Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Nature ; 543(7643): 122-125, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28178237

ABSTRACT

Human cells have twenty-three pairs of chromosomes. In cancer, however, genes can be amplified in chromosomes or in circular extrachromosomal DNA (ecDNA), although the frequency and functional importance of ecDNA are not understood. We performed whole-genome sequencing, structural modelling and cytogenetic analyses of 17 different cancer types, including analysis of the structure and function of chromosomes during metaphase of 2,572 dividing cells, and developed a software package called ECdetect to conduct unbiased, integrated ecDNA detection and analysis. Here we show that ecDNA was found in nearly half of human cancers; its frequency varied by tumour type, but it was almost never found in normal cells. Driver oncogenes were amplified most commonly in ecDNA, thereby increasing transcript level. Mathematical modelling predicted that ecDNA amplification would increase oncogene copy number and intratumoural heterogeneity more effectively than chromosomal amplification. We validated these predictions by quantitative analyses of cancer samples. The results presented here suggest that ecDNA contributes to accelerated evolution in cancer.


Subject(s)
DNA Copy Number Variations/genetics , Evolution, Molecular , Gene Amplification/genetics , Genetic Heterogeneity , Models, Genetic , Neoplasms/genetics , Oncogenes/genetics , Chromosomes, Human/genetics , Cytogenetic Analysis , DNA Mutational Analysis , Genome, Human/genetics , Humans , Metaphase/genetics , Neoplasms/classification , RNA, Messenger/analysis , RNA, Neoplasm/genetics , Reproducibility of Results , Software
2.
Nat Immunol ; 10(5): 504-13, 2009 May.
Article in English | MEDLINE | ID: mdl-19363483

ABSTRACT

Foxo transcription factors regulate cell cycle progression, cell survival and DNA-repair pathways. Here we demonstrate that deficiency in Foxo3 resulted in greater expansion of T cell populations after viral infection. This exaggerated expansion was not T cell intrinsic. Instead, it was caused by the enhanced capacity of Foxo3-deficient dendritic cells to sustain T cell viability by producing more interleukin 6. Stimulation of dendritic cells mediated by the coinhibitory molecule CTLA-4 induced nuclear localization of Foxo3, which in turn inhibited the production of interleukin 6 and tumor necrosis factor. Thus, Foxo3 acts to constrain the production of key inflammatory cytokines by dendritic cells and to control T cell survival.


Subject(s)
Dendritic Cells/immunology , Forkhead Transcription Factors/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation/immunology , Antigens, CD/immunology , Antigens, CD/metabolism , Arenaviridae Infections/immunology , Blotting, Western , CTLA-4 Antigen , Dendritic Cells/metabolism , Flow Cytometry , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Interleukin-6/immunology , Interleukin-6/metabolism , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Congenic , Mice, Transgenic , Protein Transport/immunology , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
3.
Diabetes ; 58(6): 1275-82, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19289458

ABSTRACT

OBJECTIVE: Forkhead box O (FoxO) transcription factors represent evolutionarily conserved targets of insulin signaling, regulating metabolism and cellular differentiation in response to changes in nutrient availability. Although the FoxO1 isoform is known to play a key role in adipogenesis, its physiological role in differentiated adipose tissue remains unclear. RESEARCH DESIGN AND METHODS: In this study, we analyzed the phenotype of FoxO1 haploinsufficient mice to investigate the role of FoxO1 in high-fat diet-induced obesity and adipose tissue metabolism. RESULTS: We showed that reduced FoxO1 expression protects mice against obesity-related insulin resistance with marked improvement not only in hepatic insulin sensitivity but also in skeletal muscle insulin action. FoxO1 haploinsufficiency also resulted in increased peroxisome proliferator-activated receptor (PPAR)gamma gene expression in adipose tissue, with enhanced expression of PPARgamma target genes known to influence metabolism. Moreover, treatment of mice with the PPARgamma agonist rosiglitazone caused a greater improvement in in vivo insulin sensitivity in FoxO1 haploinsufficient animals, including reductions in circulating proinflammatory cytokines. CONCLUSIONS: These findings indicate that FoxO1 proteins negatively regulate insulin action and that their effect may be explained, at least in part, by inhibition of PPARgamma function.


Subject(s)
Adipose Tissue/physiology , Dietary Fats/pharmacology , Forkhead Transcription Factors/deficiency , Hypoglycemic Agents/pharmacology , Insulin Resistance/physiology , PPAR gamma/genetics , Thiazolidinediones/pharmacology , Animals , Forkhead Box Protein O1 , Gene Deletion , Glucose Clamp Technique , Male , Mice , Phenotype , RNA, Messenger/genetics , Reference Values , Reverse Transcriptase Polymerase Chain Reaction
4.
Nat Immunol ; 9(12): 1388-98, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18978794

ABSTRACT

The transcription factors Foxo1, Foxo3 and Foxo4 modulate cell fate 'decisions' in diverse systems. Here we show that Foxo1-dependent gene expression was critical at many stages of B cell differentiation. Early deletion of Foxo1 caused a substantial block at the pro-B cell stage due to a failure to express interleukin 7 receptor-alpha. Foxo1 inactivation in late pro-B cells resulted in an arrest at the pre-B cell stage due to lower expression of the recombination-activating genes Rag1 and Rag2. Deletion of Foxo1 in peripheral B cells led to fewer lymph node B cells due to lower expression of L-selectin and failed class-switch recombination due to impaired upregulation of the gene encoding activation-induced cytidine deaminase. Thus, Foxo1 regulates a transcriptional program that is essential for early B cell development and peripheral B cell function.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation/immunology , Forkhead Transcription Factors/immunology , Animals , B-Lymphocytes/immunology , Blotting, Southern , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression/immunology , Gene Rearrangement, B-Lymphocyte/genetics , Homeodomain Proteins/immunology , Homeodomain Proteins/metabolism , Immunohistochemistry , Mice , Mice, Mutant Strains , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/immunology , Stem Cells/metabolism , Transcription, Genetic/immunology
5.
Cell Cycle ; 7(7): 837-41, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18414034

ABSTRACT

Human cancer cells frequently harbor chromosomal translocations that create chimeric fusion genes. The t(2;13) translocation is characteristic of the pediatric muscle tumor, alveolar rhabdomyosarcoma, and produces the chimeric transcription factor, PAX3-FOXO1, that contains the DNA binding elements of PAX3 and the transcriptional activation domain of FOXO1. Experiments designed to determine how PAX3-FOXO1 expression contributes to the development of muscle cell-derived tumors resulted in the discovery that the fusion protein misregulates gene expression and interrupts myogenic differentiation through a unique gain of function mechanism. These results yield new insight into how tumor-associated genetic alterations increase the likelihood of cancer formation and may lead to new therapeutic approaches.


Subject(s)
Cell Differentiation/physiology , Early Growth Response Protein 1/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Paired Box Transcription Factors/metabolism , Rhabdomyosarcoma, Alveolar/metabolism , Blotting, Western , Cell Line , Forkhead Box Protein O1 , Humans , Immunoprecipitation , Models, Biological , Myoblasts/cytology , PAX3 Transcription Factor , Ubiquitination
6.
Genome Res ; 18(1): 46-59, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18042645

ABSTRACT

By integrating genome-wide maps of RNA polymerase II (Polr2a) binding with gene expression data and H3ac and H3K4me3 profiles, we characterized promoters with enriched activity in mouse embryonic stem cells (mES) as well as adult brain, heart, kidney, and liver. We identified approximately 24,000 promoters across these samples, including 16,976 annotated mRNA 5' ends and 5153 additional sites validating cap-analysis of gene expression (CAGE) 5' end data. We showed that promoters with CpG islands are typically non-tissue specific, with the majority associated with Polr2a and the active chromatin modifications in nearly all the tissues examined. By contrast, the promoters without CpG islands are generally associated with Polr2a and the active chromatin marks in a tissue-dependent way. We defined 4396 tissue-specific promoters by adapting a quantitative index of tissue-specificity based on Polr2a occupancy. While there is a general correspondence between Polr2a occupancy and active chromatin modifications at the tissue-specific promoters, a subset of them appear to be persistently marked by active chromatin modifications in the absence of detectable Polr2a binding, highlighting the complexity of the functional relationship between chromatin modification and gene expression. Our results provide a resource for exploring promoter Polr2a binding and epigenetic states across pluripotent and differentiated cell types in mammals.


Subject(s)
Chromosome Mapping , CpG Islands/physiology , Embryonic Stem Cells/physiology , Gene Expression Regulation/physiology , Genome/physiology , Promoter Regions, Genetic/physiology , Animals , Cell Differentiation/physiology , Chromatin/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Female , Mice , Organ Specificity/physiology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/physiology , RNA Polymerase II/genetics , RNA Polymerase II/metabolism
7.
Proc Natl Acad Sci U S A ; 104(46): 18085-90, 2007 Nov 13.
Article in English | MEDLINE | ID: mdl-17986608

ABSTRACT

The chimeric protein PAX3-FOXO1, resulting from a translocation between chromosomes 2 and 13, is the most common genetic aberration in the alveolar subtype of the human skeletal muscle tumor, rhabdomyosarcoma. To understand how PAX3-FOXO1 contributes to tumor development, we isolated and characterized muscle cells from transgenic mice expressing PAX3-FOXO1 under control of the PAX3 promoter. We demonstrate that these myoblasts are unable to complete myogenic differentiation because of an inability to up-regulate p57Kip2 transcription. This defect is caused by reduced levels of the EGR1 transcriptional activator resulting from a direct, destabilizing interaction with PAX3-FOXO1. Neither PAX3 nor FOXO1 share the ability to regulate p57Kip2 transcription. Thus, the breakage and fusion of the genes encoding these transcription factors creates a unique chimeric protein that controls a key cell-cycle and -differentiation regulator.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/genetics , Early Growth Response Protein 1/metabolism , Forkhead Transcription Factors/physiology , Gene Expression Regulation/physiology , Paired Box Transcription Factors/physiology , Animals , Electrophoretic Mobility Shift Assay , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Humans , Hydrolysis , Mice , Mice, Transgenic , Muscle Neoplasms/genetics , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Rhabdomyosarcoma/genetics
8.
Cell ; 128(2): 235-7, 2007 Jan 26.
Article in English | MEDLINE | ID: mdl-17254960

ABSTRACT

The FoxO transcription factors have been implicated in many processes including tumor suppression and cell death. In this issue, two groups now report on mice that conditionally lack the three predominant FoxO transcription factors. Demonstrate that FoxOs are critical for the long-term maintenance of hematopoietic stem cells, and show that FoxOs suppress the formation of hemangiomas and lymphomas in mice.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Forkhead Transcription Factors/metabolism , Hemangioma/genetics , Hematopoietic Stem Cells/metabolism , Lymphoma/genetics , Tumor Suppressor Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic/genetics , Hemangioma/metabolism , Humans , Lymphoma/metabolism , Tumor Suppressor Proteins/genetics
9.
Transgenic Res ; 15(5): 595-614, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16952014

ABSTRACT

The t(2;13) chromosomal translocation is found in the majority of human alveolar rhabdomyosarcomas (RMS). The resulting PAX3-FKHR fusion protein contains PAX3 DNA-binding domains fused to the potent transactivation domain of FKHR, suggesting that PAX3-FKHR functions to deregulate PAX3-specific target genes and signaling pathways. We previously developed transgenic mice expressing PAX3-FKHR under the control of mouse Pax3 regulatory sequences to test this hypothesis. We reported that PAX3-FKHR interferes with normal Pax3 developmental functions, with mice exhibiting neural tube and neural crest abnormalities that mimic those found in Pax3-deficient Splotch mice. Here we expanded those studies to show that developmental expression of PAX3-FKHR results in aberrant myogenesis in the developing somites and neural tube, leading to ectopic skeletal muscle formation in the mature spinal cord. Gene expression profiling indicated that PAX3-FKHR expression in the developing neural tube induces a myogenic pattern of gene expression at the expense of the normal neurogenic program. Somite defects in PAX3-FKHR transgenic animals resulted in skeletal malformations that included rib fusions and mis-attachments. As opposed to the neural tube defects, the severity of the rib phenotype was rescued by reducing Pax3 levels through mating with Splotch mice. Embryos from the transgenic line expressing the highest levels of PAX3-FKHR had severe neural tube defects, including exencephaly, and almost half of the embryos died between gestational ages E13.5-E15.5. Nearly all of the embryos that survived to term died after birth due to severe spina bifida, rather than the absence of a muscular diaphragm. These studies reveal a prominent role for PAX3-FKHR in disrupting Pax3 functions and in deregulating skeletal muscle development, suggesting that this fusion protein plays a critical role in the pathogenesis of alveolar RMS by influencing the commitment and differentiation of the myogenic cell lineage.


Subject(s)
Choristoma/genetics , Forkhead Transcription Factors/genetics , Muscle Development/genetics , Neural Tube Defects/genetics , Paired Box Transcription Factors/genetics , Recombinant Fusion Proteins/genetics , Animals , Cell Differentiation/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors/physiology , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Neural Tube Defects/pathology , PAX3 Transcription Factor , Paired Box Transcription Factors/physiology , Recombinant Fusion Proteins/physiology , Rhabdomyosarcoma, Alveolar/etiology , Rhabdomyosarcoma, Alveolar/genetics , Rhabdomyosarcoma, Alveolar/pathology , Somites/pathology
10.
Exp Gerontol ; 41(8): 709-17, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16806782

ABSTRACT

The FOXO family of transcription factors has been implicated in several cellular processes including cell cycle arrest, cell death and protection from stress stimuli. FOXO function is influenced by multiple signaling pathways. Many of these pathways are known to be misregulated in cancer. Perturbation of FOXO function leads to uncontrolled cell proliferation and accumulation of DNA damage. It is becoming clear that active FOXO proteins play an important role in keeping cells in check and inactivation of FOXO proteins is associated with characteristics of cancer cells. FOXO proteins may represent new therapeutic targets for a broad spectrum of cancers.


Subject(s)
Cell Transformation, Neoplastic/genetics , Forkhead Transcription Factors/physiology , Neoplasms/genetics , Acetylation , Animals , Antineoplastic Agents/pharmacology , DNA Damage , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Neoplasm Proteins/physiology , Phosphorylation , Ubiquitin/metabolism
11.
Nat Genet ; 38(5): 589-93, 2006 May.
Article in English | MEDLINE | ID: mdl-16642023

ABSTRACT

The total mass of islets of Langerhans is reduced in individuals with type 2 diabetes, possibly contributing to the pathogenesis of this condition. Although the regulation of islet mass is complex, recent studies have suggested the importance of a signaling pathway that includes the insulin or insulin-like growth factor-1 receptors, insulin receptor substrate and phosphatidylinositol (PI) 3-kinase. 3-Phosphoinositide-dependent protein kinase 1 (PDK1) is a serine-threonine kinase that mediates signaling downstream of PI 3-kinase. Here we show that mice that lack PDK1 specifically in pancreatic beta cells (betaPdk1-/- mice) develop progressive hyperglycemia as a result of a loss of islet mass. The mice show reductions in islet density as well as in the number and size of cells. Haploinsufficiency of the gene for the transcription factor Foxo1 resulted in a marked increase in the number, but not the size, of cells and resulted in the restoration of glucose homeostasis in betaPdk1-/- mice. These results suggest that PDK1 is important in maintenance of pancreatic cell mass and glucose homeostasis.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Islets of Langerhans/enzymology , Islets of Langerhans/pathology , Protein Serine-Threonine Kinases/genetics , 3-Phosphoinositide-Dependent Protein Kinases , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Mice , Mice, Knockout , Signal Transduction
12.
Science ; 307(5716): 1720b-1721b, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15778202
13.
Science ; 307(5716): 1720-1, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15779114
14.
Cell ; 117(4): 421-6, 2004 May 14.
Article in English | MEDLINE | ID: mdl-15137936

ABSTRACT

Forkhead transcription factors of the FoxO subfamily are emerging as a shared component among pathways regulating diverse cellular functions, such as differentiation, metabolism, proliferation, and survival. Their transcriptional output is controlled via a two-tiered mechanism of phosphorylation and acetylation. Modest alterations of this balance can result in profound effects. The gamut of phenotypes runs from protection against diabetes and predisposition to neoplasia, conferred by FoxO loss of function, to increased cellular survival and a marked catabolic response associated with gain of function.


Subject(s)
Cell Transformation, Neoplastic/genetics , Energy Metabolism/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Acetylation , Animals , Cell Differentiation/genetics , Cell Survival/genetics , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Genes, Lethal/genetics
15.
Mol Cell ; 14(4): 416-8, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15149589

ABSTRACT

Two recent reports reveal new roles for FoxO proteins in cell proliferation and tumorigenesis. Seoane and colleagues show that FoxO proteins play key roles in the TGFbeta-dependent activation of p21Cip1 by partnering with Smad3 and Smad4. FoxG1, a protein from a distinct Fox subfamily, binds FoxO/Smad complexes and blocks p21Cip1 expression. These interactions establish a relationship between the PI3K pathway, FoxG1, and the TGFbeta/Smad pathways. The second report identifies IkappaB kinase as a negative regulator of FoxO proteins, suggesting a mechanism for relieving negative regulation of cell cycle and promoting tumor cell proliferation.


Subject(s)
Cell Transformation, Neoplastic/genetics , Signal Transduction/genetics , Transcription Factors/genetics , Animals , Apoptosis Regulatory Proteins , Carrier Proteins/genetics , Cell Division/genetics , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins/genetics , Forkhead Box Protein O1 , Forkhead Transcription Factors , Humans , Intracellular Signaling Peptides and Proteins , Mitochondrial Proteins/genetics , Nerve Tissue Proteins/genetics , Phosphatidylinositol 3-Kinases/genetics , Transforming Growth Factor beta/genetics
16.
Proc Natl Acad Sci U S A ; 101(9): 2975-80, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14978268

ABSTRACT

Genetic analysis in Caenorhabditis elegans has uncovered essential roles for DAF-16 in longevity, metabolism, and reproduction. The mammalian orthologs of DAF-16, the closely-related FOXO subclass of forkhead transcription factors (FKHR/FOXO1, FKHRL1/FOXO3a, and AFX/FOXO4), also have important roles in cell cycle arrest, apoptosis and stress responses in vitro, but their in vivo physiological roles are largely unknown. To elucidate their role in normal development and physiology, we disrupted each of the Foxo genes in mice. Foxo1-null embryos died on embryonic day 10.5 as a consequence of incomplete vascular development. Foxo1-null embryonic and yolk sac vessels were not well developed at embryonic day 9.5, and Foxo1 expression was found in a variety of embryonic vessels, suggesting a crucial role of this transcription factor in vascular formation. On the other hand, both Foxo3a- and Foxo4-null mice were viable and grossly indistinguishable from their littermate controls, indicating dispensability of these two members of the Foxo transcription factor family for normal vascular development. Foxo3a-null females showed age-dependent infertility and had abnormal ovarian follicular development. In contrast, histological analyses of Foxo4-null mice did not identify any consistent abnormalities. These results demonstrate that the physiological roles of Foxo genes are functionally diverse in mammals.


Subject(s)
Embryonic and Fetal Development/genetics , Gene Expression Regulation, Developmental/genetics , Genetic Variation , Transcription Factors/genetics , Animals , Female , Fetal Death , Forkhead Box Protein O1 , Forkhead Transcription Factors , Infertility, Female/genetics , Male , Mice , Multigene Family , Neovascularization, Physiologic/genetics , Ovary/embryology , Sequence Deletion , Yolk Sac/physiology
17.
Proc Natl Acad Sci U S A ; 101(2): 615-20, 2004 Jan 13.
Article in English | MEDLINE | ID: mdl-14722351

ABSTRACT

Prostate tumors are complex entities composed of malignant cells mixed and interacting with nonmalignant cells. However, molecular analyses by standard gene expression profiling are limited because spatial information and nontumor cell types are lost in sample preparation. We scored 88 prostate specimens for relative content of tumor, benign hyperplastic epithelium, stroma, and dilated cystic glands. The proportions of these cell types were then linked in silico to gene expression levels determined by microarray analysis, revealing unique cell-specific profiles. Gene expression differences for malignant and nonmalignant epithelial cells (tumor versus benign hyperplastic epithelium) could be identified without being confounded by contributions from stroma that dominate many samples or sacrificing possible paracrine influences. Cell-specific expression of selected genes was validated by immunohistochemistry and quantitative PCR. The results provide patterns of gene expression for these three lineages with relevance to pathogenetic, diagnostic, and therapeutic considerations.


Subject(s)
Gene Expression Profiling , Prostatic Neoplasms/genetics , Humans , Immunohistochemistry , Male , Nucleic Acid Hybridization , Prostatic Neoplasms/pathology
18.
Genomics ; 82(6): 628-36, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14611804

ABSTRACT

Human SSX was first identified as the gene involved in the t(X;18) translocation in synovial sarcoma. SSX is a multigene family, with 9 complete genes on chromosome Xp11. Normally expressed almost exclusively in testis, SSX mRNA is expressed in various human tumors, defining SSX as a cancer/testis antigen. We have now cloned the mouse ortholog of SSX. Mouse SSX genes can be divided into Ssxa and Ssxb subfamilies based on sequence homology. Ssxa has only one member, whereas 12 Ssxb genes, Ssxb1 to Ssxb12, were identified by cDNA cloning from mouse testis and mouse tumors. Both Ssxa and Ssxb are located on chromosome X and show tissue-restricted mRNA expression to testis among normal tissues. All putative human and mouse SSX proteins share conserved KRAB and SSX-RD domains. Mouse tumors were found to express some, but not all, Ssxb genes, similar to the SSX activation in human tumors.


Subject(s)
Gene Expression , Multigene Family/genetics , Neoplasm Proteins/genetics , RNA, Messenger/metabolism , Repressor Proteins/genetics , Testis/metabolism , X Chromosome/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Chromosome Mapping , Gene Components , Humans , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Messenger/genetics , Restriction Mapping , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Analysis, DNA , Sequence Homology , Tumor Cells, Cultured
19.
Dev Cell ; 4(1): 119-29, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12530968

ABSTRACT

An outstanding question in adipocyte biology is how hormonal cues are relayed to the nucleus to activate the transcriptional program that promotes adipogenesis. The forkhead transcription factor Foxo1 is regulated by insulin via Akt-dependent phosphorylation and nuclear exclusion. We show that Foxo1 is induced in the early stages of adipocyte differentiation but that its activation is delayed until the end of the clonal expansion phase. Constitutively active Foxo1 prevents the differentiation of preadipocytes, while dominant-negative Foxo1 restores adipocyte differentiation of fibroblasts from insulin receptor-deficient mice. Further, Foxo1 haploinsufficiency protects from diet-induced diabetes in mice. We propose that Foxo1 plays an important role in the integration of hormone-activated signaling pathways with the complex transcriptional cascade that promotes adipocyte differentiation.


Subject(s)
Adipocytes/cytology , Adipocytes/metabolism , Cell Differentiation , Transcription Factors/metabolism , 3T3 Cells , Adipose Tissue/growth & development , Adipose Tissue/metabolism , Animals , Cell Size , Diabetes Mellitus/chemically induced , Diabetes Mellitus/metabolism , Dietary Fats/administration & dosage , Dietary Fats/pharmacology , Fibroblasts , Forkhead Box Protein O1 , Forkhead Transcription Factors , Gene Expression Regulation , Insulin Resistance , Mice , Mutation/genetics , Phosphorylation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Transcription Factors/genetics
20.
J Clin Invest ; 110(12): 1839-47, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12488434

ABSTRACT

Diabetes is caused by an absolute (type 1) or relative (type 2) deficiency of insulin-producing beta cells. The mechanisms governing replication of terminally differentiated beta cells and neogenesis from progenitor cells are unclear. Mice lacking insulin receptor substrate-2 (Irs2) develop beta cell failure, suggesting that insulin signaling is required to maintain an adequate beta cell mass. We report that haploinsufficiency for the forkhead transcription factor Foxo1 reverses beta cell failure in Irs2(-/-) mice through partial restoration of beta cell proliferation and increased expression of the pancreatic transcription factor pancreas/duodenum homeobox gene-1 (Pdx1). Foxo1 and Pdx1 exhibit mutually exclusive patterns of nuclear localization in beta cells, and constitutive nuclear expression of a mutant Foxo1 is associated with lack of Pdx1 expression. We show that Foxo1 acts as a repressor of Foxa2-dependent (Hnf-3beta-dependent) expression from the Pdx1 promoter. We propose that insulin/IGFs regulate beta cell proliferation by relieving Foxo1 inhibition of Pdx1 expression in a subset of cells embedded within pancreatic ducts.


Subject(s)
Homeodomain Proteins , Insulin/metabolism , Islets of Langerhans/growth & development , Signal Transduction/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Diabetes Mellitus, Type 2/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors , Genes, Reporter , Humans , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Islets of Langerhans/cytology , Kidney/cytology , Mice , Mice, Knockout , Microscopy, Fluorescence , Pancreas/cytology , Pancreas/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Promoter Regions, Genetic , Protein Isoforms , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Trans-Activators/genetics , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL