Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Endocrinology ; 155(5): 1793-805, 2014 May.
Article in English | MEDLINE | ID: mdl-24517230

ABSTRACT

GH is an important regulator of body growth and composition as well as numerous other metabolic processes. In particular, liver plays a key role in the GH/IGF-I axis, because the majority of circulating "endocrine" IGF-I results from GH-stimulated liver IGF-I production. To develop a better understanding of the role of liver in the overall function of GH, we generated a strain of mice with liver-specific GH receptor (GHR) gene knockout (LiGHRKO mice). LiGHRKO mice had a 90% decrease in circulating IGF-I levels, a 300% increase in circulating GH, and significant changes in IGF binding protein (IGFBP)-1, IGFBP-2, IGFBP-3, IGFBP-5, and IGFBP-7. LiGHRKO mice were smaller than controls, with body length and body weight being significantly decreased in both sexes. Analysis of body composition over time revealed a pattern similar to those found in GH transgenic mice; that is, LiGHRKO mice had a higher percentage of body fat at early ages followed by lower percentage of body fat in adulthood. Local IGF-I mRNA levels were significantly increased in skeletal muscle and select adipose tissue depots. Grip strength was increased in LiGHRKO mice. Finally, circulating levels of leptin, resistin, and adiponectin were increased in LiGHRKO mice. In conclusion, LiGHRKO mice are smaller despite increased local mRNA expression of IGF-I in several tissues, suggesting that liver-derived IGF-I is indeed important for normal body growth. Furthermore, our data suggest that novel GH-dependent cross talk between liver and adipose is important for regulation of adipokines in vivo.


Subject(s)
Adipokines/metabolism , Aging , Endocrine Glands/metabolism , Growth Hormone/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Receptors, Somatotropin/metabolism , Adipose Tissue, White/growth & development , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Composition , Body Size , Female , Growth Hormone/blood , Insulin-Like Growth Factor I/genetics , Liver/growth & development , Male , Mice , Mice, Knockout , Mice, Transgenic , Muscle Development , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , RNA, Messenger/metabolism , Receptors, Somatotropin/genetics , Sex Characteristics , Signal Transduction
2.
Mol Cell Endocrinol ; 386(1-2): 34-45, 2014 Apr 05.
Article in English | MEDLINE | ID: mdl-24035867

ABSTRACT

The discovery of a growth hormone receptor antagonist (GHA) was initially established via expression of mutated GH genes in transgenic mice. Following this discovery, development of the compound resulted in a drug termed pegvisomant, which has been approved for use in patients with acromegaly. Pegvisomant treatment in a dose dependent manner results in normalization of IGF-1 levels in most patients. Thus, it is a very efficacious and safe drug. Since the GH/IGF-1 axis has been implicated in the progression of several types of cancers, many have suggested the use of pegvisomant as an anti-cancer therapeutic. In this manuscript, we will review the use of mouse strains that possess elevated or depressed levels of GH action for unraveling many of GH actions. Additionally, we will describe experiments in which the GHA was discovered, review results of pegvisomant's preclinical and clinical trials, and provide data suggesting pegvisomant's therapeutic value in selected types of cancer.


Subject(s)
Hormone Antagonists/pharmacology , Hormone Antagonists/therapeutic use , Receptors, Somatotropin/antagonists & inhibitors , Amino Acid Sequence , Animals , Growth Hormone/metabolism , Human Growth Hormone/analogs & derivatives , Human Growth Hormone/chemistry , Human Growth Hormone/pharmacology , Human Growth Hormone/therapeutic use , Insulin-Like Growth Factor I/metabolism , Mice , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Somatotropin/metabolism
3.
Mol Endocrinol ; 27(3): 524-35, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23349524

ABSTRACT

GH receptor (GHR) gene-disrupted mice (GHR-/-) have provided countless discoveries as to the numerous actions of GH. Many of these discoveries highlight the importance of GH in adipose tissue. For example GHR-/- mice are insulin sensitive yet obese with preferential enlargement of the sc adipose depot. GHR-/- mice also have elevated levels of leptin, resistin, and adiponectin, compared with controls leading some to suggest that GH may negatively regulate certain adipokines. To help clarify the role that GH exerts specifically on adipose tissue in vivo, we selectively disrupted GHR in adipose tissue to produce Fat GHR Knockout (FaGHRKO) mice. Surprisingly, FaGHRKOs shared only a few characteristics with global GHR-/- mice. Like the GHR-/- mice, FaGHRKO mice are obese with increased total body fat and increased adipocyte size. However, FaGHRKO mice have increases in all adipose depots with no improvements in measures of glucose homeostasis. Furthermore, resistin and adiponectin levels in FaGHRKO mice are similar to controls (or slightly decreased) unlike the increased levels found in GHR-/- mice, suggesting that GH does not regulate these adipokines directly in adipose tissue in vivo. Other features of FaGHRKO mice include decreased levels of adipsin, a near-normal GH/IGF-1 axis, and minimal changes to a large assortment of circulating factors that were measured such as IGF-binding proteins. In conclusion, specific removal of GHR in adipose tissue is sufficient to increase adipose tissue and decrease circulating adipsin. However, removal of GHR in adipose tissue alone is not sufficient to increase levels of resistin or adiponectin and does not alter glucose metabolism.


Subject(s)
Adipose Tissue/metabolism , Gene Deletion , Growth Hormone/metabolism , Receptors, Somatotropin/metabolism , Adipocytes/cytology , Adipocytes/metabolism , Adipokines/blood , Adiposity , Animals , Body Composition , Body Weight , Cell Count , Cell Size , Cytokines/blood , Female , Glucose/metabolism , Homeostasis , Insulin-Like Growth Factor Binding Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Male , Mice , Mice, Knockout , Organ Size , Organ Specificity , Triglycerides/metabolism
4.
J Pharmacol Exp Ther ; 339(1): 115-24, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21775475

ABSTRACT

Ghrelin influences a variety of metabolic functions through a direct action at its receptor, the GhrR (GhrR-1a). Ghrelin knockout (KO) and GhrR KO mice are resistant to the negative effects of high-fat diet (HFD) feeding. We have generated several classes of small-molecule GhrR antagonists and evaluated whether pharmacologic blockade of ghrelin signaling can recapitulate the phenotype of ghrelin/GhrR KO mice. Antagonist treatment blocked ghrelin-induced and spontaneous food intake; however, the effects on spontaneous feeding were absent in GhrR KO mice, suggesting target-specific effects of the antagonists. Oral administration of antagonists to HFD-fed mice improved insulin sensitivity in both glucose tolerance and glycemic clamp tests. The insulin sensitivity observed was characterized by improved glucose disposal with dramatically decreased insulin secretion. It is noteworthy that these results mimic those obtained in similar tests of HFD-fed GhrR KO mice. HFD-fed mice treated for 56 days with antagonist experienced a transient decrease in food intake but a sustained body weight decrease resulting from decreased white adipose, but not lean tissue. They also had improved glucose disposal and a striking reduction in the amount of insulin needed to achieve this. These mice had reduced hepatic steatosis, improved liver function, and no evidence of systemic toxicity relative to controls. Furthermore, GhrR KO mice placed on low- or high-fat diets had lifespans similar to the wild type, emphasizing the long-term safety of ghrelin receptor blockade. We have therefore demonstrated that chronic pharmacologic blockade of the GhrR is an effective and safe strategy for treating metabolic syndrome.


Subject(s)
Insulin Resistance/physiology , Insulin/metabolism , Receptors, Ghrelin/antagonists & inhibitors , Signal Transduction/drug effects , Animals , Anti-Obesity Agents/pharmacology , Blood Glucose/metabolism , Body Weight/drug effects , CHO Cells , Cricetinae , Cricetulus , Dietary Fats/pharmacology , Eating/drug effects , Ghrelin/antagonists & inhibitors , Ghrelin/pharmacology , Glucose Clamp Technique , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Receptors, Ghrelin/physiology , Stress, Physiological/physiology
5.
J Proteomics ; 74(5): 716-27, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21354350

ABSTRACT

The development of type 2 diabetes (T2D) is strongly associated with obesity. In humans, T2D increases the risk for end organ complications. Among these, heart disease has been ranked as the leading cause of death. We used a proteomic methodology to test the hypothesis that a pre-diabetic state generated by high-fat diet leads to changes in proteins related to heart function and structure. Over 300 protein spots were resolved by two-dimensional gel electrophoresis (2-DE). Fifteen protein spots were found to be altered (7 decreased and 8 increased) in pre-diabetic hearts. The protein spots were then identified by mass spectrometry and immunoblots. Among the decreased proteins, 3 are involved in heart structure (one isoform of desmin, troponin T2 and α-cardiac actin), 3 are involved in energy metabolism (mitochondrial ATP synthase ß subunit, adenylate kinase and creatine kinase) and one is a component of the citric acid cycle (isocitrate dehydrogenase 3). In contrast, proteins involved in fatty acid oxidation (two isoforms of peroxisomal enoyl-CoA hydratase) and the citric acid cycle (three isoforms of malate dehydrogenase) were increased in pre-diabetic hearts. The results suggest that changes in the levels of several heart proteins may have implications in the development of the cardiac phenotype associated to T2D.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Diet/adverse effects , Dietary Fats/adverse effects , Muscle Proteins/biosynthesis , Myocardium/metabolism , Proteome/biosynthesis , Proteomics/methods , Animals , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/pathology , Dietary Fats/pharmacology , Electrophoresis, Gel, Two-Dimensional , Humans , Male , Mice , Myocardium/pathology
6.
Endocr Rev ; 32(3): 356-86, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21123740

ABSTRACT

Disruption of the GH receptor (GHR) gene eliminates GH-induced intracellular signaling and, thus, its biological actions. Therefore, the GHR gene disrupted mouse (GHR-/-) has been and is a valuable tool for helping to define various parameters of GH physiology. Since its creation in 1995, this mouse strain has been used by our laboratory and others for numerous studies ranging from growth to aging. Some of the most notable discoveries are their extreme insulin sensitivity in the presence of obesity. Also, the animals have an extended lifespan, which has generated a large number of investigations into the roles of GH and IGF-I in the aging process. This review summarizes the many results derived from the GHR-/- mice. We have attempted to present the findings in the context of current knowledge regarding GH action and, where applicable, to discuss how these mice compare to GH insensitivity syndrome in humans.


Subject(s)
Receptors, Somatotropin/physiology , Age Factors , Animals , Female , Growth Hormone/physiology , Humans , Insulin Resistance , Insulin-Like Growth Factor I/physiology , Male , Mice , Mice, Knockout , Obesity/physiopathology , Phenotype , Receptors, Somatotropin/deficiency , Receptors, Somatotropin/genetics , Signal Transduction
7.
Proc Natl Acad Sci U S A ; 107(34): 15199-204, 2010 Aug 24.
Article in English | MEDLINE | ID: mdl-20699217

ABSTRACT

Androgen-independent recurrence is the major limit of androgen ablation therapy for prostate cancer. Identification of alternative pathways promoting prostate tumor growth is thus needed. Stat5 has been recently shown to promote human prostate cancer cell survival/proliferation and to be associated with early prostate cancer recurrence. Stat5 is the main signaling pathway triggered by prolactin (PRL), a growth factor whose local production is also increased in high-grade prostate cancers. The first aim of this study was to use prostate-specific PRL transgenic mice to address the mechanisms by which local PRL induces prostate tumorogenesis. We report that (i) Stat5 is the major signaling cascade triggered by local PRL in the mouse dorsal prostate, (ii) this model recapitulates prostate tumorogenesis from precancer lesions to invasive carcinoma, and (iii) tumorogenesis involves dramatic accumulation and abnormal spreading of p63-positive basal cells, and of stem cell antigen-1-positive cells identified as a stem/progenitor-like subpopulation. Because basal epithelial stem cells are proposed to serve as tumor-initiating cells, we challenged the relevance of local PRL as a previously unexplored therapeutic target. Using a double-transgenic approach, we show that Delta1-9-G129R-hPRL, a competitive PRL-receptor antagonist, prevented early stages of prostate tumorogenesis by reducing or inhibiting Stat5 activation, cell proliferation, abnormal basal-cell pattern, and frequency or grade of intraepithelial neoplasia. This study identifies PRL receptor/Stat5 as a unique pathway, initiating prostate tumorogenesis by altering basal-/stem-like cell subpopulations, and strongly supports the importance of further developing strategies to target locally overexpressed PRL in human prostate cancer.


Subject(s)
Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prolactin/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Base Sequence , Cell Proliferation , DNA Primers/genetics , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Neoplasms, Hormone-Dependent/genetics , Neoplasms, Hormone-Dependent/therapy , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Prolactin/genetics , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Rats , Receptors, Prolactin/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Tissue Distribution
8.
Growth Horm IGF Res ; 20(1): 73-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19747867

ABSTRACT

The goal of this study was to examine factors that contribute to energy balance in female GHR -/- mice. We measured energy intake, energy expenditure (EE), fuel utilization, body mass (M(b)) changes and physical activity in 17month-old female GHR -/- mice and their age-matched wild type littermates. The GHR -/- mice were smaller, consumed more food per unit M(b), had greater EE per unit M(b) and had an increase in 24-h EE/M(b) that was similar to the increase in their surface-area-to-volume ratio. Locomotor activity (LMA) was reduced in the GHR -/- mice, but the energetic cost associated with their LMA was greater than in wild type controls. Furthermore, M(b) and LMA were independent explanatory covariates of most of the variance in EE, and when adjusted for M(b) and LMA, the GHR -/- mice had higher EE during both the light and dark phases of the daily cycle. Respiratory quotient was lower in GHR -/- mice during the light phase, which indicated a greater utilization of lipid relative to carbohydrate in these mice. Additionally, GHR -/- mice had higher ratios of caloric intake to EE at several intervals during the dark phase, and this effect was greater and more sustained in the final 3h of the dark phase. Therefore, we conclude that GHR -/- mice are able to overcome the substantial energetic challenges of dwarfism through several mechanisms that promote stable M(b). Relative to wild type mice, the GHR -/- mice consumed more calories per unit M(b), which offset the disproportionate increase in their daily energy expenditure. While GHR -/- mice oxidized a greater proportion of lipid during the light phase in order to meet their energy requirements, they achieved greater energy efficiency and storage during the dark phase through a combination of higher energy consumption and lower LMA.


Subject(s)
Energy Metabolism/physiology , Receptors, Somatotropin/metabolism , Activity Cycles/physiology , Animals , Dwarfism/metabolism , Energy Intake/physiology , Female , Lipid Metabolism/physiology , Mice , Mice, Mutant Strains , Motor Activity/physiology , Receptors, Somatotropin/genetics , Receptors, Somatotropin/physiology
9.
Growth Horm IGF Res ; 17(4): 346-51, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17544797

ABSTRACT

Growth hormone's (GH) lipolytic activity in white adipose tissue (WAT) results in decreased body fat in giant GH transgenic mice and increased subcutaneous fat in dwarf growth hormone receptor/binding protein gene-disrupted mice (GHR -/-). We therefore hypothesized that GH action would affect expression of CIDE-A (cell-death-inducing DFF45-like effector-A), a protein found in white adipose tissue (WAT) and involved in lipid metabolism. CIDE-A RNA levels were determined in subcutaneous, retroperitoneal and epididymal adipose tissue isolated from wild-type and GHR -/- mice. The adipose tissue was also analyzed for adipocyte size. We determined that the lack of GH action has depot-specific effects on the levels of CIDE-A RNA and affected adipocyte cell size. CIDE-A expression is significantly reduced in GHR -/- subcutaneous fat compared to wild-type but is not altered in retroperitoneal or epididymal fat. Likewise, adipocytes are significantly enlarged in GHR -/- subcutaneous adipose tissue relative wild-type mice. A high-fat diet also influenced the level of CIDE-A RNA in mouse adipose tissue. The high-fat diet significantly reduced CIDE-A expression in wild-type subcutaneous fat but did not alter CIDE-A expression in subcutaneous fat of GHR -/- mice. The diet also reduced CIDE-A expression in wild-type retroperitoneal fat but the levels of CIDE-A in epididymal fat were unchanged. In contrast, the high-fat diet reduced CIDE-A expression in both retroperitoneal and epididymal fat of GHR -/- mice. These data demonstrate that CIDE-A levels are reduced in two different mouse models of obesity and this reduction may contribute to altered lipid metabolism.


Subject(s)
Adipose Tissue, White/metabolism , Apoptosis Regulatory Proteins/genetics , Diet, Atherogenic , Dietary Fats/pharmacology , Receptors, Somatotropin/genetics , Adipocytes, White/cytology , Adipocytes, White/drug effects , Adipose Tissue, White/drug effects , Animals , Apoptosis Regulatory Proteins/metabolism , Blood Glucose/analysis , Body Weight/drug effects , Cell Size/drug effects , Down-Regulation/drug effects , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Comp Hepatol ; 6: 4, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17472743

ABSTRACT

BACKGROUND: Increased levels of circulating fatty acids caused by insulin resistance and increased adipocyte lipolysis can accumulate within the liver resulting in steatosis. This steatosis sensitizes the liver to inflammation and further injury which can lead to liver dysfunction. We performed microarray analysis on normal mouse liver tissue at different ages and type 2 diabetic liver exhibiting steatosis to identify differentially expressed genes involved in lipid accumulation and liver dysfunction. RESULTS: Microarray analysis identified CIDE-A as the most differentially expressed gene as a function of age. Mice fed a high fat diet developed hyperinsulinemia, hyperglycemia and liver steatosis, all features of the human metabolic syndrome. Increased CIDE-A expression was observed in type 2 diabetic liver and the elevated CIDE-A expression could be reversed by weight loss and normalization of plasma insulin. Also, CIDE-A expression was found to be correlated with hepatic lipid accumulation. CONCLUSION: The corresponding increase in CIDE-A expression with hyperinsulinemia and liver steatosis suggests a novel pathway for lipid accumulation in the liver.

11.
Proteomics ; 7(7): 1140-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17390296

ABSTRACT

In this study, proteomic analysis was performed on the skin of C57BL/6J mice with type 2 diabetes and compared to nondiabetic controls. To induce obesity and subsequent diabetes, mice were placed on a high-fat diet for 16 wk. After 16 wk, both diabetic and nondiabetic control mice were sacrificed and their skin removed for analysis. Following 2-DE, proteomic profiles from the skin samples were quantified using PDQuest software. Out of more than 1000 distinct protein spots, 28 were shown to be significantly altered with 6 being decreased and 22 increased in the diabetic state compared to controls. The 28 protein spots were removed from the gels and analyzed by MALDI-TOF and MS/MS analyses. Protein identifications revealed that 17 of the 28 proteins were involved in energy metabolism (60.7% of changes observed). Collectively, none of the significantly altered proteins had been shown previously to be altered in diabetic skin. This study not only helps to identify proteins found in skin samples of obese mice with type 2 diabetes, but also shows that skin biopsies coupled with proteomic analysis may be useful as a noninvasive method for the diagnosis of hyperinsulinemia and diabetes.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Proteins/analysis , Proteomics , Skin/chemistry , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Dietary Fats/administration & dosage , Energy Metabolism , Mice , Mice, Inbred C57BL , Skin/pathology , Weight Gain
12.
J Endocrinol ; 190(2): 271-85, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16899561

ABSTRACT

Experimental, clinical, and epidemiological data support the growth-promoting role of endocrine prolactin (PRL) in mammary tumors. PRL is also produced by the breast, where it is now recognized to act as a growth/survival factor via autocrine/paracrine mechanisms. Recent transgenic (Tg) mouse models have revealed the pro-oncogenic effect of PRL over-expression in virgin mammary glands. To address the question whether PRL tumorigenicity was maintained on differentiated mammary glands, we generated mammary-specific Tg mice expressing human (h)PRL under the control of the milk whey acidic protein promoter, which directs autocrine hPRL over-expression in late gestation throughout lactation. Minimal levels of transgene expression were detected in the mammary glands of virgin animals, which at best induced partial ductal branching and lobulo-alveolar structures in older nulliparous females. As expected, expression of mammary hPRL dramatically increased at the end of first pregnancy, and from this point it never returned to baseline, although it peaked at each gestation/lactation cycle. Over-expression of hPRL that starts when the gland is already well into the differentiation process led to various morphological mammary alterations, including abnormally differentiated epithelium, atropy of the myoepithelial layer, dilated ducts, cysts, and lymphocytic infiltrates. These phenotypes tended to worsen with successive pregnancies, also reflecting cumulative damage of failure of involution. Although some older, multiparous females developed benign tumors (papillomas and metaplasias), none of the animals studied developed mammary carcinomas. In addition, we noticed that half of the Tg females exhibited lactation defects, leading to significantly increased pup mortality. This phenotype was due neither to failure of milk production nor to modification of its protein content, but rather it was correlated to lipid enrichment of the milk, which, in combination with profoundly altered morphology of the gland, led to impaired milk extrusion through the nipple. In summary, these data show that over-expression of autocrine hPRL in a differentiating mammary gland induces dramatic functional and morphological defects, but not carcinoma. This deserves further investigations on the emerging concept that autocrine PRL may have different effects on pathological development of the mammary gland depending on the differentiation state of the latter.


Subject(s)
Autocrine Communication , Lactation , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Animal/metabolism , Prolactin/metabolism , Animals , Cell Proliferation , Female , Gene Expression , Humans , Immunohistochemistry/methods , Lipid Metabolism , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/pathology , Metaplasia/pathology , Mice , Mice, Inbred Strains , Mice, Transgenic , Milk/metabolism , Milk Proteins/genetics , Papilloma/pathology , Pregnancy , Prolactin/analysis , Prolactin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic
13.
OMICS ; 9(3): 251-65, 2005.
Article in English | MEDLINE | ID: mdl-16209639

ABSTRACT

Combining results from gene microarrays, clinical chemistry, and quantitative tissue histomorphology in an integrated bioinformatics setting enables prioritization of gene families as well as individual genes in a type II diabetes animal study. This new methodology takes advantage of a time-controlled mouse study as the animals progress from a normal phenotype to that of type II diabetes. Profiles from different levels of the biological hierarchy of unpooled entities provide an encompassing, system-wide view of biological changes. Here, phenotypic changes on the tissue-structural and physiological level are used as statistical covariants to enrich the gene expression analysis, suggesting correlative processes between gene expression and phenotype unlocked by multi-sample comparisons. We apply correlative and gene set enrichment procedures and compare the results to differential analysis to identify molecular markers. Evaluation based on ontological classifications proves changes in prioritization of disease-related genes that would have been overlooked by conventional gene expression analyses strategies.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Microarray Analysis , Phenotype , Animals , Blood Glucose/analysis , Chemistry, Clinical , Computational Biology , Enzyme-Linked Immunosorbent Assay , Fasting , Gene Expression Profiling , Genetic Markers , Insulin/blood , Insulin Resistance/genetics , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Reference Standards
14.
Lipids ; 37(8): 733-40, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12371743

ABSTRACT

In mammalian cells, Sprecher has proposed that the synthesis of long-chain PUFA from the 20-carbon substrates involves two consecutive elongation steps, a delta6-desaturation step followed by retroconversion (Sprecher, H., Biochim. Biophys. Acta 1486, 219-231, 2000). We searched the database using the translated sequence of human elongase ELOVL5, whose encoded enzyme elongates monounsaturated and polyunsaturated FA, as a query to identify the enzyme(s) involved in elongation of very long chain PUFA. The database search led to the isolation of two cDNA clones from human and mouse. These clones displayed deduced amino acid sequences that had 56.4 and 58% identity, respectively, to that of ELOVL5. The open reading frame of the human clone (ELOVL2) encodes a 296-amino acid peptide, whereas the mouse clone (Elovl2) encodes a 292-amino acid peptide. Expression of these open reading frames in baker's yeast, Saccharomyces cerevisiae, demonstrated that the encoded proteins were involved in the elongation of both 20- and 22-carbon long-chain PUFA, as determined by the conversion of 20:4n-6 to 22:4n-6, 22:4n-6 to 24:4n-6, 20:5n-3 to 22:5n-3, and 22:5n-3 to 24:5n-3. The elongation activity of the mouse Elovl2 was further demonstrated in the transformed mouse L cells incubated with long-chain (C20- and C22-carbon) n-6 and n-3 PUFA substrates by the significant increase in the levels of 24:4n-6 and 24:5n-3, respectively. This report demonstrates the isolation and identification of two mammalian genes that encode very long chain PUFA specific elongation enzymes in the Sprecher pathway for DHA synthesis.


Subject(s)
Acetyltransferases/genetics , Acetyltransferases/metabolism , Fatty Acids, Omega-3/biosynthesis , Fatty Acids, Unsaturated/biosynthesis , Amino Acid Sequence , Animals , Blotting, Northern , Cloning, Molecular , DNA, Complementary/genetics , Fatty Acid Desaturases/metabolism , Fatty Acid Elongases , Fatty Acids, Omega-6 , Gas Chromatography-Mass Spectrometry , Genetic Vectors/genetics , Humans , Mice , Molecular Sequence Data , Open Reading Frames/genetics , Organ Specificity , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...