Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
BMC Med Genet ; 21(1): 215, 2020 10 31.
Article in English | MEDLINE | ID: mdl-33129256

ABSTRACT

BACKGROUND: SHORT syndrome is a rare genetic disease named with the acronyms of short stature, hyper-extensibility of joints, ocular depression, Rieger anomaly and teething delay. It is inherited in an autosomal dominant manner confirmed by the identification of heterozygous mutations in PIK3R1. This study hereby presents a 15-year-old female with intrauterine growth restriction, short stature, teething delay, characteristic facial gestalts who was identified a novel de novo nonsense mutation in PIK3R1. CASE PRESENTATION: The proband was admitted to our department due to irregular menstrual cycle and hirsutism with short stature, who had a history of intrauterine growth restriction and presented with short stature, teething delay, characteristic facial gestalts, hirsutism, and thyroid disease. Whole-exome sequencing and Sanger sequencing revealed c.1960C > T, a novel de novo nonsense mutation, leading to the termination of protein translation (p. Gln654*). CONCLUSIONS: This is the first case report of SHORT syndrome complicated with thyroid disease in China, identifying a novel de novo heterozygous nonsense mutation in PIK3R1 gene (p. Gln654*). The phenotypes are mildly different from other cases previously described in the literature, in which our patient presents with lipoatrophy, facial feature, and first reported thyroid disease. Thyroid disease may be a new clinical symptom of patients with SHORT syndrome.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Codon, Nonsense , Growth Disorders/genetics , Hypercalcemia/genetics , Metabolic Diseases/genetics , Nephrocalcinosis/genetics , Thyroid Diseases/genetics , Adolescent , Asian People , Base Sequence , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Female , Gene Expression , Genes, Dominant , Growth Disorders/complications , Growth Disorders/ethnology , Growth Disorders/pathology , Heterozygote , Humans , Hypercalcemia/complications , Hypercalcemia/ethnology , Hypercalcemia/pathology , Metabolic Diseases/complications , Metabolic Diseases/ethnology , Metabolic Diseases/pathology , Models, Molecular , Nephrocalcinosis/complications , Nephrocalcinosis/ethnology , Nephrocalcinosis/pathology , Phenotype , Protein Structure, Secondary , Thyroid Diseases/complications , Thyroid Diseases/ethnology , Thyroid Diseases/pathology , Exome Sequencing
2.
Mol Metab ; 31: 1-13, 2020 01.
Article in English | MEDLINE | ID: mdl-31918912

ABSTRACT

OBJECTIVE: An increase in mass and/or brown adipose tissue (BAT) functionality leads to an increase in energy expenditure, which may be beneficial for the prevention and treatment of obesity. Moreover, distinct class I PI3K isoforms can participate in metabolic control as well as in systemic dysfunctions associated with obesity. In this regard, we analyzed in vivo whether the lack of p85α in BAT (BATp85αKO) could modulate the activity and insulin signaling of this tissue, thereby improving diet-induced obesity and its associated metabolic complications. METHODS: We generated BATp85αKO mice using Cre-LoxP technology, specifically deleting p85α in a conditional manner. To characterize this new mouse model, we used mice of 6 and 12 months of age. In addition, BATp85αKO mice were submitted to a high-fat diet (HFD) to challenge BAT functionality. RESULTS: Our results suggest that the loss of p85α in BAT improves its thermogenic functionality, high-fat diet-induced adiposity and body weight, insulin resistance, and liver steatosis. The potential mechanisms involved in the improvement of obesity include (1) increased insulin signaling and lower activation of JNK in BAT, (2) enhanced insulin receptor isoform B (IRB) expression and association with IRS-1 in BAT, (3) lower production of proinflammatory cytokines by the adipose organ, (4) increased iWAT browning, and (5) improved liver steatosis. CONCLUSIONS: Our results provide new mechanisms involved in the resistance to obesity development, supporting the hypothesis that the gain of BAT activity induced by the lack of p85α has a direct impact on the prevention of diet-induced obesity and its associated metabolic complications.


Subject(s)
Adipose Tissue, Brown/metabolism , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Obesity/metabolism , Animals , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/chemically induced
3.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R627-R637, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29791203

ABSTRACT

Systemic insulin resistance and glucose intolerance occur with as little as 3 days of a high-fat diet (HFD) in mice and humans; the mechanisms that initiate acute insulin resistance are unknown. Most laboratories house mice at 22°C, which is below their thermoneutral temperature (~30°C). Cold stress has been shown to increase white adipose tissue (WAT) browning, alter lipid trafficking, and impair immune function, whereas energy intake and expenditure decrease with increasing ambient temperature; importantly, dysregulation of these parameters has been strongly linked to obesity-induced insulin resistance. Therefore, we compared acute changes in glucose metabolism and the metabolic phenotype in lean mice in response to a control diet or HFD housed at standard vivarium (22°C) and thermoneutral (30°C) temperatures. Glucose intolerance occurred following 1 or 5 days of HFD and was independent of housing temperature or adiposity; however, the reduction in tissue-specific glucose clearance with HFD diverged by temperature with reduced brown adipose tissue (BAT) glucose uptake at 22°C but reduced soleus glucose uptake at 30°C. Fasting glucose, food intake, and energy expenditure were significantly lower at 30°C, independent of diet. Additionally, markers of browning in both BAT and inguinal subcutaneous WAT, but not perigonadal epididymal WAT, decreased at 30°C. Together, we find housing temperature has a significant impact on the cellular pathways that regulate glucose tolerance in response to an acute HFD exposure. Thus, even short-term changes in housing temperature should be highly considered in interpretation of metabolic studies in mice.


Subject(s)
Adipose Tissue, Brown/metabolism , Blood Glucose/metabolism , Body Temperature Regulation , Diet, High-Fat , Energy Metabolism , Glucose Intolerance/blood , Housing, Animal , Subcutaneous Fat/metabolism , Temperature , Adipose Tissue, Brown/physiopathology , Animals , Biomarkers/blood , Circadian Rhythm , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Class Ia Phosphatidylinositol 3-Kinase/genetics , Disease Models, Animal , Eating , Feeding Behavior , Glucose Intolerance/etiology , Glucose Intolerance/physiopathology , Glucose Intolerance/psychology , Insulin Resistance , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Subcutaneous Fat/physiopathology , Time Factors
4.
Sci Rep ; 7(1): 6187, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28733636

ABSTRACT

We investigated the role of the PI3K p85α subunit in the development of acute colitis with a focus on intestinal macrophages. Experimental acute colitis was induced using 3% dextran sulfate sodium (DSS) in drinking water for 7 days. The severity of DSS-induced acute colitis was significantly attenuated in p85α hetero-deficient (p85α+/-) mice compared with WT mice. The expression of proinflammatory mediators in intestinal macrophages isolated from the inflamed colonic mucosa was significantly suppressed in p85α+/- colitis mice compared with WT colitis mice. Interestingly, we found that bone marrow-derived macrophages (BMDMs) from p85α+/- mice produced a significantly higher amount of IL-10 than BMDMs from WT mice. The adoptive transfer of p85α+/- BMDMs, but not WT BMDMs, significantly improved the severity in WT colitis mice, and this effect was reversed by anti-IL-10 antibody. Furthermore, the expression of IL-10 in the intestinal macrophages of p85α+/- normal colonic mucosa was significantly higher than that in the intestinal macrophages of WT normal colonic mucosa. The present results demonstrate that p85α+/- mice exhibit a reduced susceptibility to DSS-induced acute colitis. Our study suggests that a deficiency of PI3K p85α enhances the production of IL-10 in intestinal macrophages, thereby suppressing the development of DSS-induced acute colitis.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/deficiency , Colitis/drug therapy , Dextran Sulfate/adverse effects , Interleukin-10/metabolism , Macrophages/transplantation , Adoptive Transfer , Animals , Colitis/chemically induced , Colitis/metabolism , Disease Models, Animal , Macrophages/metabolism , Male , Mice , Severity of Illness Index
5.
Nat Commun ; 6: 6496, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25754093

ABSTRACT

Endoplasmic reticulum (ER) stress is associated with diabetic nephropathy (DN), but its pathophysiological relevance and the mechanisms that compromise adaptive ER signalling in podocytes remain unknown. Here we show that nuclear translocation of the transcription factor spliced X-box binding protein-1 (sXBP1) is selectively impaired in DN, inducing activating transcription factor-6 (ATF6) and C/EBP homology protein (CHOP). Podocyte-specific genetic ablation of XBP1 or inducible expression of ATF6 in mice aggravates DN. sXBP1 lies downstream of insulin signalling and attenuating podocyte insulin signalling by genetic ablation of the insulin receptor or the regulatory subunits phosphatidylinositol 3-kinase (PI3K) p85α or p85ß impairs sXBP1 nuclear translocation and exacerbates DN. Corroborating our findings from murine DN, the interaction of sXBP1 with p85α and p85ß is markedly impaired in the glomerular compartment of human DN. Thus, signalling via the insulin receptor, p85, and XBP1 maintains podocyte homeostasis, while disruption of this pathway impairs podocyte function in DN.


Subject(s)
Activating Transcription Factor 6/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , DNA-Binding Proteins/genetics , Diabetes Mellitus, Experimental/genetics , Diabetic Nephropathies/genetics , Endoplasmic Reticulum Stress/genetics , Podocytes/metabolism , Transcription Factors/genetics , Activating Transcription Factor 6/deficiency , Animals , Class Ia Phosphatidylinositol 3-Kinase/deficiency , DNA-Binding Proteins/deficiency , Databases, Factual , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation , Humans , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Podocytes/pathology , Receptor, Insulin/deficiency , Receptor, Insulin/genetics , Regulatory Factor X Transcription Factors , Signal Transduction , Streptozocin , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription Factors/deficiency , X-Box Binding Protein 1
6.
Proc Natl Acad Sci U S A ; 111(3): 1192-7, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395790

ABSTRACT

In insulin resistant states such as type 2 diabetes, there is a high demand on the ß-cell to synthesize and secrete insulin, which challenges the ability of the endoplasmic reticulum (ER) to synthesize and fold nascent proteins. This creates a state of ER stress that triggers a coordinated program referred to as the unfolded protein response (UPR) that attempts to restore ER homeostasis. We identified a role for the p85α regulatory subunit of PI3K to modulate the UPR by promoting the nuclear localization of X-box binding protein 1, a transcription factor central to the UPR. In the present study we demonstrate that reducing p85α expression in ß-cells can markedly delay the onset and severity of the diabetic phenotype observed in Akita(+/-) mice, which express a mutant insulin molecule. This is due to a decrease in activation of ER stress-dependent apoptotic pathways and a preservation of ß-cell mass and function. These data demonstrate that modulation of p85α can protect pancreatic ß-cells from ER stress, pointing to a potentially therapeutic target in diabetic states.


Subject(s)
Apoptosis , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Insulin-Secreting Cells/cytology , Alleles , Animals , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation, Enzymologic , Genotype , Glucose/metabolism , Hyperglycemia/metabolism , Insulin/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Size , Oxidative Stress , Pancreas/physiology , Phenotype , Protein Denaturation , Protein Folding , Time Factors
7.
Mol Endocrinol ; 27(9): 1468-82, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23820902

ABSTRACT

In the mammalian ovary, primordial follicles are generated early in life and remain dormant for prolonged periods. Their growth resumes via primordial follicle activation, and they continue to grow until the preovulatory stage under the regulation of hormones and growth factors, such as estrogen, FSH, and IGF-1. Both FSH and IGF-1 activate the phosphatidylinositol-3 kinase (PI3K)/Akt (acute transforming retrovirus thymoma protein kinase) signaling pathway in granulosa cells (GCs), yet it remains inconclusive whether the PI3K pathway is crucial for follicle growth. In this study, we investigated the p110δ isoform (encoded by the Pik3cd gene) of PI3K catalytic subunit expression in the mouse ovary and its function in fertility. Pik3cd-null females were subfertile, exhibited fewer growing follicles and more atretic antral follicles in the ovary, and responded poorly to exogenous gonadotropins compared with controls. Ovary transplantation showed that Pik3cd-null ovaries responded poorly to FSH stimulation in vitro; this confirmed that the follicle growth defect was intrinsically ovarian. In addition, estradiol (E2)-stimulated follicle growth and GC proliferation in preantral follicles was impaired in Pik3cd-null ovaries. FSH and E2 substantially activated the PI3K/Akt pathway in GCs of control mice but not in those of Pik3cd-null mice. However, primordial follicle activation and oocyte meiotic maturation were not affected by Pik3cd knockout. Taken together, our findings indicate that the p110δ isoform of the PI3K catalytic subunit is a key component of the PI3K pathway for both FSH and E2-stimulated follicle growth in ovarian GCs; however, it is not required for primordial follicle activation and oocyte development.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Estrogens/metabolism , Follicle Stimulating Hormone/metabolism , Ovarian Follicle/enzymology , Ovarian Follicle/growth & development , Animals , Cell Proliferation/drug effects , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Enzyme Activation/drug effects , Estradiol/pharmacology , Female , Gene Deletion , Gonadotropins/pharmacology , Granulosa Cells/drug effects , Granulosa Cells/enzymology , Granulosa Cells/pathology , Homozygote , Humans , Infertility, Female/enzymology , Infertility, Female/pathology , Meiosis/drug effects , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Oocytes/drug effects , Oocytes/pathology , Ovarian Follicle/drug effects , Ovarian Follicle/pathology , Reproducibility of Results , Signal Transduction/drug effects , Steroids/biosynthesis
8.
PLoS One ; 7(8): e43668, 2012.
Article in English | MEDLINE | ID: mdl-22928013

ABSTRACT

Of the various phosphatidylinositol 3- kinases (PI3Ks), only the class III enzyme Vps34 has been shown to regulate phagosome maturation. During studies of phagosome maturation in THP-1 cells deficient in class IA PI3K p110α, we discovered that this PI3K isoform is required for vacuole maturation to progress beyond acquisition of Rab7 leading to delivery of lysosomal markers. Bead phagosomes from THP-1 cells acquired p110α and contained PI3P and PI(3,4,5)P3; however, p110α and PI(3,4,5)P3 levels in phagosomes from p110α knockdown cells were decreased. Phagosomes from p110α knock down cells showed normal acquisition of both Rab5 and EEA-1, but were markedly deficient in the lysosomal markers LAMP-1 and LAMP-2, and the lysosomal hydrolase, ß-galactosidase. Phagosomes from p110α deficient cells also displayed impaired fusion with Texas Red dextran-loaded lysosomes. Despite lacking lysosomal components, phagosomes from p110α deficient cells recruited normal levels of Rab7, Rab-interacting lysosomal protein (RILP) and homotypic vacuole fusion and protein sorting (HOPs) components Vps41 and Vps16. The latter observations demonstrated that phagosomal Rab7 was active and capable of recruiting effectors involved in membrane fusion. Nevertheless, active Rab7 was not sufficient to bring about the delivery of lysosomal proteins to the maturing vacuole, which is shown for the first time to be dependent on a class I PI3K.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/metabolism , Phagosomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cell Line, Tumor , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Class Ia Phosphatidylinositol 3-Kinase/genetics , Gene Knockdown Techniques , Humans , Lysosomal-Associated Membrane Protein 1/metabolism , Lysosomes/metabolism , Phagosomes/enzymology , Phosphatidylinositol Phosphates/metabolism , Vesicular Transport Proteins/metabolism , beta-Galactosidase/metabolism , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
9.
Haematologica ; 97(7): 1042-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22315502

ABSTRACT

Juvenile myelomonocytic leukemia is a lethal disease of children characterized by hypersensitivity of hematopoietic progenitors to granulocyte macrophage-colony stimulating factor. Mutations in PTPN11, the gene encoding the protein tyrosine phosphatase Shp2, are common in juvenile myelomonocytic leukemia and induce hyperactivation of the phosphoinositide-3-kinase pathway. We found that genetic disruption of Pik3r1, the gene encoding the Class IA phosphoinositide-3-kinase regulatory subunits p85α, p55α and p50α, significantly reduced hyperproliferation and hyperphosphorylation of Akt in gain-of-function Shp2 E76K-expressing cells. Elevated protein levels of the phosphoinositide-3-kinase catalytic subunit, p110δ, in the Shp2 E76K-expressing Pik3r1-/- cells suggest that p110δ may be a crucial mediator of mutant Shp2-induced phosphoinositide-3-kinase hyperactivation. Consistently, treatment with the p110δ-specific inhibitor, IC87114, or the clinical grade pan-phosphoinositide-3-kinase inhibitor, GDC-0941, reduced granulocyte macrophage-colony stimulating factor hypersensitivity. Treatment with the farnesyltransferase inhibitor, tipifarnib, showed that Shp2 E76K induces hyperactivation of phosphoinositide-3-kinase by both Ras-dependent and Ras-independent mechanisms. Collectively, these findings implicate Class IA phosphoinositide-3-kinase as a relevant molecular target in juvenile myelomonocytic leukemia.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Child , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/genetics , Farnesyltranstransferase/antagonists & inhibitors , Farnesyltranstransferase/genetics , Gene Expression/drug effects , Humans , Leukemia, Myelomonocytic, Juvenile/drug therapy , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/metabolism , Mice , Mutation , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/drug effects
10.
J Exp Med ; 209(3): 463-70, 2012 Mar 12.
Article in English | MEDLINE | ID: mdl-22351933

ABSTRACT

Whole exome sequencing was used to determine the causative gene in patients with B cell defects of unknown etiology. A homozygous premature stop codon in exon 6 of PIK3R1 was identified in a young woman with colitis and absent B cells. The mutation results in the absence of p85α but normal expression of the p50α and p55α regulatory subunits of PI3K. Bone marrow aspirates from the patient showed <0.1% CD19(+) B cells with normal percentages of TdT(+)VpreB(+)CD19(-) B cell precursors. This developmental block is earlier than that seen in patients with defects in the B cell receptor signaling pathway or in a strain of engineered mice with a similar defect in p85α. The number and function of the patient's T cells were normal. However, Western blot showed markedly decreased p110δ, as well as absent p85α, in patient T cells, neutrophils, and dendritic cells. The patient had normal growth and development and normal fasting glucose and insulin. Mice with p85α deficiency have insulin hypersensitivity, defective platelet function, and abnormal mast cell development. In contrast, the absence of p85α in the patient results in an early and severe defect in B cell development but minimal findings in other organ systems.


Subject(s)
Agammaglobulinemia/enzymology , Agammaglobulinemia/genetics , B-Lymphocytes/enzymology , B-Lymphocytes/immunology , Class Ia Phosphatidylinositol 3-Kinase/deficiency , Class Ia Phosphatidylinositol 3-Kinase/genetics , Agammaglobulinemia/immunology , Amino Acid Sequence , Animals , B-Lymphocytes/pathology , Base Sequence , Case-Control Studies , Cell Differentiation/genetics , Codon, Nonsense , Cytokines/biosynthesis , DNA Mutational Analysis , Dendritic Cells/immunology , Exons , Female , Homozygote , Humans , Male , Mice , Mice, Knockout , Pedigree , Young Adult
11.
Nat Genet ; 21(2): 230-5, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9988280

ABSTRACT

The hallmark of type 2 diabetes, the most common metabolic disorder, is a defect in insulin-stimulated glucose transport in peripheral tissues. Although a role for phosphoinositide-3-kinase (PI3K) activity in insulin-stimulated glucose transport and glucose transporter isoform 4 (Glut4) translocation has been suggested in vitro, its role in vivo and the molecular link between activation of PI3K and translocation has not yet been elucidated. To determine the role of PI3K in glucose homeostasis, we generated mice with a targeted disruption of the gene encoding the p85alpha regulatory subunit of PI3K (Pik3r1; refs 3-5). Pik3r1-/- mice showed increased insulin sensitivity and hypoglycaemia due to increased glucose transport in skeletal muscle and adipocytes. Insulin-stimulated PI3K activity associated with insulin receptor substrates (IRSs) was mediated via full-length p85 alpha in wild-type mice, but via the p50 alpha alternative splicing isoform of the same gene in Pik3r1-/- mice. This isoform switch was associated with an increase in insulin-induced generation of phosphatidylinositol(3,4,5)triphosphate (PtdIns(3,4,5)P3) in Pik3r1-/- adipocytes and facilitation of Glut4 translocation from the low-density microsome (LDM) fraction to the plasma membrane (PM). This mechanism seems to be responsible for the phenotype of Pik3r1-/- mice, namely increased glucose transport and hypoglycaemia. Our work provides the first direct evidence that PI3K and its regulatory subunit have a role in glucose homeostasis in vivo.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/deficiency , Class Ia Phosphatidylinositol 3-Kinase/genetics , Hypoglycemia/genetics , Insulin/pharmacology , Phosphatidylinositol 3-Kinases/deficiency , Phosphatidylinositol 3-Kinases/genetics , Animals , Biological Transport/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Crosses, Genetic , Deoxyglucose/metabolism , Enzyme Activation/genetics , Glucose/metabolism , Isoenzymes/deficiency , Isoenzymes/genetics , Isoenzymes/metabolism , Kinetics , Mice , Mice, Knockout , Muscle, Skeletal/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Subcellular Fractions/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...