Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Endocrinology ; 150(3): 1259-68, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18948405

ABSTRACT

Androgens are important regulators of body composition and promote myogenic differentiation and inhibit adipogenesis of mesenchymal, multipotent cells. Here, we investigated the mechanisms by which androgens induce myogenic differentiation of mesenchymal multipotent cells. Incubation of mesenchymal multipotent C3H 10T1/2 cells with testosterone and dihydrotestosterone promoted nuclear translocation of androgen receptor (AR)/beta-catenin complex and physical interaction of AR, beta-catenin, and T-cell factor-4 (TCF-4). Inhibition of beta-catenin by small inhibitory RNAs significantly decreased testosterone-induced stimulation of myogenic differentiation. Overexpression of TCF-4, a molecule downstream of beta-catenin in Wnt signaling cascade, in C3H 10T1/2 cells significantly up-regulated expression of myoD and myosin heavy chain II proteins and of follistatin (Fst), which binds and antagonizes native ligands of the TGF-beta/Smad pathway. Gene array analysis of C3H 10T1/2 cells treated with testosterone revealed that testosterone up-regulated the expression of Fst and modified the expression of several signaling molecules involved in the TGF-beta/Smad pathway, including Smad7. Lowering of testosterone levels in mice by orchidectomy led to a significant decrease in Fst and Smad7 expression; conversely, testosterone supplementation in castrated mice up-regulated Fst and Smad7 mRNA expression in androgen-responsive levator ani muscle. Testosterone-induced up-regulation of MyoD and myosin heavy chain II proteins in C3H 10T1/2 cells was abolished in cells simultaneously treated with anti-Fst antibody, suggesting an essential role of Fst during testosterone regulation of myogenic differentiation. In conclusion, our data suggest the involvement of AR, beta-catenin, and TCF-4 pathway during androgen action to activate a number of Wnt target genes, including Fst, and cross communication with the Smad signaling pathway.


Subject(s)
Androgens/pharmacology , Follistatin/physiology , Muscle Development/drug effects , Receptors, Androgen/physiology , Transforming Growth Factor beta/physiology , beta Catenin/physiology , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Differentiation/drug effects , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Follistatin/metabolism , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/physiology , Orchiectomy , Protein Transport/drug effects , RNA, Small Interfering/pharmacology , Receptor Cross-Talk/physiology , Receptors, Androgen/metabolism , Signal Transduction/drug effects , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , TCF Transcription Factors/physiology , Transcription Factor 4 , Transforming Growth Factor beta/metabolism , beta Catenin/antagonists & inhibitors , beta Catenin/metabolism
2.
Metabolism ; 55(9): 1239-47, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16919545

ABSTRACT

Excess glucocorticoids (GCs) cause muscle atrophy. Glucocorticoid-induced muscle atrophy is associated with increased intramuscular myostatin expression. Myostatin is a negative regulator of skeletal muscle mass. Glutamine prevents GC-induced muscle atrophy. We hypothesized that glutamine effect on reversal of GC-induced muscle atrophy is mediated in part by suppression of myostatin. We administered daily to male Sprague-Dawley rats dexamethasone, dexamethasone plus glutamine, saline or saline plus glutamine, all pair-fed. Animals were killed on day 5. Body weight and weights of gastrocnemius muscles were measured. Myostatin expression was measured by Northern and Western blots, and was compared with glyceraldehyde-3-phosphate dehydrogenase. Myoblast C2C12 cells were exposed to dexamethasone, or dexamethasone and glutamine, and their myostatin messenger RNA and protein expression compared with glyceraldehyde-3-phosphate dehydrogenase. Myostatin promoter activity was measured by luciferase activity of transfected C2C12 cells, grown in medium including dexamethasone, or dexamethasone plus glutamine. Rats that received dexamethasone showed significant body and muscle weight loss accompanied by an increase in intramuscular myostatin expression, compared with their saline-treated controls. Pair-fed rats given dexamethasone plus glutamine had significantly less reduction in body and muscle weights and lower myostatin expression when compared with those treated with dexamethasone alone. In C2C12 myoblast cells, addition of glutamine to dexamethasone prevented the hyperexpression of myostatin induced by dexamethasone. Myostatin promoter activity increased in cells exposed to dexamethasone, but this increase was partially blocked by addition of the glutamine. Administration of glutamine partially prevents GC-induced myostatin expression and muscle atrophy, providing a potential mechanism for the prevention of muscle atrophy induced by glucocorticoids.


Subject(s)
Glucocorticoids/pharmacology , Glutamine/pharmacology , Muscular Atrophy/drug therapy , Transforming Growth Factor beta/genetics , Animals , Body Weight , Cell Line , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Drug Antagonism , Glucocorticoids/administration & dosage , Glutamine/administration & dosage , Male , Muscle, Skeletal/pathology , Myostatin , Organ Size , Promoter Regions, Genetic , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley
3.
Endocrinology ; 147(1): 141-54, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16210377

ABSTRACT

Testosterone supplementation in men decreases fat mass; however, the mechanisms by which it inhibits fat mass are unknown. We hypothesized that testosterone inhibits adipogenic differentiation of preadipocytes by activation of androgen receptor (AR)/beta-catenin interaction and subsequent translocation of this complex to the nucleus thereby bypassing canonical Wnt signaling. We tested this hypothesis in 3T3-L1 cells that differentiate to form fat cells in adipogenic medium. We found that these cells express AR and that testosterone and dihydrotestosterone dose-dependently inhibited adipogenic differentiation as analyzed by Oil Red O staining and down-regulation of CCAAT/enhancer binding protein-alpha and -delta and peroxisome proliferator-activated receptor-gamma2 protein and mRNA. These inhibitory effects of androgens were partially blocked by flutamide or bicalutamide. Androgen treatment was associated with nuclear translocation of beta-catenin and AR. Immunoprecipitation studies demonstrated association of beta-catenin with AR and T-cell factor 4 (TCF4) in the presence of androgens. Transfection of TCF4 cDNA inhibited adipogenic differentiation, whereas a dominant negative TCF4 cDNA construct induced adipogenesis and blocked testosterone's inhibitory effects. Our gene array analysis indicates that testosterone treatment led to activation of some Wnt target genes. Expression of constitutively activated AR fused with VP-16 did not inhibit the expression of CCAAT/enhancer binding protein-alpha in the absence of androgens. Testosterone and dihydrotestosterone inhibit adipocyte differentiation in vitro through an AR-mediated nuclear translocation of beta-catenin and activation of downstream Wnt signaling. These data provide evidence for a regulatory role for androgens in inhibiting adipogenic differentiation and a mechanistic explanation consistent with the observed reduction in fat mass in men treated with androgens.


Subject(s)
Adipocytes/cytology , Adipose Tissue/cytology , Cell Nucleus/metabolism , Receptors, Androgen/metabolism , TCF Transcription Factors/metabolism , Testosterone/pharmacology , Transcription Factors/genetics , Wnt Proteins/physiology , beta Catenin/metabolism , 3T3 Cells , Adipocytes/drug effects , Animals , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Mice , Protein Transport/drug effects , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology , Transcription Factor 7-Like 2 Protein , Transcription, Genetic/drug effects , Wnt Proteins/genetics
4.
Kidney Int ; 68(1): 352-61, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15954927

ABSTRACT

BACKGROUND: Maintenance hemodialysis patients often display evidence for protein-energy malnutrition, inflammation, and sarcopenia. We therefore investigated whether sedentary maintenance hemodialysis patients have decreased skeletal muscle mRNA levels and muscle and serum protein concentrations of certain growth factors. METHODS: Fifty-one clinically stable maintenance hemodialysis patients (32 men and 19 women), and 21 normal adults (16 men and five women) of similar age, gender mix, racial/ethnic backgrounds, serum albumin, body composition, and level of sedentary activity were studied. Individuals underwent biopsy of the right vastus lateralis muscle, and real-time polymerase chain reaction (PCR) amplification of mRNAs for insulin-like growth factor-I (IGF-I), IGF-II, IGF-I receptor (IGF-IR), IGF-IIR, and myostatin (44 patients) was performed. Serum and muscle IGF-I and IGF-II, serum proinflammatory cytokines, and leg muscle strength, power, and fatigability were measured. RESULTS: Maintenance hemodialysis patients displayed significantly reduced mRNA levels for IGF-IEa mRNA (P < 0.05), IGF-II (P < 0.001), and IGF-IR (P < 0.001), and no difference in mRNAs for IGF-IEc, IGF-IIR, or myostatin as compared to normal controls. Muscle mRNA levels, in general, followed the same pattern in male and female maintenance hemodialysis patients considered separately. In the maintenance hemodialysis patients, muscle IGF-I protein, serum IGF-II and tumor necrosis factor-alpha (TNF-alpha) were each increased, whereas serum C-reactive protein (CRP) and interleukin-6 (IL-6) were normal. Muscle strength and power, but not fatigability, were reduced in the maintenance hemodialysis patients. CONCLUSION: In sedentary, clinically stable maintenance hemodialysis patients as compared to sedentary normal individuals, the mRNA levels for IGF-IEa, IGF-II, and the IGF-I receptor are decreased in vastus lateralis muscle. Protein levels for muscle IGF-I and serum IGF-II are increased.


Subject(s)
Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor I/genetics , Kidney Failure, Chronic/physiopathology , Muscle, Skeletal/physiology , Receptor, IGF Type 1/genetics , Adult , Female , Humans , Insulin-Like Growth Factor I/metabolism , Insulin-Like Growth Factor II/metabolism , Kidney Failure, Chronic/therapy , Life Style , Male , Middle Aged , Motor Activity , Muscle, Skeletal/pathology , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , Myostatin , RNA, Messenger/analysis , Receptor, IGF Type 2/genetics , Renal Dialysis , Transforming Growth Factor beta/genetics
5.
J Clin Endocrinol Metab ; 90(2): 855-63, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15522925

ABSTRACT

Previous studies of Delta 4-androstene-3,17-dione (4-androstenedione) administration in men have not demonstrated sustained increments in testosterone levels, fat-free mass (FFM), and muscle strength, and failure to demonstrate androstenedione's androgenic/anabolic effects has stifled efforts to regulate its sales. To determine whether 4-androstenedione has androgenic/anabolic properties, we evaluated its association with androgen receptor (AR) and its effects on myogenesis in vitro. Additionally, we studied the effects of a high dose of 4-androstenedione on testosterone levels, FFM, and muscle strength in hypogonadal men. We determined the dissociation constant (K(d)) for 4-androstenedione using fluorescence anisotropy measurement of competitive displacement of fluorescent androgen from AR ligand-binding domain. AR nuclear translocation and myogenic activity of androstenedione were evaluated in mesenchymal, pluripotent C3H10T1/2 cells, in which androgens stimulate myogenesis through an AR pathway. We determined effects of a high dose of androstenedione (500 mg thrice daily) given for 12 wk on FFM, muscle strength, and hormone levels in nine healthy, hypogonadal men. 4-Androstenedione competitively displaced fluorescent androgen from AR ligand-binding domain with a lower affinity than dihydrotestosterone (K(d), 648 +/- 21 and 10 +/- 0.4 nm, respectively). In C3H10T1/2 cells, 4-androstenedione caused nuclear translocation of AR and stimulated myogenesis, as indicated by a dose-dependent increase in myosin heavy chain II+ myotube area and up-regulation of MyoD protein. Stimulatory effects of 4-androstenedione on myosin heavy chain II+ myotubes and myogenic determination factor expression were attenuated by bicalutamide, an AR antagonist. Administration of 1500 mg 4-androstenedione daily to hypogonadal men significantly increased serum androstenedione, total and free testosterone, estradiol, and estrone levels and suppressed SHBG and high-density lipoprotein cholesterol levels. 4-androstenedione administration was associated with significant gains in FFM (+1.7 +/- 0.5 kg; P = 0.012) and muscle strength in bench press (+4.3 +/- 3.1 kg; P = 0.006) and leg press exercises (+18.8 +/- 17.3 kg; P = 0.045). 4-androstenedione is an androgen that binds AR, induces AR nuclear translocation, and promotes myogenesis in vitro, with substantially lower potency than dihydrotestosterone. 4-androstenedione administration in high doses to hypogonadal men increases testosterone levels, FFM, and muscle strength, although at the dose tested, the anabolic effects in hypogonadal men are likely because of its conversion to testosterone.


Subject(s)
Androstenedione/therapeutic use , Hypogonadism/physiopathology , Muscle, Skeletal/physiopathology , Receptors, Androgen/metabolism , Testosterone/blood , 3T3 Cells , Adult , Androstenedione/pharmacology , Animals , Cell Differentiation/drug effects , Humans , Hypogonadism/classification , Hypogonadism/drug therapy , Kinetics , Ligands , Male , Mice , Middle Aged , Muscle, Skeletal/drug effects , Reference Values
6.
J Clin Endocrinol Metab ; 89(10): 5245-55, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15472231

ABSTRACT

Androgens stimulate myogenesis, but we do not know what cell types within human skeletal muscle express the androgen receptor (AR) protein and are the target of androgen action. Because testosterone promotes the commitment of pluripotent, mesenchymal cells into myogenic lineage, we hypothesized that AR would be expressed in mesenchymal precursor cells in the skeletal muscle. AR expression was evaluated by immunohistochemical staining, confocal immunofluorescence, and immunoelectron microscopy in sections of vastus lateralis from healthy men before and after treatment with a supraphysiological dose of testosterone enanthate. Satellite cell cultures from human skeletal muscle were also tested for AR expression. AR protein was expressed predominantly in satellite cells, identified by their location outside sarcolemma and inside basal lamina, and by CD34 and C-met staining. Many myonuclei in muscle fibers also demonstrated AR immunostaining. Additionally, CD34+ stem cells in the interstitium, fibroblasts, and mast cells expressed AR immunoreactivity. AR expression was also observed in vascular endothelial and smooth muscle cells. Immunoelectron microscopy revealed aggregation of immunogold particles in nucleoli of satellite cells and myonuclei; testosterone treatment increased nucleolar AR density. In enriched cultures of human satellite cells, more than 95% of cells stained for CD34 and C-met, confirming their identity as satellite cells, and expressed AR protein. AR mRNA and protein expression in satellite cell cultures was confirmed by RT-PCR, reverse transcription and real-time PCR, sequencing of RT-PCR product, and Western blot analysis. Incubation of satellite cell cultures with supraphysiological testosterone and dihydrotestosterone concentrations (100 nm testosterone and 30 nm dihydrotestosterone) modestly increased AR protein levels. We conclude that AR is expressed in several cell types in human skeletal muscle, including satellite cells, fibroblasts, CD34+ precursor cells, vascular endothelial, smooth muscle cells, and mast cells. Satellite cells are the predominant site of AR expression. These observations support the hypothesis that androgens increase muscle mass in part by acting on several cell types to regulate the differentiation of mesenchymal precursor cells in the skeletal muscle.


Subject(s)
Muscle, Skeletal/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Satellite Cells, Skeletal Muscle/metabolism , Testosterone/analogs & derivatives , Testosterone/administration & dosage , Adult , Blotting, Western , Cells, Cultured , Humans , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Immunoelectron , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Satellite Cells, Skeletal Muscle/ultrastructure , Up-Regulation
7.
Oncogene ; 23(52): 8557-62, 2004 Nov 04.
Article in English | MEDLINE | ID: mdl-15378028

ABSTRACT

KIT ligand (KL) and its receptor, c-kit, are coexpressed in many types of cancer cells and have been implicated in tumor growth and angiogenesis. While Sertoli cell-specific regulation of the KL promoter has been well characterized, regulation in cancer cells remains to be elucidated. We recently reported microarray results demonstrating that increased high-mobility group (HMG) A1a protein expression correlates with increased KL transcription in MCF-7 human breast cancer cells. Sequence analysis indicates a potential for multiple HMGA1 binding sites within the human KL promoter. In order to better define the underlying molecular mechanisms that HMGA1 uses to facilitate malignant transformation of cancer cells, we have used a variety of methods to determine whether HMGA1a directly regulates the human KL promoter in breast and ovarian cancer cells. Our results indicate that: (i) KL promoter activity is significantly higher in MCF-7 cells overexpressing HMGA1a; (ii) HMGA1a protein binds to AT-rich regions of the KL promoter DNA both in vitro and in vivo; (iii) mutation of the AT-rich regions inhibits HMGA1a binding in vitro; and (iv) HMGA1a-specific inhibition significantly decreases transcription of KL in OCC1 human ovarian cancer cells. In addition, MCF-7 cells with transgenic HMGA1 overexpression stained positive for the KL protein by immunocytochemistry and immunohistochemistry, and were growth-inhibited by KL neutralization. The cumulative evidence indicates that HMGA1 positively regulates the human KL promoter in breast and ovarian cancer cells and implicates serum KL as a diagnostic marker for HMGA1-positive carcinomas.


Subject(s)
Breast Neoplasms/genetics , HMGA1a Protein/metabolism , Ovarian Neoplasms/genetics , Promoter Regions, Genetic , Stem Cell Factor/genetics , Animals , Base Sequence , Breast Neoplasms/metabolism , Female , Humans , Mice , Molecular Sequence Data , Ovarian Neoplasms/metabolism , Stem Cell Factor/metabolism
8.
J Gerontol A Biol Sci Med Sci ; 58(12): M1103-10, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14684707

ABSTRACT

Testosterone supplementation increases muscle mass primarily by inducing muscle fiber hypertrophy; however, the mechanisms by which testosterone exerts its anabolic effects on the muscle are poorly understood. The prevalent view is that testosterone improves net muscle protein balance by stimulating muscle protein synthesis, decreasing muscle protein degradation, and improving the reutilization of amino acids. However, the muscle protein synthesis hypothesis does not adequately explain testosterone-induced changes in fat mass, myonuclear number, and satellite cell number. We postulate that testosterone promotes the commitment of pluripotent stem cells into the myogenic lineage and inhibits their differentiation into the adipogenic lineage. The hypothesis that the primary site of androgen action is the pluripotent stem cell provides a unifying explanation for the observed reciprocal effects of testosterone on muscle and fat mass.


Subject(s)
Body Composition/drug effects , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Pluripotent Stem Cells/metabolism , Testosterone/administration & dosage , Testosterone/physiology , Adolescent , Adult , Biopsy, Needle , Body Composition/physiology , Child , Child, Preschool , Humans , Immunohistochemistry , Male , Muscle Development/drug effects , Muscle Development/physiology , Muscle Fibers, Skeletal/drug effects , Muscle Proteins/drug effects , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Sensitivity and Specificity , Stem Cells/drug effects , Stem Cells/metabolism
9.
Endocrinology ; 144(11): 5081-8, 2003 Nov.
Article in English | MEDLINE | ID: mdl-12960001

ABSTRACT

Testosterone supplementation increases skeletal muscle mass and decreases fat mass; however, the underlying mechanisms are unknown. We hypothesized that testosterone regulates body composition by promoting the commitment of mesenchymal pluripotent cells into myogenic lineage and inhibiting their differentiation into adipogenic lineage. Mouse C3H 10T1/2 pluripotent cells were treated with testosterone (0-300 nM) or dihydrotestosterone (DHT, 0-30 nM) for 0-14 d, and myogenic conversion was evaluated by immunocytochemical staining for early (MyoD) and late (myosin heavy chain II; MHC) myogenic markers and by measurements of MyoD and MHC mRNA and protein. Adipogenic differentiation was assessed by adipocyte counting and by measurements of peroxisomal proliferator-activated receptor gamma 2 (PPAR gamma 2) mRNA and PPAR gamma 2 protein and CCAAT/enhancer binding protein alpha. The number of MyoD+ myogenic cells and MHC+ myotubes and MyoD and MHC mRNA and protein levels increased dose dependently in response to testosterone and DHT treatment. Both testosterone and DHT decreased the number of adipocytes and down-regulated the expression of PPAR gamma 2 mRNA and PPAR gamma 2 protein and CCAAT/enhancer binding protein alpha. Androgen receptor mRNA and protein levels were low at baseline but increased after testosterone or DHT treatment. The effects of testosterone and DHT on myogenesis and adipogenesis were blocked by bicalutamide. Therefore, testosterone and DHT regulate lineage determination in mesenchymal pluripotent cells by promoting their commitment to the myogenic lineage and inhibiting their differentiation into the adipogenic lineage through an androgen receptor-mediated pathway. The observation that differentiation of pluripotent cells is androgen dependent provides a unifying explanation for the reciprocal effects of androgens on muscle and fat mass in men.


Subject(s)
Adipocytes/cytology , Androgens/pharmacology , Dihydrotestosterone/pharmacology , Muscle, Skeletal/cytology , Pluripotent Stem Cells/cytology , Receptors, Androgen/physiology , Testosterone/pharmacology , Androgen Antagonists/pharmacology , Anilides/pharmacology , Animals , Cell Differentiation/drug effects , Cell Division/drug effects , Cell Line , Dihydrotestosterone/antagonists & inhibitors , Mice , Mice, Inbred C3H , Nitriles , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Receptors, Androgen/metabolism , Testosterone/antagonists & inhibitors , Tosyl Compounds
10.
J Gravit Physiol ; 9(2): 61-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-14638460

ABSTRACT

To clarify the mechanism of skeletal muscle wasting during spaceflights, we investigated whether intramuscular gene expression profiles are affected, by using DNA microarray methods. Male rats sent on the 17-day NASA STS-90 Neurolab spaceflight were sacrificed 24 hours after return to earth (MG group). Ground control rats were maintained for 17 days in flight-simulated cages (CS group). Spaceflight induced a 19% and 23% loss of tibialis anterior and gastrocnemius muscle mass, respectively, as compared to ground controls. Muscle RNA was analyzed by the Clontech Atlas DNA expression array in four rats, with two MG/ CS pairs for the tibialis anterior, and one pair for the gastrocnemius. Alterations in gene expression were verified for selected genes by reverse-transcription PCR. In both muscles of MG rats, mRNAs for 12 genes were up-regulated by over 2-fold, and 38 were down-regulated compared to controls. There was inhibition of genes for cell proliferation and growth factor cascades, including cell cycle genes and signal transduction proteins, such as p21 Cip1, retinoblastoma (Rb), cyclins G1/S, -E and -D3, MAP kinase 3, MAD3, and ras related protein RAB2. These data indicate that following exposure to microgravity, there is downregulation of genes involved in regulation of muscle satellite cell replication.


Subject(s)
Gene Expression/physiology , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Space Flight , Weightlessness , Animals , Male , Muscle Proteins/metabolism , Muscle, Skeletal/physiology , Muscle, Skeletal/physiopathology , Muscular Atrophy/metabolism , Muscular Atrophy/physiopathology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...