Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
J Mol Biol ; 432(14): 3989-4009, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32304700

ABSTRACT

The impenetrability of the blood-brain barrier (BBB) to most conventional drugs impedes the treatment of central nervous system (CNS) disorders. Interventions for diseases like brain cancer, neurodegeneration, or age-associated inflammatory processes require varied approaches to CNS drug delivery. Cystine-dense peptides (CDPs) have drawn recent interest as drugs or drug-delivery vehicles. Found throughout the phylogenetic tree, often in drug-like roles, their size, stability, and protein interaction capabilities make CDPs an attractive mid-size biologic scaffold to complement conventional antibody-based drugs. Here, we describe the identification, maturation, characterization, and utilization of a CDP that binds to the transferrin receptor (TfR), a native receptor and BBB transporter for the iron chaperone transferrin. We developed variants with varying binding affinities (KD as low as 216 pM), co-crystallized it with the receptor, and confirmed murine cross-reactivity. It accumulates in the mouse CNS at ~25% of blood levels (CNS blood content is only ~1%-6%) and delivers neurotensin, an otherwise non-BBB-penetrant neuropeptide, at levels capable of modulating CREB signaling in the mouse brain. Our work highlights the utility of CDPs as a diverse, easy-to-screen scaffold family worthy of inclusion in modern drug discovery strategies, demonstrated by the discovery of a candidate CNS drug delivery vehicle ready for further optimization and preclinical development.


Subject(s)
Blood-Brain Barrier/drug effects , Central Nervous System Diseases/drug therapy , Drug Delivery Systems , Peptides/pharmacology , Animals , Antigens, CD/chemistry , Antigens, CD/drug effects , Antigens, CD/genetics , Antigens, CD/pharmacology , Central Nervous System/drug effects , Cystine/chemistry , Cystine/genetics , Humans , Inflammation/drug therapy , Inflammation/pathology , Mice , Neuropeptides/chemistry , Neuropeptides/pharmacology , Neurotensin/chemistry , Neurotensin/pharmacology , Peptides/chemistry , Protein Binding/drug effects , Receptors, Transferrin/chemistry , Receptors, Transferrin/drug effects , Receptors, Transferrin/genetics
2.
Sci Transl Med ; 12(533)2020 03 04.
Article in English | MEDLINE | ID: mdl-32132215

ABSTRACT

On-target, off-tissue toxicity limits the systemic use of drugs that would otherwise reduce symptoms or reverse the damage of arthritic diseases, leaving millions of patients in pain and with limited physical mobility. We identified cystine-dense peptides (CDPs) that rapidly accumulate in cartilage of the knees, ankles, hips, shoulders, and intervertebral discs after systemic administration. These CDPs could be used to concentrate arthritis drugs in joints. A cartilage-accumulating peptide, CDP-11R, reached peak concentration in cartilage within 30 min after administration and remained detectable for more than 4 days. Structural analysis of the peptides by crystallography revealed that the distribution of positive charge may be a distinguishing feature of joint-accumulating CDPs. In addition, quantitative whole-body autoradiography showed that the disulfide-bonded tertiary structure is critical for cartilage accumulation and retention. CDP-11R distributed to joints while carrying a fluorophore imaging agent or one of two different steroid payloads, dexamethasone (dex) and triamcinolone acetonide (TAA). Of the two payloads, the dex conjugate did not advance because the free drug released into circulation was sufficient to cause on-target toxicity. In contrast, the CDP-11R-TAA conjugate alleviated joint inflammation in the rat collagen-induced model of rheumatoid arthritis while avoiding toxicities that occurred with nontargeted steroid treatment at the same molar dose. This conjugate shows promise for clinical development and establishes proof of concept for multijoint targeting of disease-modifying therapeutic payloads.


Subject(s)
Arthritis, Experimental , Adrenal Cortex Hormones , Animals , Arthritis, Experimental/drug therapy , Cartilage , Humans , Peptides , Rats , Steroids
3.
Nat Med ; 24(11): 1752-1761, 2018 11.
Article in English | MEDLINE | ID: mdl-30349086

ABSTRACT

Brain tumors are the leading cause of cancer-related death in children. Genomic studies have provided insights into molecular subgroups and oncogenic drivers of pediatric brain tumors that may lead to novel therapeutic strategies. To evaluate new treatments, better preclinical models adequately reflecting the biological heterogeneity are needed. Through the Children's Oncology Group ACNS02B3 study, we have generated and comprehensively characterized 30 patient-derived orthotopic xenograft models and seven cell lines representing 14 molecular subgroups of pediatric brain tumors. Patient-derived orthotopic xenograft models were found to be representative of the human tumors they were derived from in terms of histology, immunohistochemistry, gene expression, DNA methylation, copy number, and mutational profiles. In vivo drug sensitivity of targeted therapeutics was associated with distinct molecular tumor subgroups and specific genetic alterations. These models and their molecular characterization provide an unprecedented resource for the cancer community to study key oncogenic drivers and to evaluate novel treatment strategies.


Subject(s)
Biological Specimen Banks , Brain Neoplasms/pathology , Immunohistochemistry , Xenograft Model Antitumor Assays/methods , Animals , Cell Line, Tumor , Child , Child, Preschool , DNA Methylation/genetics , Female , Genomics , Humans , Male , Mice , Mutation , Pediatrics
4.
Sci Transl Med ; 7(284): 284ra58, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25904742

ABSTRACT

A fundamental problem in cancer drug development is that antitumor efficacy in preclinical cancer models does not translate faithfully to patient outcomes. Much of early cancer drug discovery is performed under in vitro conditions in cell-based models that poorly represent actual malignancies. To address this inconsistency, we have developed a technology platform called CIVO, which enables simultaneous assessment of up to eight drugs or drug combinations within a single solid tumor in vivo. The platform is currently designed for use in animal models of cancer and patients with superficial tumors but can be modified for investigation of deeper-seated malignancies. In xenograft lymphoma models, CIVO microinjection of well-characterized anticancer agents (vincristine, doxorubicin, mafosfamide, and prednisolone) induced spatially defined cellular changes around sites of drug exposure, specific to the known mechanisms of action of each drug. The observed localized responses predicted responses to systemically delivered drugs in animals. In pair-matched lymphoma models, CIVO correctly demonstrated tumor resistance to doxorubicin and vincristine and an unexpected enhanced sensitivity to mafosfamide in multidrug-resistant lymphomas compared with chemotherapy-naïve lymphomas. A CIVO-enabled in vivo screen of 97 approved oncology agents revealed a novel mTOR (mammalian target of rapamycin) pathway inhibitor that exhibits significantly increased tumor-killing activity in the drug-resistant setting compared with chemotherapy-naïve tumors. Finally, feasibility studies to assess the use of CIVO in human and canine patients demonstrated that microinjection of drugs is toxicity-sparing while inducing robust, easily tracked, drug-specific responses in autochthonous tumors, setting the stage for further application of this technology in clinical trials.


Subject(s)
Antineoplastic Agents/chemistry , Drug Screening Assays, Antitumor/methods , Lymphoma/drug therapy , Neoplasms/drug therapy , Animals , Biomarkers , Cell Line, Tumor , Cyclophosphamide/analogs & derivatives , Cyclophosphamide/chemistry , Dogs , Doxorubicin/chemistry , Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Humans , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Prednisolone/chemistry , TOR Serine-Threonine Kinases/metabolism , Vincristine/chemistry
5.
Neuroreport ; 20(12): 1098-103, 2009 Aug 05.
Article in English | MEDLINE | ID: mdl-19590393

ABSTRACT

Neuroinflammation is a prominent feature of many neurodegenerative diseases, however, little is known about neuroinflammation in Huntington's disease. We used quantitative real time-PCR to compare the expression level of neuroinflammation-associated mediators in the striatum, cortex, and cerebellum from post-mortem Huntington's disease patient samples with controls. We found increased expression of several key inflammatory mediators, including CCL2 and IL-10, specifically in the striatum of Huntington's disease patients, the main area affected by this pathology. Remarkably, we also found upregulation of IL-6, IL-8, and MMP9, in the cortex and notably the cerebellum, a brain area commonly thought to be spared by Huntington's disease. Our data suggest that neuroinflammation is a prominent feature associated with Huntington's disease and may constitute a novel target for therapeutic intervention.


Subject(s)
Cerebellum/immunology , Cerebral Cortex/immunology , Corpus Striatum/immunology , Huntington Disease/immunology , Adult , Aged , Cerebellum/metabolism , Cerebral Cortex/metabolism , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Corpus Striatum/metabolism , Female , Gene Expression , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Middle Aged , RNA, Messenger/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
6.
J Neurosci ; 27(43): 11758-68, 2007 Oct 24.
Article in English | MEDLINE | ID: mdl-17959817

ABSTRACT

Many pathways have been proposed as contributing to Huntington's disease (HD) pathogenesis, but generally the in vivo effects of their perturbation have not been compared with reference data from human patients. Here we examine how accurately mechanistically motivated and genetic HD models recapitulate the striatal gene expression phenotype of human HD. The representative genetic model was the R6/2 transgenic mouse, which expresses a fragment of the huntingtin protein containing a long CAG repeat. Pathogenic mechanisms examined include mitochondrial dysfunction; profiled in 3-nitropropionic acid-treated rats, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated mice, and PGC-1alpha knock-out mice; and depletion of brain-derived neurotrophic factor (BDNF) using heterozygous and forebrain-specific BDNF-knock-out mice (BDNF(HET), Emx-BDNF(KO)). Based on striatal gene expression, we find the BDNF models, both heterozygous and homozygous knock-outs, to be more like human HD than the other HD models. This implicates reduced trophic support as a major pathway contributing to striatal degeneration in HD. Because the majority of striatal BDNF is synthesized by cortical neurons, the data also imply that cortical dysfunction contributes to HD's hallmark effects on the basal ganglia. Finally, the results suggest that striatal lesions caused by mitochondrial toxins may arise via pathways different from those that drive neurodegeneration in HD. Based on these findings, we present a testable model of HD pathogenesis that, unlike most models, begins to account for regional specificity in human HD and the absence of such specificity in genetic mouse models of HD.


Subject(s)
Brain-Derived Neurotrophic Factor/deficiency , Corpus Striatum/metabolism , Disease Models, Animal , Gene Expression Profiling/methods , Huntington Disease/metabolism , Nerve Degeneration/metabolism , Animals , Brain-Derived Neurotrophic Factor/genetics , Corpus Striatum/pathology , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Knockout , Mice, Transgenic , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Rats , Rats, Inbred Lew
7.
Hum Mol Genet ; 16(15): 1845-61, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17519223

ABSTRACT

To test the hypotheses that mutant huntingtin protein length and wild-type huntingtin dosage have important effects on disease-related transcriptional dysfunction, we compared the changes in mRNA in seven genetic mouse models of Huntington's disease (HD) and postmortem human HD caudate. Transgenic models expressing short N-terminal fragments of mutant huntingtin (R6/1 and R6/2 mice) exhibited the most rapid effects on gene expression, consistent with previous studies. Although changes in the brains of knock-in and full-length transgenic models of HD took longer to appear, 15- and 22-month CHL2(Q150/Q150), 18-month Hdh(Q92/Q92) and 2-year-old YAC128 animals also exhibited significant HD-like mRNA signatures. Whereas it was expected that the expression of full-length huntingtin transprotein might result in unique gene expression changes compared with those caused by the expression of an N-terminal huntingtin fragment, no discernable differences between full-length and fragment models were detected. In addition, very high correlations between the signatures of mice expressing normal levels of wild-type huntingtin and mice in which the wild-type protein is absent suggest a limited effect of the wild-type protein to change basal gene expression or to influence the qualitative disease-related effect of mutant huntingtin. The combined analysis of mouse and human HD transcriptomes provides important temporal and mechanistic insights into the process by which mutant huntingtin kills striatal neurons. In addition, the discovery that several available lines of HD mice faithfully recapitulate the gene expression signature of the human disorder provides a novel aspect of validation with respect to their use in preclinical therapeutic trials.


Subject(s)
Corpus Striatum/metabolism , Gene Expression , Huntington Disease/genetics , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Gene Dosage , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Phenotype , RNA, Messenger/metabolism
8.
PLoS Genet ; 3(4): e59, 2007 Apr 20.
Article in English | MEDLINE | ID: mdl-17447843

ABSTRACT

Many neurodegenerative diseases have a hallmark regional and cellular pathology. Gene expression analysis of healthy tissues may provide clues to the differences that distinguish resistant and sensitive tissues and cell types. Comparative analysis of gene expression in healthy mouse and human brain provides a framework to explore the ability of mice to model diseases of the human brain. It may also aid in understanding brain evolution and the basis for higher order cognitive abilities. Here we compare gene expression profiles of human motor cortex, caudate nucleus, and cerebellum to one another and identify genes that are more highly expressed in one region relative to another. We separately perform identical analysis on corresponding brain regions from mice. Within each species, we find that the different brain regions have distinctly different expression profiles. Contrasting between the two species shows that regionally enriched genes in one species are generally regionally enriched genes in the other species. Thus, even when considering thousands of genes, the expression ratios in two regions from one species are significantly correlated with expression ratios in the other species. Finally, genes whose expression is higher in one area of the brain relative to the other areas, in other words genes with patterned expression, tend to have greater conservation of nucleotide sequence than more widely expressed genes. Together these observations suggest that region-specific genes have been conserved in the mammalian brain at both the sequence and gene expression levels. Given the general similarity between patterns of gene expression in healthy human and mouse brains, we believe it is reasonable to expect a high degree of concordance between microarray phenotypes of human neurodegenerative diseases and their mouse models. Finally, these data on very divergent species provide context for studies in more closely related species that address questions such as the origins of cognitive differences.


Subject(s)
Brain/metabolism , Evolution, Molecular , Gene Expression , Adult , Aged , Animals , Conserved Sequence , Female , Gene Expression Profiling , Genetic Variation , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Organ Specificity , Tissue Distribution
9.
Cell Metab ; 4(5): 349-62, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17055784

ABSTRACT

Huntington's disease (HD) is a fatal, dominantly inherited disorder caused by polyglutamine repeat expansion in the huntingtin (htt) gene. Here, we observe that HD mice develop hypothermia associated with impaired activation of brown adipose tissue (BAT). Although sympathetic stimulation of PPARgamma coactivator 1alpha (PGC-1alpha) was intact in BAT of HD mice, uncoupling protein 1 (UCP-1) induction was blunted. In cultured cells, expression of mutant htt suppressed UCP-1 promoter activity; this was reversed by PGC-1alpha expression. HD mice showed reduced food intake and increased energy expenditure, with dysfunctional BAT mitochondria. PGC-1alpha is a known regulator of mitochondrial function; here, we document reduced expression of PGC-1alpha target genes in HD patient and mouse striatum. Mitochondria of HD mouse brain show reduced oxygen consumption rates. Finally, HD striatal neurons expressing exogenous PGC-1alpha were resistant to 3-nitropropionic acid treatment. Altered PGC-1alpha function may thus link transcription dysregulation and mitochondrial dysfunction in HD.


Subject(s)
Adipose Tissue, Brown/physiopathology , Body Temperature Regulation/genetics , Heat-Shock Proteins/metabolism , Huntington Disease/etiology , Transcription Factors/metabolism , Animals , Body Temperature/genetics , Cells, Cultured , Disease Models, Animal , Heat-Shock Proteins/genetics , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Mice, Transgenic , Mitochondria/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Signal Transduction/genetics , Transcription Factors/genetics , Transcription, Genetic
10.
BMC Bioinformatics ; 7: 211, 2006 Apr 19.
Article in English | MEDLINE | ID: mdl-16623940

ABSTRACT

BACKGROUND: Gene expression microarray experiments are expensive to conduct and guidelines for acceptable quality control at intermediate steps before and after the samples are hybridised to chips are vague. We conducted an experiment hybridising RNA from human brain to 117 U133A Affymetrix GeneChips and used these data to explore the relationship between 4 pre-chip variables and 22 post-chip outcomes and quality control measures. RESULTS: We found that the pre-chip variables were significantly correlated with each other but that this correlation was strongest between measures of RNA quality and cRNA yield. Post-mortem interval was negatively correlated with these variables. Four principal components, reflecting array outliers, array adjustment, hybridisation noise and RNA integrity, explain about 75% of the total post-chip measure variability. Two significant canonical correlations existed between the pre-chip and post-chip variables, derived from MAS 5.0, dChip and the Bioconductor packages affy and affyPLM. The strongest (CANCOR 0.838, p < 0.0001) correlated RNA integrity and yield with post chip quality control (QC) measures indexing 3'/5' RNA ratios, bias or scaling of the chip and scaling of the variability of the signal across the chip. Post-mortem interval was relatively unimportant. We also found that the RNA integrity number (RIN) could be moderately well predicted by post-chip measures B_ACTIN35, GAPDH35 and SF. CONCLUSION: We have found that the post-chip variables having the strongest association with quantities measurable before hybridisation are those reflecting RNA integrity. Other aspects of quality, such as noise measures (reflecting the execution of the assay) or measures reflecting data quality (outlier status and array adjustment variables) are not well predicted by the variables we were able to determine ahead of time. There could be other variables measurable pre-hybridisation which may be better associated with expression data quality measures. Uncovering such connections could create savings on costly microarray experiments by eliminating poor samples before hybridisation.


Subject(s)
Artifacts , Gene Expression Profiling/instrumentation , Models, Genetic , Oligonucleotide Array Sequence Analysis/instrumentation , Quality Assurance, Health Care/methods , Computer Simulation , Data Interpretation, Statistical , Equipment Design , Equipment Failure Analysis , Models, Statistical , Quality Control , Reproducibility of Results , Sensitivity and Specificity
11.
Hum Mol Genet ; 15(6): 965-77, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16467349

ABSTRACT

Huntington's disease (HD) pathology is well understood at a histological level but a comprehensive molecular analysis of the effect of the disease in the human brain has not previously been available. To elucidate the molecular phenotype of HD on a genome-wide scale, we compared mRNA profiles from 44 human HD brains with those from 36 unaffected controls using microarray analysis. Four brain regions were analyzed: caudate nucleus, cerebellum, prefrontal association cortex [Brodmann's area 9 (BA9)] and motor cortex [Brodmann's area 4 (BA4)]. The greatest number and magnitude of differentially expressed mRNAs were detected in the caudate nucleus, followed by motor cortex, then cerebellum. Thus, the molecular phenotype of HD generally parallels established neuropathology. Surprisingly, no mRNA changes were detected in prefrontal association cortex, thereby revealing subtleties of pathology not previously disclosed by histological methods. To establish that the observed changes were not simply the result of cell loss, we examined mRNA levels in laser-capture microdissected neurons from Grade 1 HD caudate compared to control. These analyses confirmed changes in expression seen in tissue homogenates; we thus conclude that mRNA changes are not attributable to cell loss alone. These data from bona fide HD brains comprise an important reference for hypotheses related to HD and other neurodegenerative diseases.


Subject(s)
Brain/metabolism , Gene Expression Profiling , Huntington Disease/genetics , Huntington Disease/metabolism , Adult , Aged , Axons/metabolism , Brain/pathology , Cell Death/genetics , Female , Humans , Huntington Disease/pathology , Male , Middle Aged , Oligonucleotide Array Sequence Analysis , RNA, Messenger/biosynthesis , Signal Transduction/genetics
12.
Hum Mol Genet ; 14(20): 3065-78, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16183657

ABSTRACT

In postmortem Huntington's disease brains, mutant htt is present in both nuclear and cytoplasmic compartments. To dissect the impact of nuclear and extranuclear mutant htt on the initiation and progression of disease, we generated a series of transgenic mouse lines in which nuclear localization or nuclear export signal sequences have been placed N-terminal to the htt exon 1 protein carrying 144 glutamines. Our data indicate that the exon 1 mutant protein is present in the nucleus as part of an oligomeric or aggregation complex. Increasing the concentration of the mutant transprotein in the nucleus is sufficient for and dramatically accelerates the onset and progression of behavioral phenotypes. Furthermore, nuclear exon 1 mutant protein is sufficient to induce cytoplasmic neurodegeneration and transcriptional dysregulation. However, our data suggest that cytoplasmic mutant exon 1 htt, if present, contributes to disease progression.


Subject(s)
Cell Nucleus/metabolism , Disease Models, Animal , Huntington Disease/metabolism , Huntington Disease/pathology , Peptides/metabolism , Animals , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Cytoplasm/metabolism , Gene Expression Profiling , Gene Expression Regulation , Humans , Huntingtin Protein , Macromolecular Substances , Mice , Mice, Transgenic , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Localization Signals , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rotarod Performance Test , Transcription, Genetic , Transgenes/genetics
13.
Stat Med ; 24(15): 2281-98, 2005 Aug 15.
Article in English | MEDLINE | ID: mdl-15889452

ABSTRACT

Which significance test is carried out when the number of repeats is small in microarray experiments can dramatically influence the results. When in two sample comparisons both conditions have fewer than, say, five repeats traditional test statistics require extreme results, before a gene is considered statistically significant differentially expressed after a multiple comparisons correction. In the literature many approaches to circumvent this problem have been proposed. Some of these proposals use (empirical) Bayes arguments to moderate the variance estimates for individual genes. Other proposals try to stabilize these variance estimate by combining groups of genes or similar experiments. In this paper we compare several of these approaches, both on data sets where both experimental conditions are the same, and thus few statistically significant differentially expressed genes should be identified, and on experiments where both conditions do differ. This allows us to identify which approaches are most powerful without identifying many false positives. We conclude that after balancing the numbers of false positives and true positives an empirical Bayes approach and an approach which combines experiments perform best. Standard t-tests are inferior and offer almost no power when the sample size is small.


Subject(s)
Bayes Theorem , Data Interpretation, Statistical , Oligonucleotide Array Sequence Analysis/methods , Animals , Computer Simulation , DNA Fingerprinting/methods , False Positive Reactions , Humans , Huntington Disease/genetics , Mice
14.
Hum Mol Genet ; 14(13): 1863-76, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15888475

ABSTRACT

Huntington's disease (HD) is an incurable and fatal neurodegenerative disorder. Improvements in the objective measurement of HD will lead to more efficient clinical trials and earlier therapeutic intervention. We hypothesized that abnormalities seen in the R6/2 mouse, a greatly accelerated HD model, might highlight subtle phenotypes in other mouse models and human HD. In this paper, we identify common gene expression changes in skeletal muscle from R6/2 mice, Hdh(CAG(150)) homozygous knock-in mice and HD patients. This HD-triggered gene expression phenotype is consistent with the beginnings of a transition from fast-twitch to slow-twitch muscle fiber types. Metabolic adaptations similar to those induced by diabetes or fasting are also present but neither metabolic disorder can explain the full phenotype of HD muscle. The HD-induced gene expression changes reflect disease progression. This raises the possibility that muscle gene expression may be used as an objective biomarker to complement clinical HD-rating systems. Furthermore, an understanding of the molecular basis of muscle dysfunction in HD should provide insight into mechanisms involved in neuronal abnormalities and neurodegeneration.


Subject(s)
Gene Expression Regulation , Huntington Disease/metabolism , Muscle, Skeletal/metabolism , Animals , Biomarkers , Disease Models, Animal , Gene Expression Profiling , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Knockout , Muscle Fibers, Fast-Twitch/metabolism , Muscle Fibers, Fast-Twitch/pathology , Muscle Fibers, Slow-Twitch/metabolism , Muscle Fibers, Slow-Twitch/pathology , Muscle, Skeletal/pathology , Oligonucleotide Array Sequence Analysis
15.
J Cell Biochem ; 90(5): 1068-78, 2003 Dec 01.
Article in English | MEDLINE | ID: mdl-14624465

ABSTRACT

Large-scale gene expression measurements with oligonucleotide microarrays have contributed tremendously to biological research. However, to distinguish between relevant expression changes and falsely identified positives, the source and magnitude of errors must be understood. Here, we report a source of biological variability in microarray experiments with stably transfected cell lines. Mouse embryonic fibroblast (MEF/3T3) and rat schwannoma (RT4) cell lines were generated to provide regulatable schwannomin expression. The expression levels of 29 samples from five different mouse embryonic fibroblast clonal cell lines and 18 samples from 3 RT4 cell lines were monitored with oligonucleotide microarrays. Using hierarchical clustering, we determined that the changes in gene expression induced by schwannomin overexpression were subtle when compared with those detected as a consequence of clonal selection during generation of the cell lines. The hierarchical clustering implies that significant alterations of gene expression were introduced during the transfection and selection processes. A total of 28 genes were identified by Kruskal-Wallis rank test that showed significant variation between clonal lines. Most of them were related to cytoskeletal function and signaling pathways. Based on these analyses, we recommend that replications of experiments with several selected cell lines are necessary to assess biological effects of induced gene expression.


Subject(s)
Fibroblasts/metabolism , Gene Expression Profiling , Genetic Heterogeneity , Neurilemmoma/metabolism , Oligonucleotide Array Sequence Analysis/statistics & numerical data , Research Design/trends , Animals , Cell Line , Cluster Analysis , Cytoskeleton/metabolism , Gene Expression Regulation , Mice , Rats , Reproducibility of Results , Signal Transduction , Transfection
16.
Nat Med ; 9(8): 1033-8, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12872164

ABSTRACT

The mechanisms of retinoid activity in tumors remain largely unknown. Here we establish that retinoids cause extensive apoptosis of medulloblastoma cells. In a xenograft model, retinoids largely abrogated tumor growth. Using receptor-specific retinoid agonists, we defined a subset of mRNAs that were induced by all active retinoids in retinoid-sensitive cell lines. We also identified bone morphogenetic protein-2 (BMP-2) as a candidate mediator of retinoid activity. BMP-2 protein induced medulloblastoma cell apoptosis, whereas the BMP-2 antagonist noggin blocked both retinoid and BMP-2-induced apoptosis. BMP-2 also induced p38 mitogen-activated protein kinase (MAPK), which is necessary for BMP-2- and retinoid-induced apoptosis. Retinoid-resistant medulloblastoma cells underwent apoptosis when treated with BMP-2 or when cultured with retinoid-sensitive medulloblastoma cells. Retinoid-induced expression of BMP-2 is thus necessary and sufficient for apoptosis of retinoid-responsive cells, and expression of BMP-2 by retinoid-sensitive cells is sufficient to induce apoptosis in surrounding retinoid-resistant cells.


Subject(s)
Apoptosis , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/metabolism , Medulloblastoma/metabolism , Paracrine Communication , Retinoids/pharmacology , Transforming Growth Factor beta , Animals , Bone Morphogenetic Protein 2 , Bone Morphogenetic Protein Receptors , Brain Neoplasms/pathology , Female , Gene Expression Profiling , Humans , Medulloblastoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Receptors, Growth Factor/genetics , Receptors, Growth Factor/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
17.
Hum Mol Genet ; 11(19): 2207-21, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12217949

ABSTRACT

We present a simple method to assign approximate P-values to gene expression changes detected with Affymetrix oligonucleotide arrays and software. The method pools data for groups of genes and a small number of like-to-like comparisons in order to estimate the significance of changes observed for single genes in comparisons of experimental interest. Statistical significance levels are based on the observed variability in the fractional majority of probe pairs that indicate increasing or decreasing differential expression in comparisons of technical replicates. From this reference distribution or error model, we compute the expected frequency for fractional majorities in comparisons for N > or = 2. These computed distributions are the source of P-value estimates for changes seen in the experimental comparisons. The method is intended to complement the Affymetrix software and to rationalize gene selection for experimental designs involving limited replication.


Subject(s)
Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Animals , Data Interpretation, Statistical , Disease Models, Animal , Mice
18.
Hum Mol Genet ; 11(19): 2223-32, 2002 Sep 15.
Article in English | MEDLINE | ID: mdl-12217950

ABSTRACT

A randomization procedure to evaluate the significance level and the false-discovery rate in complex microarray experiments is proposed. A related graph can be used to compare different test statistics that can be used to analyze the same experiment. This graph is closely related to receiver operator characteristic (ROC) curves. The proposed method is applied to a subset of the data from a cell-line experiment related to Huntington's disease. A small simulation study compares the effectiveness of the proposed procedure with the significance analysis of microarrays (SAM) procedure.


Subject(s)
Data Interpretation, Statistical , Oligonucleotide Array Sequence Analysis/methods , Computer Simulation , Huntington Disease/genetics , Models, Genetic , ROC Curve , Random Allocation , Regression Analysis
19.
Hum Mol Genet ; 11(17): 1911-26, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12165554

ABSTRACT

Previous analyses of gene expression in a mouse model of Huntington's disease (R6/2) indicated that an N-terminal fragment of mutant huntingtin causes downregulation of striatal signaling genes and particularly those normally induced by cAMP and retinoic acid. The present study expands the regional and temporal scope of this previous work by assessing whether similar changes occur in other brain regions affected in Huntington's disease and other polyglutamine diseases and by discerning whether gene expression changes precede the appearance of disease signs. Oligonucleotide microarrays were employed to survey the expression of approximately 11,000 mRNAs in the cerebral cortex, cerebellum and striatum of symptomatic R6/2 mice. The number and nature of gene expression changes were similar among these three regions, influenced as expected by regional differences in baseline gene expression. Time-course studies revealed that mRNA changes could only reliably be detected after 4 weeks of age, coincident with development of early pathologic and behavioral changes in these animals. In addition, we discovered that skeletal muscle is also a target of polyglutamine-related perturbations in gene expression, showing changes in mRNAs that are dysregulated in brain and also muscle-specific mRNAs. The complete dataset is available at www.neumetrix.info.


Subject(s)
Brain/metabolism , Huntington Disease/genetics , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Peptides/genetics , Animals , Base Sequence , Blotting, Northern , Blotting, Western , Brain/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , Immunoenzyme Techniques , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Muscle, Skeletal/pathology , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Peptides/metabolism , RNA, Messenger/metabolism
20.
Hum Mol Genet ; 11(17): 1927-37, 2002 Aug 15.
Article in English | MEDLINE | ID: mdl-12165555

ABSTRACT

Recent evidence indicates that transcriptional abnormalities may play an important role in the pathophysiology of polyglutamine diseases. In the present study, we have explored the extent to which polyglutamine-related changes in gene expression may be independent of protein context by comparing mouse models of dentatorubral-pallidoluysian atrophy (DRPLA) and Huntington's disease (HD). Microarray gene expression profiling was conducted in mice of the same background strain in which the same promoter was employed to direct the expression of full-length atrophin-1 or partial huntingtin transproteins (At-65Q or N171-82Q mice). A large number of overlapping gene expression changes were observed in the cerebella of At-65Q and N171-82Q mice. Six of the gene expression changes common to both huntingtin and atrophin-1 transgenic mice were also observed in the cerebella of mouse models expressing full-length mutant ataxin-7 or the androgen receptor. These results demonstrate that some of the gene expression effects of expanded polyglutamine proteins occur independently of protein context.


Subject(s)
Brain/metabolism , Gene Expression Regulation , Huntington Disease/genetics , Myoclonic Epilepsies, Progressive/genetics , Nerve Tissue Proteins/genetics , Peptides/genetics , Animals , Ataxin-7 , Base Sequence , Blotting, Northern , Brain/pathology , DNA-Binding Proteins , Disease Models, Animal , Humans , Huntington Disease/metabolism , Huntington Disease/pathology , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Myoclonic Epilepsies, Progressive/metabolism , Myoclonic Epilepsies, Progressive/pathology , Nerve Tissue Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Peptides/metabolism , RNA, Messenger/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Transcription, Genetic , Trinucleotide Repeat Expansion
SELECTION OF CITATIONS
SEARCH DETAIL