Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Hum Mol Genet ; 29(13): 2109-2123, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32186706

ABSTRACT

Cobalamin C (cblC) deficiency, the most common inborn error of intracellular cobalamin metabolism, is caused by mutations in MMACHC, a gene responsible for the processing and intracellular trafficking of vitamin B12. This recessive disorder is characterized by a failure to metabolize cobalamin into adenosyl- and methylcobalamin, which results in the biochemical perturbations of methylmalonic acidemia, hyperhomocysteinemia and hypomethioninemia caused by the impaired activity of the downstream enzymes, methylmalonyl-CoA mutase and methionine synthase. Cobalamin C deficiency can be accompanied by a wide spectrum of clinical manifestations, including progressive blindness, and, in mice, manifests with very early embryonic lethality. Because zebrafish harbor a full complement of cobalamin metabolic enzymes, we used genome editing to study the loss of mmachc function and to develop the first viable animal model of cblC deficiency. mmachc mutants survived the embryonic period but perished in early juvenile life. The mutants displayed the metabolic and clinical features of cblC deficiency including methylmalonic acidemia, severe growth retardation and lethality. Morphologic and metabolic parameters improved when the mutants were raised in water supplemented with small molecules used to treat patients, including hydroxocobalamin, methylcobalamin, methionine and betaine. Furthermore, mmachc mutants bred to express rod and/or cone fluorescent reporters, manifested a retinopathy and thin optic nerves (ON). Expression analysis using whole eye mRNA revealed the dysregulation of genes involved in phototransduction and cholesterol metabolism. Zebrafish with mmachc deficiency recapitulate the several of the phenotypic and biochemical features of the human disorder, including ocular pathology, and show a response to established treatments.


Subject(s)
Carrier Proteins/genetics , Morphogenesis/genetics , Vitamin B 12 Deficiency/genetics , Vitamin B 12/genetics , Zebrafish Proteins/genetics , Animals , Homocystinuria/genetics , Homocystinuria/pathology , Humans , Mice , Mutation/genetics , Optic Nerve/growth & development , Optic Nerve/pathology , Oxidoreductases/genetics , Retina/growth & development , Retina/metabolism , Vitamin B 12/analogs & derivatives , Vitamin B 12/metabolism , Vitamin B 12 Deficiency/metabolism , Vitamin B 12 Deficiency/pathology , Zebrafish/genetics , Zebrafish/growth & development
2.
JCI Insight ; 3(23)2018 12 06.
Article in English | MEDLINE | ID: mdl-30518688

ABSTRACT

Methylmalonic acidemia (MMA), an organic acidemia characterized by metabolic instability and multiorgan complications, is most frequently caused by mutations in methylmalonyl-CoA mutase (MUT). To define the metabolic adaptations in MMA in acute and chronic settings, we studied a mouse model generated by transgenic expression of Mut in the muscle. Mut-/-;TgINS-MCK-Mut mice accurately replicate the hepatorenal mitochondriopathy and growth failure seen in severely affected patients and were used to characterize the response to fasting. The hepatic transcriptome in MMA mice was characterized by the chronic activation of stress-related pathways and an aberrant fasting response when compared with controls. A key metabolic regulator, Fgf21, emerged as a significantly dysregulated transcript in mice and was subsequently studied in a large patient cohort. The concentration of plasma FGF21 in MMA patients correlated with disease subtype, growth indices, and markers of mitochondrial dysfunction but was not affected by renal disease. Restoration of liver Mut activity, by transgenesis and liver-directed gene therapy in mice or liver transplantation in patients, drastically reduced plasma FGF21 and was associated with improved outcomes. Our studies identify mitocellular hormesis as a hepatic adaptation to metabolic stress in MMA and define FGF21 as a highly predictive disease biomarker.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Fibroblast Growth Factors/metabolism , Hormesis , Methylmalonyl-CoA Mutase/metabolism , Stress, Physiological , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Biomarkers/blood , Disease Models, Animal , Female , Fibroblast Growth Factors/blood , Genetic Therapy , Humans , Kidney Diseases/metabolism , Liver/metabolism , Liver/pathology , Liver Transplantation , Male , Methylmalonyl-CoA Mutase/genetics , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mitochondria/pathology , Phenotype , Transcriptome
3.
PLoS Biol ; 16(5): e2003648, 2018 05.
Article in English | MEDLINE | ID: mdl-29723194

ABSTRACT

Melanocyte stem cells (McSCs) and mouse models of hair graying serve as useful systems to uncover mechanisms involved in stem cell self-renewal and the maintenance of regenerating tissues. Interested in assessing genetic variants that influence McSC maintenance, we found previously that heterozygosity for the melanogenesis associated transcription factor, Mitf, exacerbates McSC differentiation and hair graying in mice that are predisposed for this phenotype. Based on transcriptome and molecular analyses of Mitfmi-vga9/+ mice, we report a novel role for MITF in the regulation of systemic innate immune gene expression. We also demonstrate that the viral mimic poly(I:C) is sufficient to expose genetic susceptibility to hair graying. These observations point to a critical suppressor of innate immunity, the consequences of innate immune dysregulation on pigmentation, both of which may have implications in the autoimmune, depigmenting disease, vitiligo.


Subject(s)
Adult Stem Cells , Hair Color/immunology , Immunity, Innate , Melanocytes , Microphthalmia-Associated Transcription Factor/physiology , Animals , Female , Gene Expression Regulation , Genetic Predisposition to Disease , Hair Color/genetics , Interferon Type I/metabolism , Mice , Mice, Transgenic , Poly I-C
4.
Appetite ; 111: 142-150, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28043857

ABSTRACT

Common reports of over-response to food cues, difficulties with calorie restriction, and difficulty adhering to dietary guidelines suggest that eating behaviors could be interrelated in ways that influence weight management efforts. The feasibility of identifying robust eating phenotypes (showing face, content, and criterion validity) was explored based on well-validated individual eating behavior assessments. Adults (n = 260; mean age 34 years) completed online questionnaires with measurements of nine eating behaviors including: appetite for palatable foods, binge eating, bitter taste sensitivity, disinhibition, food neophobia, pickiness and satiety responsiveness. Discovery-based visualization procedures that have the combined strengths of heatmaps and hierarchical clustering were used to investigate: 1) how eating behaviors cluster, 2) how participants can be grouped within eating behavior clusters, and 3) whether group clustering is associated with body mass index (BMI) and dietary self-efficacy levels. Two distinct eating behavior clusters and participant groups that aligned within these clusters were identified: one with higher drive to eat and another with food avoidance behaviors. Participants' BMI (p = 0.0002) and dietary self-efficacy (p < 0.0001) were associated with cluster membership. Eating behavior clusters showed content and criterion validity based on their association with BMI (associated, but not entirely overlapping) and dietary self-efficacy. Identifying eating behavior phenotypes appears viable. These efforts could be expanded and ultimately inform tailored weight management interventions.


Subject(s)
Eating/psychology , Feeding Behavior/psychology , Phenotype , Self Efficacy , Adult , Appetite , Body Mass Index , Bulimia/psychology , Cluster Analysis , Feasibility Studies , Female , Humans , Inhibition, Psychological , Male , Satiation , Surveys and Questionnaires , Taste Perception
5.
Nat Med ; 22(4): 388-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26998835

ABSTRACT

Smith-Lemli-Opitz syndrome (SLOS) is a malformation disorder caused by mutations in DHCR7, which impair the reduction of 7-dehydrocholesterol (7DHC) to cholesterol. SLOS results in cognitive impairment, behavioral abnormalities and nervous system defects, though neither affected cell types nor impaired signaling pathways are fully understood. Whether 7DHC accumulation or cholesterol loss is primarily responsible for disease pathogenesis is also unclear. Using induced pluripotent stem cells (iPSCs) from subjects with SLOS, we identified cellular defects that lead to precocious neuronal specification within SLOS derived neural progenitors. We also demonstrated that 7DHC accumulation, not cholesterol deficiency, is critical for SLOS-associated defects. We further identified downregulation of Wnt/ß-catenin signaling as a key initiator of aberrant SLOS iPSC differentiation through the direct inhibitory effects of 7DHC on the formation of an active Wnt receptor complex. Activation of canonical Wnt signaling prevented the neural phenotypes observed in SLOS iPSCs, suggesting that Wnt signaling may be a promising therapeutic target for SLOS.


Subject(s)
Cell Differentiation/genetics , Induced Pluripotent Stem Cells/metabolism , Smith-Lemli-Opitz Syndrome/genetics , Wnt Signaling Pathway/genetics , Animals , Cholesterol/biosynthesis , Cholesterol/metabolism , Dehydrocholesterols/metabolism , Humans , Induced Pluripotent Stem Cells/transplantation , Mice , Mutation , Neurons/metabolism , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Smith-Lemli-Opitz Syndrome/metabolism , Smith-Lemli-Opitz Syndrome/pathology
6.
Aging Cell ; 15(2): 267-78, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26685868

ABSTRACT

Alternative splicing (AS) is a key regulatory mechanism for the development of different tissues; however, not much is known about changes to alternative splicing during aging. Splicing events may become more frequent and widespread genome-wide as tissues age and the splicing machinery stringency decreases. Using skin, skeletal muscle, bone, thymus, and white adipose tissue from wild-type C57BL6/J male mice (4 and 18 months old), we examined the effect of age on splicing by AS analysis of the differential exon usage of the genome. The results identified a considerable number of AS genes in skeletal muscle, thymus, bone, and white adipose tissue between the different age groups (ranging from 27 to 246 AS genes corresponding to 0.3-3.2% of the total number of genes analyzed). For skin, skeletal muscle, and bone, we included a later age group (28 months old) that showed that the number of alternatively spliced genes increased with age in all three tissues (P < 0.01). Analysis of alternatively spliced genes across all tissues by gene ontology and pathway analysis identified 158 genes involved in RNA processing. Additional analysis of AS in a mouse model for the premature aging disease Hutchinson-Gilford progeria syndrome was performed. The results show that expression of the mutant protein, progerin, is associated with an impaired developmental splicing. As progerin accumulates, the number of genes with AS increases compared to in wild-type skin. Our results indicate the existence of a mechanism for increased AS during aging in several tissues, emphasizing that AS has a more important role in the aging process than previously known.


Subject(s)
Aging, Premature/genetics , Aging/genetics , Alternative Splicing/genetics , Animals , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/pathology , Skin/pathology , Skin Physiological Phenomena/genetics
7.
J Endocrinol Metab ; 5(3): 199-210, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26504501

ABSTRACT

BACKGROUND: Adipose tissues play important role in the pathophysiology of obesity-related diseases including type 2 diabetes (T2D). To describe gene expression patterns and functional pathways in obesity-related T2D, we performed global transcript profiling of omental adipose tissue (OAT) in morbidly obese individuals with or without T2D. METHODS: Twenty morbidly obese (mean BMI: about 54 kg/m2) subjects were studied, including 14 morbidly obese individuals with T2D (cases) and 6 morbidly obese individuals without T2D (reference group). Gene expression profiling was performed using the Affymetrix U133 Plus 2.0 human genome expression array. Analysis of covariance was performed to identify differentially expressed genes (DEGs). Bioinformatics tools including PANTHER and Ingenuity Pathway Analysis (IPA) were applied to the DEGs to determine biological functions, networks and canonical pathways that were overrepresented in these individuals. RESULTS: At an absolute fold-change threshold of 2 and false discovery rate (FDR) < 0.05, 68 DEGs were identified in cases compared to the reference group. Myosin X (MYO10) and transforming growth factor beta regulator 1 (TBRG1) were upregulated. MYO10 encodes for an actin-based motor protein that has been associated with T2D. Telomere extension by telomerase (HNRNPA1, TNKS2), D-myo-inositol (1, 4, 5)-trisphosphate biosynthesis (PIP5K1A, PIP4K2A), and regulation of actin-based motility by Rho (ARPC3) were the most significant canonical pathways and overlay with T2D signaling pathway. Upstream regulator analysis predicted 5 miRNAs (miR-320b, miR-381-3p, miR-3679-3p, miR-494-3p, and miR-141-3p,) as regulators of the expression changes identified. CONCLUSION: This study identified a number of transcripts and miRNAs in OAT as candidate novel players in the pathophysiology of T2D in African Americans.

8.
Oncotarget ; 6(31): 31557-68, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26384308

ABSTRACT

Metastasis is the primary cause of death in breast cancer. Earlier studies using a mammary tumorigenesis mouse model identified Necdin (Ndn)as a germline modifier of metastasis. Differential expression of Ndn induces a gene-expression signature that predicts prognosis in human breast cancer. Additionally, a non-synonymous germline single nucleotide polymorphism (T50C; V17A) in Ndn distinguishes mouse strains with differing metastatic capacities. To better understand how hereditary factors influence metastasis in breast cancer, we characterized NDN-mediated transcription. Haplotype analysis in a well-characterized breast cancer cohort revealed that NDN germline variation is associated with both NDN expression levels and patient outcome. To examine the role of NDN in mammary tumor metastasis and transcriptional regulation, mouse mammary tumor cell lines stably over-expressing either the wildtype 50T or variant 50C Ndn allele were generated. Cells over-expressing Ndn 50T, but not Ndn 50C, exhibited significant decrease in cell invasiveness and pulmonary metastases compared to control cells. Transcriptome analyses identified a 71-gene expression signature that distinguishes cells over-expressing the two Ndn allelic variants. Furthermore, ChIP assays revealed c-Myc, a target gene of NDN, to be differentially regulated by the allelic variants. These data demonstrate that NDN and the T50C allele regulate gene expression and metastasis efficiency.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Germ-Line Mutation/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Animals , Breast Neoplasms/genetics , Cell Proliferation , Chromatin Immunoprecipitation , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Staging , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Prognosis , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
9.
J Clin Invest ; 125(2): 870-80, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25607839

ABSTRACT

The use of adeno-associated virus (AAV) as a gene therapy vector has been approved recently for clinical use and has demonstrated efficacy in a growing number of clinical trials. However, the safety of AAV as a vector has been challenged by a single study that documented hepatocellular carcinoma (HCC) after AAV gene delivery in mice. Most studies have not noted genotoxicity following AAV-mediated gene delivery; therefore, the possibility that there is an association between AAV and HCC is controversial. Here, we performed a comprehensive study of HCC in a large number of mice following therapeutic AAV gene delivery. Using a sensitive high-throughput integration site-capture technique and global expressional analysis, we found that AAV integration into the RNA imprinted and accumulated in nucleus (Rian) locus, and the resulting overexpression of proximal microRNAs and retrotransposon-like 1 (Rtl1) were associated with HCC. In addition, we demonstrated that the AAV vector dose, enhancer/promoter selection, and the timing of gene delivery are all critical factors for determining HCC incidence after AAV gene delivery. Together, our results define aspects of AAV-mediated gene therapy that influence genotoxicity and suggest that these features should be considered for design of both safer AAV vectors and gene therapy studies.


Subject(s)
Carcinoma, Hepatocellular , Dependovirus , Genetic Therapy/methods , Genetic Vectors , Liver Neoplasms , Transduction, Genetic , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Mice , Mice, Mutant Strains , Pregnancy Proteins/genetics , Pregnancy Proteins/metabolism
10.
Mol Cancer Res ; 12(12): 1818-28, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25092915

ABSTRACT

UNLABELLED: Overexpression of ribosomal RNA processing 1 homolog B (RRP1B) induces a transcriptional profile that accurately predicts patient outcome in breast cancer. However, the mechanism by which RRP1B modulates transcription is unclear. Here, the chromatin-binding properties of RRP1B were examined to define how it regulates metastasis-associated transcription. To identify genome-wide RRP1B-binding sites, high-throughput ChIP-seq was performed in the human breast cancer cell line MDA-MB-231 and HeLa cells using antibodies against endogenous RRP1B. Global changes in repressive marks such as histone H3 lysine 9 trimethylation (H3K9me3) were also examined by ChIP-seq. Analysis of these samples identified 339 binding regions in MDA-MB-231 cells and 689 RRP1B-binding regions in HeLa cells. Among these, 136 regions were common to both cell lines. Gene expression analyses of these RRP1B-binding regions revealed that transcriptional repression is the primary result of RRP1B binding to chromatin. ChIP-reChIP assays demonstrated that RRP1B co-occupies loci with decreased gene expression with the heterochromatin-associated proteins, tripartite motif-containing protein 28 (TRIM28/KAP1), and heterochromatin protein 1-α (CBX5/HP1α). RRP1B occupancy at these loci was also associated with higher H3K9me3 levels, indicative of heterochromatinization mediated by the TRIM28/HP1α complex. In addition, RRP1B upregulation, which is associated with metastasis suppression, induced global changes in histone methylation. IMPLICATIONS: RRP1B, a breast cancer metastasis suppressor, regulates gene expression through heterochromatinization and transcriptional repression, which helps our understanding of mechanisms that drive prognostic gene expression in human breast cancer.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/pathology , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Repressor Proteins/genetics , Animals , Apoptosis Regulatory Proteins/chemistry , Binding Sites , Breast Neoplasms/genetics , Cell Line, Tumor , Chromatin/metabolism , Chromatin Immunoprecipitation , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/chemistry , Epigenesis, Genetic , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Histones/metabolism , Humans , Lung Neoplasms/pathology , Methylation , Mice , Neoplasms, Experimental , Transcription, Genetic , Tripartite Motif-Containing Protein 28
11.
Blood ; 124(12): 1926-30, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25114263

ABSTRACT

Familial platelet disorder with predisposition to acute myeloid leukemia (FPD/AML) is an autosomal dominant disease of the hematopoietic system that is caused by heterozygous mutations in RUNX1. FPD/AML patients have a bleeding disorder characterized by thrombocytopenia with reduced platelet numbers and functions, and a tendency to develop AML. No suitable animal models exist for FPD/AML, as Runx11/2 mice and zebra fish do not develop bleeding disorders or leukemia. Here we derived induced pluripotent stem cells (iPSCs) from 2 patients in a family with FPD/AML, and found that the FPD iPSCs display defects in megakaryocytic differentiation in vitro. We corrected the RUNX1 mutation in 1 FPD iPSC line through gene targeting, which led to normalization of megakaryopoiesis of the iPSCs in culture. Our results demonstrate successful in vitro modeling of FPD with patient-specific iPSCs and confirm that RUNX1 mutations are responsible for megakaryopoietic defects in FPD patients.


Subject(s)
Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/therapy , Blood Platelet Disorders/genetics , Blood Platelet Disorders/therapy , Core Binding Factor Alpha 2 Subunit/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Mutation, Missense , Targeted Gene Repair/methods , Animals , Blood Coagulation Disorders, Inherited/pathology , Blood Platelet Disorders/pathology , Core Binding Factor Alpha 2 Subunit/chemistry , Gene Expression Profiling , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Induced Pluripotent Stem Cells/transplantation , Leukemia, Myeloid, Acute/pathology , Mice , Thrombopoiesis/genetics
12.
Pigment Cell Melanoma Res ; 27(5): 777-87, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24767210

ABSTRACT

The complex genetic changes underlying metastatic melanoma need to be deciphered to develop new and effective therapeutics. Previously, genome-wide microarray analyses of human melanoma identified two reciprocal gene expression programs, including transcripts regulated by either transforming growth factor, beta 1 (TGFß1) pathways, or microphthalmia-associated transcription factor (MITF)/SRY-box containing gene 10 (SOX10) pathways. We extended this knowledge by discovering that melanoma cell lines with these two expression programs exhibit distinctive microRNA (miRNA) expression patterns. We also demonstrated that hypoxia-inducible factor 1 alpha (HIF1A) is increased in TGFß1 pathway-expressing melanoma cells and that HIF1A upregulates miR-210, miR-218, miR-224, and miR-452. Reduced expression of these four miRNAs in TGFß1 pathway-expressing melanoma cells arrests the cell cycle, while their overexpression in mouse melanoma cells increases the expression of the hypoxic response gene Bnip3. Taken together, these data suggest that HIF1A may regulate some of the gene expression and biological behavior of TGFß1 pathway-expressing melanoma cells, in part via alterations in these four miRNAs.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , MicroRNAs/metabolism , Animals , Cell Cycle , Cell Line, Tumor , Cluster Analysis , Genome , Humans , Melanoma/pathology , Membrane Proteins/metabolism , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Mitochondrial Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Transforming Growth Factor beta1/metabolism , Up-Regulation
13.
Proc Natl Acad Sci U S A ; 110(33): 13552-7, 2013 Aug 13.
Article in English | MEDLINE | ID: mdl-23898205

ABSTRACT

Isolated methylmalonic acidemia (MMA), caused by deficiency of the mitochondrial enzyme methylmalonyl-CoA mutase (MUT), is often complicated by end stage renal disease that is resistant to conventional therapies, including liver transplantation. To establish a viable model of MMA renal disease, Mut was expressed in the liver of Mut(-/-) mice as a stable transgene under the control of an albumin (INS-Alb-Mut) promoter. Mut(-/-);Tg(INS-Alb-Mut) mice, although completely rescued from neonatal lethality that was displayed by Mut(-/-) mice, manifested a decreased glomerular filtration rate (GFR), chronic tubulointerstitial nephritis and ultrastructural changes in the proximal tubule mitochondria associated with aberrant tubular function, as demonstrated by single-nephron GFR studies. Microarray analysis of Mut(-/-);Tg(INS-Alb-Mut) kidneys identified numerous biomarkers, including lipocalin-2, which was then used to monitor the response of the GFR to antioxidant therapy in the mouse model. Renal biopsies and biomarker analysis from a large and diverse patient cohort (ClinicalTrials.gov identifier: NCT00078078) precisely replicated the findings in the animals, establishing Mut(-/-);Tg(INS-Alb-Mut) mice as a unique model of MMA renal disease. Our studies suggest proximal tubular mitochondrial dysfunction is a key pathogenic mechanism of MMA-associated kidney disease, identify lipocalin-2 as a biomarker of increased oxidative stress in the renal tubule, and demonstrate that antioxidants can attenuate the renal disease of MMA.


Subject(s)
Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/enzymology , Antioxidants/pharmacology , Disease Models, Animal , Kidney Tubules, Proximal/physiopathology , Methylmalonyl-CoA Mutase/deficiency , Amino Acid Metabolism, Inborn Errors/pathology , Animals , Antioxidants/therapeutic use , Biomarkers/metabolism , Blotting, Western , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Fluorescein-5-isothiocyanate , Genotype , Glomerular Filtration Rate/genetics , Humans , Immunohistochemistry , Methylmalonyl-CoA Mutase/genetics , Methylmalonyl-CoA Mutase/metabolism , Mice , Mice, Knockout , Microarray Analysis , Microscopy, Electron, Transmission , Nephritis, Interstitial/genetics , Real-Time Polymerase Chain Reaction , Transgenes/genetics , Ubiquinone/pharmacology
14.
Mol Genet Metab ; 110(1-2): 188-90, 2013.
Article in English | MEDLINE | ID: mdl-23850077

ABSTRACT

Mutations in individuals with the lysosomal storage disorder Niemann-Pick disease, type C1 (NPC1) are heterogeneous, not localized to specific protein domains, and not correlated to time of onset or disease severity. We demonstrate direct correlation of the time of neurological symptom onset with the severity of lysosomal defects in NPC1 patient-derived fibroblasts. This is a novel assay for NPC1 individuals that may be predictive of NPC1 disease progression and broadly applicable to other lysosomal disorders.


Subject(s)
Lysosomal Storage Diseases/genetics , Lysosomes/metabolism , Membrane Glycoproteins/genetics , Niemann-Pick Disease, Type C/genetics , Adolescent , Adult , Biological Transport/genetics , Cells, Cultured , Child , Child, Preschool , Disease Progression , Female , Fibroblasts , Humans , Infant , Infant, Newborn , Lysosomal Storage Diseases/metabolism , Lysosomal Storage Diseases/pathology , Lysosomes/genetics , Lysosomes/pathology , Male , Membrane Glycoproteins/metabolism , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Niemann-Pick Disease, Type C/metabolism , Niemann-Pick Disease, Type C/pathology , Protein Structure, Tertiary
15.
BMC Genomics ; 13: 505, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-23006843

ABSTRACT

BACKGROUND: Metastasis is characterized by spreading of neoplastic cells to an organ other than where they originated and is the predominant cause of death among cancer patients. This holds true for melanoma, whose incidence is increasing more rapidly than any other cancer and once disseminated has few therapeutic options. Here we performed whole exome sequencing of two sets of matched normal and metastatic tumor DNAs. RESULTS: Using stringent criteria, we evaluated the similarities and differences between the lesions. We find that in both cases, 96% of the single nucleotide variants are shared between the two metastases indicating that clonal populations gave rise to the distant metastases. Analysis of copy number variation patterns of both metastatic sets revealed a trend similar to that seen with our single nucleotide variants. Analysis of pathway enrichment on tumor sets shows commonly mutated pathways enriched between individual sets of metastases and all metastases combined. CONCLUSIONS: These data provide a proof-of-concept suggesting that individual metastases may have sufficient similarity for successful targeting of driver mutations.


Subject(s)
DNA/analysis , Exome/genetics , Melanoma/genetics , Melanoma/pathology , Sequence Analysis, DNA , Cell Line, Tumor , DNA Copy Number Variations , DNA Mutational Analysis , Humans , Neoplasm Metastasis , Polymorphism, Single Nucleotide
16.
Exp Biol Med (Maywood) ; 236(5): 628-36, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21540249

ABSTRACT

Gene expression profiles consisting of ten thousands of transcripts are used for clustering of tissue, such as tumors, into subtypes, often without considering the underlying reason that the distinct patterns of expression arise because of constraints in the realization of gene expression profiles imposed by the gene regulatory network. The topology of this network has been suggested to consist of a regulatory core of genes represented most prominently by transcription factors (TFs) and microRNAs, that influence the expression of other genes, and of a periphery of 'enslaved' effector genes that are regulated but not regulating. This 'medusa' architecture implies that the core genes are much stronger determinants of the realized gene expression profiles. To test this hypothesis, we examined the clustering of gene expression profiles into known tumor types to quantitatively demonstrate that TFs, and even more pronounced, microRNAs, are much stronger discriminators of tumor type specific gene expression patterns than a same number of randomly selected or metabolic genes. These findings lend support to the hypothesis of a medusa architecture and of the canalizing nature of regulation by microRNAs. They also reveal the degree of freedom for the expression of peripheral genes that are less stringently associated with a tissue type specific global gene expression profile.


Subject(s)
Gene Regulatory Networks/genetics , Neoplasms/classification , Neoplasms/genetics , Transcription Factors/metabolism , Cluster Analysis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Neoplasm/genetics , Humans , Lung Neoplasms/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Principal Component Analysis , Transcription Factors/genetics
17.
J Neurosci Res ; 72(6): 756-67, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12774316

ABSTRACT

Alcohol abuse is a common human disorder with high rate of comorbidity with other psychiatric disorders. To identify candidate mechanisms for alcohol abuse, the expression of 12,626 genes was measured in postmortem temporal cortex from 11 subjects with a history of alcohol abuse or dependence, with or without other psychiatric diagnoses and compared pairwise with the expression in 11 nonalcoholic subjects matched for the other psychiatric diagnoses and demographics. Genes were defined to have altered expression in alcohol abuse if: 1) the gene showed decreased expression in at least 10 of 11 subjects with alcohol abuse, or showed increased expression in at least 10 of 11 subjects with this diagnosis compared to matched non-abusers (P < 0.007, chi(2)test); or 2) the difference in the mean abuser/non-abuser ratio for the gene from value of 1.0 was significant at P < 0.05 (one sample t-test). In subjects with a history of alcohol abuse or dependence, 163 genes were changed significantly. The most abundant and consistent changes were in gene families encoding mitochondrial proteins, the ubiquitin system, and signal transduction. These alterations indicate disturbances in energy metabolism and multiple signaling mechanisms in the temporal cortex of subjects with a history of alcohol abuse or dependence. We hypothesize that these mechanisms may be related to alcohol abuse traits or long-term effects of alcohol.


Subject(s)
Alcoholism/genetics , Brain Chemistry/genetics , Gene Expression Profiling/methods , Mitochondria/genetics , Signal Transduction/genetics , Transcription, Genetic , Ubiquitin/genetics , Adult , Alcoholism/metabolism , Chi-Square Distribution , Female , Gene Expression Profiling/statistics & numerical data , Gene Expression Regulation/physiology , Humans , Male , Middle Aged , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis/methods , Ubiquitin/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...