
Adrenal Gland Neoplasms/pathology , Genes, p53/physiology , Pheochromocytoma/pathology , Retinoblastoma Protein/physiology , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/metabolism , Animals , Catecholamines/metabolism , Female , Immunoenzyme Techniques , Integrases/metabolism , Male , Mice , Mice, Inbred C57BL , Pheochromocytoma/genetics , Pheochromocytoma/metabolism , Transgenes/physiology
17.
Neurochem Int ; 55(8): 783-8, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19664668

ABSTRACT

Anomalies in neuropeptides and neuroactive amino acids have been postulated to play a role in neurodegeneration in a variety of diseases including the inherited neuronal ceroid lipofuscinoses (NCLs, Batten disease). These are often indicated by concentration changes in cerebrospinal fluid (CSF). Here we compare CSF neuropeptide concentrations in patients with the classical juvenile CLN3 form of NCL and the classical late infantile CLN2 form with neuropeptide and neuroactive amino acid concentrations in CSF from sheep with the late infantile variant CLN6 form. A marked disease related increase in CSF concentrations of neuron specific enolase and tau protein was noted in the juvenile CLN3 patients but this was not observed in an advanced CLN2 patient nor CLN6 affected sheep. No changes were noted in S-100b, GFAP or MBP in patients or of S-100b, GFAP or IGF-1 in affected sheep. There were no disease related changes in CSF concentrations of the neuroactive amino acids, aspartate, glutamate, serine, glutamine, glycine, taurine and GABA in these sheep. The changes observed in the CLN3 patients may be progressive markers of neurodegeneration, or of underlying metabolic changes perhaps associated with CLN3 specific changes in neuroactive amino acids, as have been postulated. The lack of changes in the CLN2 and CLN6 subjects indicate that these changes are not shared by the CLN2 or CLN6 forms and changes in CSF concentrations of these compounds are unreliable as biomarkers of neurodegeneration in the NCLs in general.


Subject(s)
Amino Acids/cerebrospinal fluid , Neuronal Ceroid-Lipofuscinoses/cerebrospinal fluid , Neuropeptides/cerebrospinal fluid , Neurotransmitter Agents/cerebrospinal fluid , Age Factors , Amino Acids/analysis , Animals , Biomarkers/analysis , Biomarkers/cerebrospinal fluid , Child , Child, Preschool , Female , Glial Fibrillary Acidic Protein/analysis , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Humans , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/cerebrospinal fluid , Male , Myelin Basic Protein/analysis , Myelin Basic Protein/cerebrospinal fluid , Nerve Growth Factors/analysis , Nerve Growth Factors/cerebrospinal fluid , Neuronal Ceroid-Lipofuscinoses/physiopathology , Neuropeptides/analysis , Neurotransmitter Agents/analysis , Phosphopyruvate Hydratase/analysis , Phosphopyruvate Hydratase/cerebrospinal fluid , Predictive Value of Tests , S100 Calcium Binding Protein beta Subunit , S100 Proteins/analysis , S100 Proteins/cerebrospinal fluid , Sheep , Tripeptidyl-Peptidase 1 , tau Proteins/analysis , tau Proteins/cerebrospinal fluid
18.
Biol Reprod ; 81(5): 921-32, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19571264

ABSTRACT

An estimated 25%-40% of infertile men have idiopathic infertility associated with deficient sperm numbers and quality. Here, we identify the membrane-anchored serine protease PRSS21, also known as testisin, to be a novel proteolytic factor that directs epididymal sperm cell maturation and sperm-fertilizing ability. PRSS21-deficient spermatozoa show decreased motility, angulated and curled tails, fragile necks, and dramatically increased susceptibility to decapitation. These defects reflect aberrant maturation during passage through the epididymis, because histological and electron microscopic structural analyses showed an increased tendency for curled and detached tails as spermatozoa transit from the corpus to the cauda epididymis. Cauda epididymal spermatozoa deficient in PRSS21 fail to mount a swelling response when exposed to hypotonic conditions, suggesting an impaired ability to respond to osmotic challenges facing maturing spermatozoa in the female reproductive tract. These data suggest that aberrant regulation of PRSS21 may underlie certain secondary male infertility syndromes, such as "easily decapitated" spermatozoa in humans.


Subject(s)
Fertilization/physiology , Membrane Proteins/metabolism , Serine Endopeptidases/metabolism , Sperm Maturation/physiology , Sperm Motility/physiology , Spermatozoa/cytology , Animals , Blotting, Western , Cell Count , Cell Shape , Cell Survival , Copulation/physiology , Female , Fertilization in Vitro , GPI-Linked Proteins , Humans , Immunohistochemistry , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Phosphorylation , Serine Endopeptidases/genetics , Spermatozoa/metabolism , Staining and Labeling
20.
Int J Cancer ; 124(5): 1122-32, 2009 Mar 01.
Article in English | MEDLINE | ID: mdl-19058182

ABSTRACT

Heterozygous disruption of the Men1 gene predisposes mice to the development of multiple endocrine tumors, accurately mimicking the human MEN1 cancer predisposition syndrome. Additionally, Men1(+/-) mice frequently develop sex cord adenomas. The mechanism underlying the susceptibility of these mice to sex cord tumor development has not been fully determined, but data suggest it may involve transcriptional regulation of key growth promoting/repressing genes. To identify potential menin-regulated genes that may be important for tumor suppression in sex cord cells, we compared the global gene expression profiles of testis and ovary adenomas with other endocrine tumors of the pancreas and pituitary from Men1 heterozygous mice and with control tissues. Gonadal tumors clustered separately from pancreas and pituitary tumors with only a few genes (e.g., Cdkn2c) commonly dysregulated in all tumor types. Testis and ovary tumors displayed a higher level of transcriptional similarity to each other than they did to their respective control tissues. Among genes that had decreased expression in tumors was significant over-representation of genes associated with the TGF-beta, hedgehog and Wnt signaling, indicating that loss of menin function affects these pathways at the level of transcription. Aberrant protein expression in Leydig and granulosa cells of 2 transcriptionally dysregulated gene products, Gata6 and Csf1r were confirmed by immunohistochemistry. We propose that sex cord tumor susceptibility in Men1(+/-) mice involves deregulated cell proliferation due to dysregulation of multiple cell growth regulating genes including: reduced Cdkn2c transcription, loss of TGF-beta pathway tumor suppressor function (e.g., Gata6) and transcriptional activation of Csf1r.


Subject(s)
GATA6 Transcription Factor/analysis , Gene Expression Profiling , Proto-Oncogene Proteins/physiology , Receptor, Macrophage Colony-Stimulating Factor/analysis , Sex Cord-Gonadal Stromal Tumors/etiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Animals , Cluster Analysis , Immunohistochemistry , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...