595 results on '"Ronald A, Depinho"'
Search Results
352. Take care of your chromosomes lest cancer take care of you
- Author
-
Richard S. Maser and Ronald A. DePinho
- Subjects
Chromosome Aberrations ,Genetics ,Cancer Research ,DNA Repair ,DNA damage ,Mutant ,Cancer ,Mice, Transgenic ,Cell Biology ,Biology ,Genes, p53 ,medicine.disease ,Mice ,chemistry.chemical_compound ,Cell Transformation, Neoplastic ,chemistry ,Oncology ,Neoplasms ,Models, Animal ,medicine ,Animals ,Humans ,DNA ,Carcinogen ,DNA Damage - Abstract
The analysis of compound mouse mutants for nonhomologous end-joining DNA double-strand break repair and those deficient for the p53 checkpoint pathway has provided a fascinating look at the carcinogenic consequences of the failure to properly repair DNA damage and to elicit appropriate checkpoints.
- Published
- 2003
- Full Text
- View/download PDF
353. Foxk1 promotes cell proliferation and represses myogenic differentiation by regulating Foxo4 and Mef2
- Author
-
Daniel J. Garry, Robert D. Gerard, Alicia M. Wallis, Ronald A. DePinho, Mary G. Garry, Xiaozhong Shi, Robert W. Grange, and Kevin A. Voelker
- Subjects
Male ,Transcription, Genetic ,Cellular differentiation ,Population ,Cell Cycle Proteins ,Biology ,Muscle Development ,Mice ,medicine ,Myocyte ,Animals ,Regeneration ,Progenitor cell ,education ,Muscle, Skeletal ,Cell Proliferation ,Mice, Knockout ,education.field_of_study ,Cell growth ,MEF2 Transcription Factors ,Cell Cycle ,Skeletal muscle ,Cell Differentiation ,Forkhead Transcription Factors ,Cell Biology ,DNA ,musculoskeletal system ,Repressor Proteins ,medicine.anatomical_structure ,Myogenic Regulatory Factors ,Myogenic regulatory factors ,Cancer research ,C2C12 ,tissues ,Research Article ,Protein Binding - Abstract
In response to severe injury, adult skeletal muscle exhibits a remarkable regenerative capacity due to a resident muscle stem/progenitor cell population. While a number of factors are expressed in the muscle progenitor cell (MPC) population, the molecular networks that govern this cell population remain an area of active investigation. In this study, utilizing knockdown techniques and overexpression of Foxk1 in the myogenic lineage, we observed dysregulation of Foxo and Mef2 downstream targets. Utilizing an array of technologies, we establish that Foxk1 represses the transcriptional activity of Foxo4 and Mef2 and physically interacts with Foxo4 and Mef2, thus promoting MPC proliferation and antagonizing the myogenic lineage differentiation program, respectively. Correspondingly, knockdown of Foxk1 in C2C12 myoblasts results in cell cycle arrest, and Foxk1 overexpression in C2C12CAR-myoblasts retards muscle differentiation. Collectively, we have established that Foxk1 promotes MPC proliferation by repressing Foxo4 transcriptional activity and inhibits myogenic differentiation by repressing Mef2 activity. These studies enhance our understanding of the transcriptional networks that regulate the MPC population and muscle regeneration.
- Published
- 2012
354. FOXO1 orchestrates the bone-suppressing function of gut-derived serotonin
- Author
-
Stavroula Kousteni, Bin Zhou, Ioanna Mosialou, Tim M. Townes, X. Edward Guo, Marie Therese Rached, Barbara C. Silva, René Hen, Ji Wang, Aruna Kode, and Ronald A. DePinho
- Subjects
medicine.medical_specialty ,Serotonin ,Genotype ,Transcription, Genetic ,Duodenum ,MAP Kinase Signaling System ,FOXO1 ,Activating Transcription Factor 4 ,CREB ,Bone remodeling ,Mice ,Genes, Reporter ,Osteogenesis ,Stress, Physiological ,Internal medicine ,medicine ,Animals ,Homeostasis ,Insulin-Like Growth Factor I ,Cyclic AMP Response Element-Binding Protein ,Promoter Regions, Genetic ,Transcription factor ,Cells, Cultured ,Osteoblasts ,biology ,Forkhead Box Protein O1 ,ATF4 ,Osteoblast ,Forkhead Transcription Factors ,General Medicine ,medicine.anatomical_structure ,Endocrinology ,Low Density Lipoprotein Receptor-Related Protein-5 ,Gene Expression Regulation ,Blood-Brain Barrier ,Organ Specificity ,biology.protein ,Receptor, Serotonin, 5-HT1B ,Bone Remodeling ,Cell Division ,Research Article - Abstract
Serotonin is a critical regulator of bone mass, fulfilling different functions depending on its site of synthesis. Brain-derived serotonin promotes osteoblast proliferation, whereas duodenal-derived serotonin suppresses it. To understand the molecular mechanisms of duodenal-derived serotonin action on osteoblasts, we explored its transcriptional mediation in mice. We found that the transcription factor FOXO1 is a crucial determinant of the effects of duodenum-derived serotonin on bone formation We identified two key FOXO1 complexes in osteoblasts, one with the transcription factor cAMP-responsive element-binding protein 1 (CREB) and another with activating transcription factor 4 (ATF4). Under normal levels of circulating serotonin, the proliferative activity of FOXO1 was promoted by a balance between its interaction with CREB and ATF4. However, high circulating serotonin levels prevented the association of FOXO1 with CREB, resulting in suppressed osteoblast proliferation. These observations identify FOXO1 as the molecular node of an intricate transcriptional machinery that confers the signal of duodenal-derived serotonin to inhibit bone formation.
- Published
- 2012
355. Optochemogenetics (OCG) allows more precise control of genetic engineering in mice with CreER regulators
- Author
-
Claudio Vinegoni, Xin Lu, Ronald A. DePinho, Sarit S. Agasti, Ralph Weissleder, and Peter Waterman
- Subjects
ved/biology.organism_classification_rank.species ,Cell ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,Computational biology ,Cellular level ,Biology ,Article ,Mass Spectrometry ,Mice ,medicine ,Animals ,Model organism ,Gene ,Chromatography, High Pressure Liquid ,Pharmacology ,Genetics ,ved/biology ,Extramural ,Organic Chemistry ,Tamoxifen ,medicine.anatomical_structure ,Spectrophotometry, Ultraviolet ,Genetic Engineering ,Biotechnology - Abstract
New approaches that allow precise spatiotemporal control of gene expression in model organisms at the single cell level are necessary to better dissect the role of specific genes and cell populations in development, disease, and therapy. Here, we describe a new optochemogenetic switch (OCG switch) to control CreER/loxP-mediated recombination via photoactivatable ("caged") tamoxifen analogues in individual cells in cell culture, organoid culture, and in vivo in adult mice. This approach opens opportunities to more fully exploit existing CreER transgenic mouse strains to achieve more precise temporal- and location-specific regulation of genetic events and gene expression.
- Published
- 2012
356. Abstract IA16: Kras-directed anabolic processes in PDAC tumor maintenance
- Author
-
Ronald A. DePinho
- Subjects
endocrine system diseases ,Cancer ,Tumor initiation ,Biology ,Pentose phosphate pathway ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,Transcriptome ,Biochemistry ,Pancreatic cancer ,medicine ,Cancer research ,Glycolysis ,KRAS ,Biogenesis - Abstract
Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context-dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. However, its function to maintain pancreatic cancer has not been characterized in vivo. Here, an inducible KrasG12D-driven PDAC mouse model establishes that advanced PDAC remain strictly dependent on KrasG12D expression. Transcriptome and metabolomic analysis indicate that KrasG12D serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that KrasG12D drives glycolysis intermediates into the nonoxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC. Citation Format: Ronald A. DePinho. Kras-directed anabolic processes in PDAC tumor maintenance. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Progress and Challenges; Jun 18-21, 2012; Lake Tahoe, NV. Philadelphia (PA): AACR; Cancer Res 2012;72(12 Suppl):Abstract nr IA16.
- Published
- 2012
- Full Text
- View/download PDF
357. Abstract A101: Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism
- Author
-
Sujun Hua, Jonathan L. Coloff, Haoqiang Ying, Alexander R. Guimaraes, Hongwu Zheng, Jason W. Locasale, Ronald A. DePinho, Eric S. Martin, Yonghong Xiao, Ralph Weissleder, Samuel R. Perry, Y. Alan Wang, Gerald C. Chu, Jeffery Chang, John M. Asara, Lynda Chin, Aram F. Hezel, Boyi Gan, Andrea Viale, Hailei Zhang, Alec C. Kimmelman, Carol Lim, Jaekyoung Son, Wei Wang, Haiyan Yan, Eliot Fletcher-Sananikone, Shujuan Chen, Costas A. Lyssiotis, Jihye Paik, Lewis C. Cantley, and Jian Hu
- Subjects
Genetics ,Glucose uptake ,Cancer ,Tumor initiation ,Pentose phosphate pathway ,Biology ,medicine.disease ,medicine.disease_cause ,Transcriptome ,Pancreatic cancer ,Cancer research ,medicine ,Glycolysis ,KRAS - Abstract
Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context-dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. Here, an inducible KrasG12D-driven PDAC mouse model establishes that advanced PDAC remain strictly dependent on KrasG12D expression. Transcriptome and metabolomic analysis indicate that KrasG12D serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that KrasG12D drives glycolysis intermediates into the non-oxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC. Citation Format: Haoqiang Ying, Hailei Zhang, Jonathan L. Coloff, Haiyan Yan, Wei Wang, Shujuan Chen, Andrea Viale, Hongwu Zheng, Ji-hye Paik, Carol Lim, Alexander R. Guimaraes, Alec C. Kimmelman, Eric S. Martin, Jeffery Chang, Aram Hezel, Samuel R. Perry, Jian Hu, Boyi Gan, Yonghong Xiao, John M. Asara, Ralph Weissleder, Y. Alan Wang, Costas A. Lyssiotis, Lynda Chin, Lewis C. Cantley, Ronald A. DePinho, Sujun Hua, Gerald C. Chu, Eliot Fletcher-Sananikone, Jason W. Locasale, Jaekyoung Son. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Progress and Challenges; Jun 18-21, 2012; Lake Tahoe, NV. Philadelphia (PA): AACR; Cancer Res 2012;72(12 Suppl):Abstract nr A101.
- Published
- 2012
- Full Text
- View/download PDF
358. Abstract PR4: Neuroplastic changes and pain-related behavior in a transgenic mouse model of pancreatic ductal adenocarcinoma (PDAC)
- Author
-
Rachelle E. Stopczynski, Brian M. Davis, Klaus Bielefeldt, Ronald A. DePinho, Kathryn M. Albers, and Haoqiang Ying
- Subjects
Pathology ,medicine.medical_specialty ,biology ,business.industry ,Artemin ,Tropomyosin receptor kinase B ,Tropomyosin receptor kinase A ,medicine.disease ,medicine.anatomical_structure ,Nerve growth factor ,Neurotrophic factors ,Pancreatic cancer ,medicine ,Glial cell line-derived neurotrophic factor ,biology.protein ,business ,Pancreas - Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with significant morbidity and mortality. Morbidity in PDAC is partly due to the severe pain reported by patients with the disease. Tumor-nerve interactions including intrapancreatic perineural invasion, neurogenic inflammation, and neuritis are key features of pancreatic malignancies and are thought to play an important role in pancreatic cancer-related pain. Furthermore, neurotrophic factors such as nerve growth factor (NGF), glial cell line-derived neurotrophic factor (GDNF), GDNF family member artemin (Artn), and brain-derived neurotrophic factor (BDNF) have been implicated in the development of PDAC-related pain. These neurotrophic factors have been shown to produce hypersensitivity in sensory afferents and may drive the neuropathology underlying PDAC pain. Thus, we hypothesized that neurotrophic factors would be increased in the pancreas of PDAC mice as tumors develop, leading to altered pancreatic innervation and changes in pain-related behavior. Experiments were performed with transgenic mice that have pancreas-specific expression of a mutated Kras oncogene and heterozygous deletion of the p53 tumor suppressor gene (p48Cre; LSL-KRASG12D; p53lox/+). PDAC mice demonstrated a varied disease time course but generally developed multifocal pancreatic cancer by week 16 as evidenced by nodular-appearing pancreata. Obstruction of the biliary tree, tumor involvement throughout the mesentery, and metastases to the liver were also observed in some PDAC mice at more advanced stages of disease (> 25 weeks). Sex- and age-matched littermate transgenic controls were used for all experiments. Pancreas RNA was isolated from PDAC mice and controls at 16-30 weeks of age and levels of neurotrophic factor and neurotrophic factor receptor mRNA expression was measured using PCR and qRT-PCR. Indeed, expression of NGF, the NGF receptor TrkA, Artn, GDNF, the GDNF receptor GFRα1, BDNF, and the BDNF receptor TrkB were all increased in the pancreas of PDAC mice compared to controls. Immunohistochemical studies were performed to examine the density and distribution of nerve fibers in the pancreas of PDAC mice. Large nerve bundles that stained intensely with the pan-neuronal marker PGP 9.5, tyrosine hydroxylase (TH; NGF-responsive sympathetic fibers), calcitonin gene-related peptide (CGRP; sensory fibers), and growth-associated protein 43 (GAP-43; nerve sprouting) were observed in the pancreas of PDAC mice but not controls. Open-field, exploratory behavior was monitored in PDAC mice between 16 30 weeks of age. Specifically, animals were placed in Plexiglas boxes and their activity in both the horizontal plane and vertical plane was measured photoelectrically for a period of 15 minutes. PDAC mice spent less time moving horizontally and vertically, had a reduced number of reaching movements into the vertical plane, traveled less distance in the vertical plane, and made fewer movements in the vertical plane. These data indicate that significant changes in pancreatic innervation and neurotrophic factor expression occur in a transgenic mouse model of pancreatic cancer and these changes correlate with pain-related decreases in ambulatory and reaching activity. Thus, this animal model will enable us to systematically study the impact of neuroplastic changes in the PDAC microenvironment on the development of pancreatic cancer-related pain. This proffered talk is also presented as Poster A81. Citation Format: Rachelle E. Stopczynski, Kathryn M. Albers, Klaus Bielefeldt, Ronald A. DePinho, Haoqiang Ying, Brian M. Davis. Neuroplastic changes and pain-related behavior in a transgenic mouse model of pancreatic ductal adenocarcinoma (PDAC). [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Progress and Challenges; Jun 18-21, 2012; Lake Tahoe, NV. Philadelphia (PA): AACR; Cancer Res 2012;72(12 Suppl):Abstract nr PR4.
- Published
- 2012
- Full Text
- View/download PDF
359. Abstract A51: Oncogenic Kras is required for pancreatic tumor maintenance through the regulation of anabolic glucose metabolism
- Author
-
Alec C. Kimmelman, Ronald A. DePinho, Lewis C. Cantley, Costas A. Lyssiotis, and Haoqiang Ying
- Subjects
Glucose uptake ,Tumor initiation ,Biology ,Pentose phosphate pathway ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,Transcriptome ,Biochemistry ,Pancreatic tumor ,Pancreatic cancer ,medicine ,Cancer research ,Glycolysis ,KRAS - Abstract
Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context-dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. Here, an inducible KrasG12D-driven PDAC mouse model establishes that advanced PDAC remain strictly dependent on KrasG12D expression. Transcriptome and metabolomic analysis indicate that KrasG12D serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that KrasG12D drives glycolysis intermediates into the non-oxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC. Citation Format: Costas Lyssiotis, Haoqiang Ying, Alec Kimmelman, Ronald DePinho, Lewis Cantley. Oncogenic Kras is required for pancreatic tumor maintenance through the regulation of anabolic glucose metabolism. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Progress and Challenges; Jun 18-21, 2012; Lake Tahoe, NV. Philadelphia (PA): AACR; Cancer Res 2012;72(12 Suppl):Abstract nr A51.
- Published
- 2012
- Full Text
- View/download PDF
360. STAR RNA-binding protein Quaking suppresses cancer via stabilization of specific miRNA
- Author
-
Richard M. Mortensen, Jessica A. Hurt, Y. Alan Wang, Pamela A. Silver, Shobha Vasudevan, Jihye Paik, Lynda Chin, Ronald A. DePinho, Caroline Dong, Jian Hu, An Jou Chen, Natalie G. Farny, Haoqiang Ying, Baoli Hu, Sachet A. Shukla, Yonghong Xiao, and Hailei Zhang
- Subjects
In silico ,RNA Stability ,Epistasis and functional genomics ,RNA-binding protein ,Biology ,law.invention ,Cell Line ,Mice ,law ,Gene expression ,microRNA ,Genetics ,Animals ,Humans ,RNA ,RNA-Binding Proteins ,Molecular biology ,Cell biology ,MicroRNAs ,Suppressor ,Signal transduction ,Tumor Suppressor Protein p53 ,Glioblastoma ,Developmental Biology ,Signal Transduction ,Research Paper - Abstract
Multidimensional cancer genome analysis and validation has defined Quaking (QKI), a member of the signal transduction and activation of RNA (STAR) family of RNA-binding proteins, as a novel glioblastoma multiforme (GBM) tumor suppressor. Here, we establish that p53 directly regulates QKI gene expression, and QKI protein associates with and leads to the stabilization of miR-20a; miR-20a, in turn, regulates TGFβR2 and the TGFβ signaling network. This pathway circuitry is substantiated by in silico epistasis analysis of its components in the human GBM TCGA (The Cancer Genome Atlas Project) collection and by their gain- and loss-of-function interactions in in vitro and in vivo complementation studies. This p53–QKI–miR-20a–TGFβ pathway expands our understanding of the p53 tumor suppression network in cancer and reveals a novel tumor suppression mechanism involving regulation of specific cancer-relevant microRNAs.
- Published
- 2012
361. Distinct requirements for Sin3a in perinatal male gonocytes and differentiating spermatogonia
- Author
-
Jan Hermen Dannenberg, Ronald A. DePinho, Christopher J. Payne, Robert E. Braun, Jessica M. Huszar, Amber E. Kofman, and Shannon J. Gallagher
- Subjects
Male ,Cellular differentiation ,Apoptosis ,Histones ,Mice ,0302 clinical medicine ,Testis ,Transcriptional regulation ,Nuclear Receptor Subfamily 4, Group A, Member 1 ,Phosphorylation ,Cell proliferation ,Regulation of gene expression ,0303 health sciences ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Cell cycle ,Sertoli cell ,Immunohistochemistry ,Cell biology ,Sin3 Histone Deacetylase and Corepressor Complex ,medicine.anatomical_structure ,Differentiation ,Gene Targeting ,Chromatin Immunoprecipitation ,Biology ,Real-Time Polymerase Chain Reaction ,Article ,03 medical and health sciences ,Histone H3 ,Gonocyte ,medicine ,Animals ,Cell Lineage ,Mitosis ,Molecular Biology ,030304 developmental biology ,Adaptor Proteins, Signal Transducing ,DNA Primers ,Gene Expression Profiling ,Proteins ,Cell Biology ,Sin3a ,Microarray Analysis ,Molecular biology ,Spermatogonia ,Repressor Proteins ,Germ Cells ,030217 neurology & neurosurgery ,Developmental Biology ,DNA Damage - Abstract
Chromatin modifier Swi-independent 3a (SIN3A), together with associated histone deacetylases, influences gene expression during development and differentiation through a variety of transcription factors in a cell-specific manner. Sin3a is essential for the maintenance of inner cell mass cells of mouse blastocysts, embryonic fibroblasts, and myoblasts, but is not required for the survival of trophectoderm or Sertoli cells. To better understand how this transcriptional regulator modulates cells at different developmental stages within a single lineage, we used conditional gene targeting in mice to ablate Sin3a from perinatal quiescent male gonocytes and from postnatal differentiating spermatogonia. Mitotic germ cells expressing stimulated by retinoic acid gene 8 (Stra8) that lacked Sin3a exhibited increased DNA damage and apoptosis, yet collectively progressed through meiosis and spermiogenesis and generated epididymal sperm at approximately 50% of control levels, sufficient for normal fertility. In contrast, perinatal gonocytes lacking Sin3a underwent rapid depletion that coincided with cell cycle reentry, exhibiting 2.5-fold increased histone H3 phosphorylation upon cycling that suggested a prophase/metaphase block; germ cells were almost entirely absent two weeks after birth, resulting in sterility. Gene expression profiling of neonatal testes containing Sin3a-deleted gonocytes identified upregulated transcripts highly associated with developmental processes and pattern formation, and downregulated transcripts involved in nuclear receptor activity, including Nr4a1 (Nur77). Interestingly, Nr4a1 levels were elevated in testes containing Stra8-expressing, Sin3a-deleted spermatogonia. SIN3A directly binds to the Nr4a1 promoter, and Nr4a1 expression is diminished upon spermatogonial differentiation in vitro. We conclude that within the male germline, Sin3a is required for the mitotic reentry of gonocytes, but is dispensable for the maintenance of differentiating spermatogonia and subsequent spermatogenic processes.
- Published
- 2012
362. Telomeres and Mitochondria in the Aging Heart
- Author
-
Ronald A. DePinho, Ergiin Sahin, and Javid Moslehi
- Subjects
Senescence ,Telomerase ,medicine.medical_specialty ,Aging ,Physiology ,DNA damage ,Oxidative phosphorylation ,Mitochondrion ,Biology ,Article ,Mitochondria, Heart ,Internal medicine ,medicine ,Animals ,Humans ,Heart metabolism ,Heat-Shock Proteins ,Telomere Shortening ,Age Factors ,RNA-Binding Proteins ,Telomere ,Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha ,Endocrinology ,Mitochondrial biogenesis ,Cardiovascular Diseases ,Tumor Suppressor Protein p53 ,Cardiology and Cardiovascular Medicine ,Carrier Proteins ,Energy Metabolism ,Signal Transduction ,Transcription Factors - Abstract
Studies in humans and in mice have highlighted the importance of short telomeres and impaired mitochondrial function in driving age-related functional decline in the heart. Although telomere and mitochondrial dysfunction have been viewed mainly in isolation, recent studies in telomerase-deficient mice have provided evidence for an intimate link between these two processes. Telomere dysfunction induces a profound p53-dependent repression of the master regulators of mitochondrial biogenesis and function, peroxisome proliferator-activated receptor gamma coactivator (PGC)-1α and PGC-1β in the heart, which leads to bioenergetic compromise due to impaired oxidative phosphorylation and ATP generation. This telomere-p53-PGC mitochondrial/metabolic axis integrates many factors linked to heart aging including increased DNA damage, p53 activation, mitochondrial, and metabolic dysfunction and provides a molecular basis of how dysfunctional telomeres can compromise cardiomyocytes and stem cell compartments in the heart to precipitate cardiac aging.
- Published
- 2012
363. Emerging insights into the molecular and cellular basis of glioblastoma
- Author
-
Alan Wang, Webster K. Cavenee, Benito Campos, Cameron Brennan, Pankaj K. Agarwalla, Mikael L. Rinne, Lynda Chin, Frank B. Furnari, Jill Wykosky, William C. Hahn, Gavin P. Dunn, Ian F. Dunn, Keith L. Ligon, Giannicola Genovese, Ronald A. DePinho, Steven N. Quayle, and Milan G. Chheda
- Subjects
Cellular basis ,IDH1 ,Transcription, Genetic ,Malignant brain tumor ,Genomics ,Computational biology ,Disease ,Review ,Biology ,Bioinformatics ,Genetics ,medicine ,Humans ,Genes, Tumor Suppressor ,neoplasms ,Neovascularization, Pathologic ,Brain Neoplasms ,Gene Expression Profiling ,Biological classification ,medicine.disease ,nervous system diseases ,Gene expression profiling ,Glioblastoma ,Developmental Biology - Abstract
Glioblastoma is both the most common and lethal primary malignant brain tumor. Extensive multiplatform genomic characterization has provided a higher-resolution picture of the molecular alterations underlying this disease. These comprehensive data sets give a clearer picture of glioblastoma as a heterogeneous collection of distinct diseases. In this review by Hahn and colleagues, the recent efforts to classify glioblastoma based on these new data and novel insights into the molecular pathways regulating glioblastoma are discussed.
- Published
- 2012
364. Suppression of Myc, but not E1a, transformation activity by Max-associated proteins, Mad and Mxi1
- Author
-
Enrique Gomez Lahoz, Lin Xu, Nicole Schreiber-Agus, and Ronald A. DePinho
- Subjects
Leucine zipper ,animal structures ,Genes, myc ,Gene Expression ,Repressor ,In Vitro Techniques ,Biology ,DNA-binding protein ,Structure-Activity Relationship ,Transactivation ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Genes, Tumor Suppressor ,RNA, Messenger ,Binding site ,Transcription factor ,Leucine Zippers ,Multidisciplinary ,Basic helix-loop-helix ,Tumor Suppressor Proteins ,Helix-Loop-Helix Motifs ,Molecular biology ,Rats ,DNA-Binding Proteins ,Repressor Proteins ,DNA binding site ,Cell Transformation, Neoplastic ,Mutagenesis, Site-Directed ,Adenovirus E1A Proteins ,Transcription Factors ,Research Article - Abstract
Mad and Mxi1, two members of the Myc-related basic-region helix-loop-helix/leucine-zipper family of proteins, associate directly with Max to form sequence-specific DNA binding heterodimers that are transactivation-incompetent. Mad-Max complexes have been shown to exert a strong repressive effect on Myc-induced transactivation, perhaps through the competitive occupation of common promoter binding sites also recognized by active Myc-Max heterodimers. To place these recent biochemical observations in a biological context, mad and mxi1 expression vectors were tested for their ability to influence Myc transformation activity in the rat embryo fibroblast cooperation assay. Addition of an equimolar amount of mad or mxi1 expression vector to mouse c-myc/ras cotransfections resulted in a dramatic reduction in both the number of foci generated and the severity of the malignant phenotype. Myc-specific suppression by Mad and Mxi1 was demonstrated by their ability to affect c- and N-myc-, but not ela-, induced transformation. In contrast, mad and mxi1 expression constructs bearing deletions in the basic region exerted only mild repressive effects on Myc transformation activity, suggesting that occupation of common DNA binding sites by transactivation-incompetent Mad-Max or Mxi1-Max complexes appears to play a more dominant role in this suppression than titration of limited intracellular pools of Max away from active Myc-Max complexes. Thus, these biological data support a current model for regulation of Myc function in which relative intracellular levels of Mad and Mxi1 in comparison to those of Myc may determine the degree of activation of Myc-responsive growth pathways.
- Published
- 1994
- Full Text
- View/download PDF
365. Telomerase extracurricular activities
- Author
-
Sandy Chang and Ronald A. DePinho
- Subjects
Telomerase ,Multidisciplinary ,Cell division ,biology ,Cell growth ,DNA polymerase ,Reverse transcriptase ,Homologous Recombination Pathway ,Telomere ,chemistry.chemical_compound ,chemistry ,Cancer research ,biology.protein ,DNA - Abstract
The extended growth potential of cancer cells is critically dependent upon the maintenance of functional telomeres, G-rich repeat sequences that cap the ends of most eukaryotic chromosomes and serve to protect natural DNA ends from being recognized as double-stranded breaks (reviewed in ref. 1). The inability of DNA polymerase to synthesize fully the terminal ends of the lagging strand leads to progressive telomere shortening with each round of cell division. The gradual erosion of telomeres to a critically short length elicits the successive cellular responses of senescence and crisis; each has been shown to represent formidable barriers to continued cell growth in culture (2, 3). These cell culture-based studies also seeded the view that long-term cancer cell growth and survival requires activation of one of two known mechanisms of telomere maintenance. The first and most common mechanism involves reactivation of the enzyme telomerase (4), a specialized ribonucleoprotein complex that contains a complementary RNA template (TERC) and a reverse transcriptase catalytic subunit (TERT). In telomerase reactivation, transcriptional up-regulation of the TERT gene is often the limiting event (5, 6), although TERT activity can be controlled on multiple posttranscriptional and posttranslational levels (7). The second telomere maintenance mechanism, encountered in only a minority of cancer cells, involves a telomerase-independent process termed ALT (for alternative lengthening of telomeres), which is, perhaps, mediated by the homologous recombination pathway (8, 9). The consistent presence of either mechanism in advanced human cancers has supported the assumption that the key, and perhaps only, factor in the promotion of full malignant transformation is adequate telomere reserves, and that the particular telomere maintenance mechanism used was less relevant. In this issue of PNAS, Stewart et al. (10) force a re-evaluation of this commonly held view with a provocative set of experiments showing that the actions of …
- Published
- 2002
- Full Text
- View/download PDF
366. Keeping telomerase in its place
- Author
-
Ronald A. DePinho and Richard S. Maser
- Subjects
Telomerase ,DNA damage ,Nucleolus ,DNA replication ,Cancer ,General Medicine ,Cell cycle ,Biology ,medicine.disease ,DNA-binding protein ,General Biochemistry, Genetics and Molecular Biology ,Telomere ,Cell biology ,medicine - Abstract
A new study shows that TERT, a component of telomerase, shuttles between nuclear compartments during the cell cycle. TERT localization is disrupted in cancer and following ionizing radiation, perhaps affecting genome stability.
- Published
- 2002
- Full Text
- View/download PDF
367. Prognostic determinants in prostate cancer
- Author
-
Massimo Loda, Ronald A. DePinho, Neil E. Martin, and Lorelei A. Mucci
- Subjects
PCA3 ,Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,MEDLINE ,Disease ,Article ,Prostate cancer ,Prostate ,Internal medicine ,medicine ,Animals ,Humans ,Pathological ,Neoplasm Staging ,business.industry ,Molecular pathogenesis ,Prostatic Neoplasms ,Prostate-Specific Antigen ,medicine.disease ,Prognosis ,Immunohistochemistry ,Prostate-specific antigen ,medicine.anatomical_structure ,business - Abstract
Clinical outcomes in prostate cancer are heterogeneous, and given the high prevalence of the disease, there is a pressing need to identify clinically useful markers of prognosis. Many clinical, pathological, molecular, and genetic factors have been investigated in this capacity, although relatively few are routinely used. With a growing understanding of the molecular pathogenesis of prostate cancer, there is the potential that the next generation of makers will prove sufficiently robust to guide the optimal management of men with prostate cancer. Here, we review the various clinical and molecular prognostic determinants in prostate cancer.
- Published
- 2011
368. Hepatic suppression of Foxo1 and Foxo3 causes hypoglycemia and hyperlipidemia in mice
- Author
-
Boyi Gan, Ronald A. DePinho, Shaodong Guo, Xiaoping Zhu, Kebin Zhang, Travis Averitt, Yajuan Qi, and Ling Li
- Subjects
Blood Glucose ,medicine.medical_specialty ,medicine.medical_treatment ,FOXO1 ,Cell Cycle Proteins ,Hyperlipidemias ,Hypoglycemia ,Carbohydrate metabolism ,Biology ,chemistry.chemical_compound ,Mice ,Endocrinology ,Mice, Inbred NOD ,Internal medicine ,medicine ,Animals ,Homeostasis ,Mice, Knockout ,Cholesterol ,Forkhead Box Protein O1 ,Insulin ,Forkhead Box Protein O3 ,Gluconeogenesis ,Diabetes-Insulin-Glucagon-Gastrointestinal ,Forkhead Transcription Factors ,medicine.disease ,Lipid Metabolism ,Glucose ,chemistry ,Gene Expression Regulation ,Liver ,FOXO4 ,FOXO3 - Abstract
Dysregulation of blood glucose and triglycerides are the major characteristics of type 2 diabetes mellitus. We sought to identify the mechanisms regulating blood glucose and lipid homeostasis. Cell-based studies established that the Foxo forkhead transcription factors Forkhead box O (Foxo)-1, Foxo3, and Foxo4 are inactivated by insulin via a phosphatidylinositol 3-kinase/Akt-dependent pathway, but the role of Foxo transcription factors in the liver in regulating nutrient metabolism is incompletely understood. In this study, we used the Cre/LoxP genetic approach to delete the Foxo1, Foxo3, and Foxo4 genes individually or a combination of two or all in the liver of lean or db/db mice and assessed the role of Foxo inactivation in regulating glucose and lipid homeostasis in vivo. In the lean mice or db/db mice, hepatic deletion of Foxo1, rather than Foxo3 or Foxo4, caused a modest reduction in blood glucose concentrations and barely affected lipid homeostasis. Combined deletion of Foxo1 and Foxo3 decreased blood glucose levels, elevated serum triglyceride and cholesterol concentrations, and increased hepatic lipid secretion and caused hepatosteatosis. Analysis of the liver transcripts established a prominent role of Foxo1 in regulating gene expression of gluconeogenic enzymes and Foxo3 in the expression of lipogenic enzymes. Our findings indicate that Foxo1 and Foxo3 inactivation serves as a potential mechanism by which insulin reduces hepatic glucose production and increases hepatic lipid synthesis and secretion in healthy and diabetic states.
- Published
- 2011
369. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism
- Author
-
Alexander R. Guimaraes, Hongwu Zheng, Jihye Paik, Y. Alan Wang, Aram F. Hezel, Jason W. Locasale, Lewis C. Cantley, Boyi Gan, Gerald C. Chu, Jaekyoung Son, Wei Wang, Haiyan Yan, Sujun Hua, Jonathan L. Coloff, Carol Lim, Yonghong Xiao, Ronald A. DePinho, Ralph Weissleder, Alec C. Kimmelman, Eric S. Martin, Eliot Fletcher-Sananikone, Andrea Viale, Jeffery Chang, Jian Hu, Hailei Zhang, Lynda Chin, Shujuan Chen, Costas A. Lyssiotis, Haoqiang Ying, John M. Asara, and Samuel R. Perry
- Subjects
endocrine system diseases ,Transcription, Genetic ,Tumor initiation ,Pentose phosphate pathway ,Biology ,Adenocarcinoma ,medicine.disease_cause ,General Biochemistry, Genetics and Molecular Biology ,Article ,Transcriptome ,Proto-Oncogene Proteins p21(ras) ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Pancreatic cancer ,Ribose ,medicine ,Animals ,Humans ,Glycolysis ,030304 developmental biology ,0303 health sciences ,Oncogene ,Biochemistry, Genetics and Molecular Biology(all) ,medicine.disease ,digestive system diseases ,Pancreatic Neoplasms ,Disease Models, Animal ,chemistry ,030220 oncology & carcinogenesis ,Cancer research ,KRAS - Abstract
Tumor maintenance relies on continued activity of driver oncogenes, although their rate-limiting role is highly context dependent. Oncogenic Kras mutation is the signature event in pancreatic ductal adenocarcinoma (PDAC), serving a critical role in tumor initiation. Here, an inducible Kras(G12D)-driven PDAC mouse model establishes that advanced PDAC remains strictly dependent on Kras(G12D) expression. Transcriptome and metabolomic analyses indicate that Kras(G12D) serves a vital role in controlling tumor metabolism through stimulation of glucose uptake and channeling of glucose intermediates into the hexosamine biosynthesis and pentose phosphate pathways (PPP). These studies also reveal that oncogenic Kras promotes ribose biogenesis. Unlike canonical models, we demonstrate that Kras(G12D) drives glycolysis intermediates into the nonoxidative PPP, thereby decoupling ribose biogenesis from NADP/NADPH-mediated redox control. Together, this work provides in vivo mechanistic insights into how oncogenic Kras promotes metabolic reprogramming in native tumors and illuminates potential metabolic targets that can be exploited for therapeutic benefit in PDAC.
- Published
- 2011
370. AKT/FOXO Signaling Enforces Reversible Differentiation Blockade in Myeloid Leukemias
- Author
-
Stephen M, Sykes, Steven W, Lane, Lars, Bullinger, Demetrios, Kalaitzidis, Rushdia, Yusuf, Borja, Saez, Francesca, Ferraro, Francois, Mercier, Harshabad, Singh, Kristina M, Brumme, Sanket S, Acharya, Claudia, Scholl, Claudia, Schöll, Zuzana, Tothova, Eyal C, Attar, Stefan, Fröhling, Ronald A, DePinho, Scott A, Armstrong, D Gary, Gilliland, and David T, Scadden
- Subjects
endocrine system ,Myeloid ,Cellular differentiation ,Cell ,Antigens, CD34 ,Apoptosis ,Bone Marrow Cells ,FOXO1 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Cell Line, Tumor ,hemic and lymphatic diseases ,medicine ,Reversible differentiation ,Animals ,Humans ,Protein kinase B ,Cells, Cultured ,PI3K/AKT/mTOR pathway ,030304 developmental biology ,0303 health sciences ,Biochemistry, Genetics and Molecular Biology(all) ,Forkhead Box Protein O3 ,fungi ,JNK Mitogen-Activated Protein Kinases ,Myeloid leukemia ,Cell Differentiation ,Forkhead Transcription Factors ,medicine.disease ,Blockade ,Disease Models, Animal ,Leukemia ,medicine.anatomical_structure ,Leukemia, Myeloid ,030220 oncology & carcinogenesis ,embryonic structures ,Neoplastic Stem Cells ,Cancer research ,Proto-Oncogene Proteins c-akt ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
Summary AKT activation is associated with many malignancies, where AKT acts, in part, by inhibiting FOXO tumor suppressors. We show a converse role for AKT/FOXOs in acute myeloid leukemia (AML). Rather than decreased FOXO activity, we observed that FOXOs are active in ∼40% of AML patient samples regardless of genetic subtype. We also observe this activity in human MLL-AF9 leukemia allele-induced AML in mice, where either activation of Akt or compound deletion of FoxO1/3/4 reduced leukemic cell growth, with the latter markedly diminishing leukemia-initiating cell (LIC) function in vivo and improving animal survival. FOXO inhibition resulted in myeloid maturation and subsequent AML cell death. FOXO activation inversely correlated with JNK/c-JUN signaling, and leukemic cells resistant to FOXO inhibition responded to JNK inhibition. These data reveal a molecular role for AKT/FOXO and JNK/c-JUN in maintaining a differentiation blockade that can be targeted to inhibit leukemias with a range of genetic lesions. PaperClip
- Published
- 2011
371. Cancer drug discovery faces the FACT
- Author
-
Giulio Draetta and Ronald A. DePinho
- Subjects
business.industry ,Extramural ,Translational medicine ,High Mobility Group Proteins ,NF-kappa B ,Cancer ,Tumor cells ,Antineoplastic Agents ,General Medicine ,Computational biology ,Pharmacology ,medicine.disease ,Article ,Cancer drug discovery ,DNA-Binding Proteins ,Medicine ,Animals ,Humans ,Transcriptional Elongation Factors ,Tumor Suppressor Protein p53 ,business - Abstract
Effective eradication of cancer requires treatment directed against multiple targets. The p53 and nuclear factor κB (NF-κB) pathways are dysregulated in nearly all tumors, making them attractive targets for therapeutic activation and inhibition, respectively. We have isolated and structurally optimized small molecules, curaxins, that simultaneously activate p53 and inhibit NF-κB without causing detectable genotoxicity. Curaxins demonstrated anticancer activity against all tested human tumor xenografts grown in mice. We report here that the effects of curaxins on p53 and NF-κB, as well as their toxicity to cancer cells, result from “chromatin trapping” of the FACT (facilitates chromatin transcription) complex. This FACT inaccessibility leads to phosphorylation of the p53 Ser392 by casein kinase 2 and inhibition of NF-κB–dependent transcription, which requires FACT activity at the elongation stage. These results identify FACT as a prospective anticancer target enabling simultaneous modulation of several pathways frequently dysregulated in cancer without induction of DNA damage. Curaxins have the potential to be developed into effective and safe anticancer drugs.
- Published
- 2011
372. Crosstalk between NOTCH and AKT signaling during murine megakaryocyte lineage specification
- Author
-
Philippe Rameau, Cristina Lo Celso, D. Gary Gilliland, David T. Scadden, Vinciane Mabialah, Zuzana Tothova, Tulasi Khandan, Thomas Mercher, Stephen M. Sykes, Jon C. Aster, Ronald A. DePinho, Cécile K. Lopez, Melanie G. Cornejo, and Paola Rivera-Munoz
- Subjects
Hematopoiesis and Stem Cells ,Immunology ,Notch signaling pathway ,Mice, Transgenic ,Biology ,Cell fate determination ,Biochemistry ,Thrombopoiesis ,Mice ,medicine ,Animals ,Cell Lineage ,PI3K/AKT/mTOR pathway ,Cells, Cultured ,Megakaryopoiesis ,Receptors, Notch ,Hematopoietic stem cell differentiation ,PTEN Phosphohydrolase ,Hematopoietic stem cell ,Cell Differentiation ,Forkhead Transcription Factors ,Cell Biology ,Hematology ,Receptor Cross-Talk ,Cell biology ,Mice, Inbred C57BL ,Oncogene Protein v-akt ,medicine.anatomical_structure ,Notch proteins ,Stem cell ,Megakaryocytes ,Signal Transduction - Abstract
The NOTCH signaling pathway is implicated in a broad range of developmental processes, including cell fate decisions. However, the molecular basis for its role at the different steps of stem cell lineage commitment is unclear. We recently identified the NOTCH signaling pathway as a positive regulator of megakaryocyte lineage specification during hematopoiesis, but the developmental pathways that allow hematopoietic stem cell differentiation into the erythro-megakaryocytic lineages remain controversial. Here, we investigated the role of downstream mediators of NOTCH during megakaryopoiesis and report crosstalk between the NOTCH and PI3K/AKT pathways. We demonstrate the inhibitory role of phosphatase with tensin homolog and Forkhead Box class O factors on megakaryopoiesis in vivo. Finally, our data annotate developmental mechanisms in the hematopoietic system that enable a decision to be made either at the hematopoietic stem cell or the committed progenitor level to commit to the megakaryocyte lineage, supporting the existence of 2 distinct developmental pathways.
- Published
- 2011
373. Proinvasion Metastasis Drivers in Early-Stage Melanoma Are Oncogenes
- Author
-
Sabin Dhakal, Gerald C. Chu, Jason A. Hanna, Tamar Feinberg, Lisa A. Cameron, Chang-Jiun Wu, Scott R. Granter, Rhamy Zeid, Ronald A. DePinho, Cristina W. Nogueira, Lynda Chin, Yonghong Xiao, Timothy P. Heffernan, David L. Rimm, Kenneth L. Scott, George F. Vande Woude, Chengyin Min, Samuel R. Perry, Remco van Doorn, Marcus Bosenberg, Minjung Kim, and Mariela Jaskelioff
- Subjects
Cancer Research ,Skin Neoplasms ,Acid Phosphatase ,Kaplan-Meier Estimate ,Biology ,Bioinformatics ,Article ,Metastasis ,Evolution, Molecular ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Cell Lineage ,Neoplasm Invasiveness ,Neoplasm Metastasis ,Phosphorylation ,Gene ,Melanoma ,Conserved Sequence ,Neoplasm Staging ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Genome ,Tartrate-Resistant Acid Phosphatase ,Gene Expression Profiling ,Reproducibility of Results ,Oncogenes ,Cell Biology ,medicine.disease ,Primary tumor ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Isoenzymes ,Oncology ,Tissue Array Analysis ,Genetically Engineered Mouse ,030220 oncology & carcinogenesis ,Cancer research ,Genetic screen - Abstract
SummaryClinical and genomic evidence suggests that the metastatic potential of a primary tumor may be dictated by prometastatic events that have additional oncogenic capability. To test this “deterministic” hypothesis, we adopted a comparative oncogenomics-guided function-based strategy involving: (1) comparison of global transcriptomes of two genetically engineered mouse models with contrasting metastatic potential, (2) genomic and transcriptomic profiles of human melanoma, (3) functional genetic screen for enhancers of cell invasion, and (4) evidence of expression selection in human melanoma tissues. This integrated effort identified six genes that are potently proinvasive and oncogenic. Furthermore, we show that one such gene, ACP5, confers spontaneous metastasis in vivo, engages a key pathway governing metastasis, and is prognostic in human primary melanomas.
- Published
- 2011
374. Hepatic FoxOs Regulate Lipid Metabolism via Modulation of Expression of the Nicotinamide Phosphoribosyltransferase Gene*
- Author
-
Rongya Tao, Hanlin Gao, Dan Wei, Ronald A. DePinho, Yunlong Liu, and X. Charlie Dong
- Subjects
Nicotinamide phosphoribosyltransferase ,Mice, Transgenic ,Biology ,Biochemistry ,Gene Expression Regulation, Enzymologic ,chemistry.chemical_compound ,NAMPT Gene ,Mice ,Animals ,Humans ,Insulin ,Nicotinamide Phosphoribosyltransferase ,Molecular Biology ,Triglycerides ,Regulation of gene expression ,Gene knockdown ,Forkhead Box Protein O1 ,Fatty Acids ,FOXO Family ,Lipid metabolism ,Forkhead Transcription Factors ,Cell Biology ,Triglyceride homeostasis ,Lipid Metabolism ,NAD ,Lipids ,Cell biology ,Metabolism ,chemistry ,Gene Expression Regulation ,Liver ,NAD+ kinase - Abstract
FoxO transcription factors have been implicated in lipid metabolism; however, the underlying mechanisms are not well understood. Here, in an effort to elucidate such mechanisms, we examined the phenotypic consequences of liver-specific deletion of three members of the FoxO family: FoxO1, FoxO3, and FoxO4. These liver-specific triply null mice, designated LTKO, exhibited elevated triglycerides in the liver on regular chow diet. More remarkably, LTKO mice developed severe hepatic steatosis following placement on a high fat diet. Further analyses revealed that hepatic NAD(+) levels and Sirt1 activity were decreased in the liver of the LTKO mice relative to controls. At the mechanistic level, expression profile analyses showed that LTKO livers had significantly down-regulated expression of the nicotinamide phosphoribosyltransferase (Nampt) gene encoding the rate-limiting enzyme in the salvage pathway of NAD(+) biosynthesis. Luciferase reporter assays and chromatin immunoprecipitation analyses demonstrated that Nampt is a transcriptional target gene of FoxOs. Significantly, overexpression of Nampt gene reduced, whereas knockdown increased, hepatic triglyceride levels in vitro and in vivo. Thus, FoxOs control the Nampt gene expression and the NAD(+) signaling in the regulation of hepatic triglyceride homeostasis.
- Published
- 2011
375. Correction: Targeting EGFR Induced Oxidative Stress by PARP1 Inhibition in Glioblastoma Therapy
- Author
-
Alan D. D'Andrea, Jayne M. Stommel, Santosh Kesari, Katherine A. Hoadley, Richard E. Kennedy, Webster K. Cavenee, Masayuki Nitta, Pascal O. Zinn, Frank B. Furnari, David Kozono, Clark C. Chen, Lynda Chin, Deepa Kushwaha, Kimberly Ng, and Ronald A. DePinho
- Subjects
Multidisciplinary ,business.industry ,Science ,Section (typography) ,lcsh:R ,Correction ,lcsh:Medicine ,Pascal (programming language) ,medicine.disease ,Medicine ,lcsh:Q ,Analysis tools ,business ,lcsh:Science ,computer ,Classics ,computer.programming_language ,Glioblastoma - Abstract
The authors would like to add Maria-del-Mar Inda and Jill Wykosky to the byline. The updated byline is: Masayuki Nitta1#, David Kozono1,2#, Richard Kennedy3, Jayne Stommel4, Kimberly Ng1, Pascal O. Zinn1, Deepa Kushwaha1, Santosh Kesari5, Maria-del-Mar Inda6, Jill Wykosky6, Frank Furnari6, Katherine A. Hoadley7, Lynda Chin4, Ronald A. DePinho4, Webster K. Cavenee6, Alan D'Andrea1, Clark C. Chen1,8* The two new authors should be added to the author contributions "Contributed reagents/materials/analysis tools" section. The section should read "Contributed reagents/materials/analysis tools: JS SK M-d-MI JW FBF LC RAD WKC ADD CCC."
- Published
- 2011
376. Zebra Fish myc Family and max Genes: Differential Expression and Oncogenic Activity throughout Vertebrate Evolution
- Author
-
Jim Horner, Ronald A. DePinho, Nicole Schreiber-Agus, Richard Torres, and Fung Chow Chiu
- Subjects
Trout ,Xenopus ,Molecular Sequence Data ,Restriction Mapping ,Genes, myc ,Gene Expression ,Protein Max ,Gene product ,Mice ,Open Reading Frames ,Sequence Homology, Nucleic Acid ,biology.animal ,Gene expression ,Animals ,Humans ,Amino Acid Sequence ,Zebrafish ,Gene ,Transcription factor ,Molecular Biology ,MYC Family Gene ,Genetics ,Base Sequence ,Sequence Homology, Amino Acid ,biology ,Basic Helix-Loop-Helix Leucine Zipper Transcription Factors ,Vertebrate ,Hominidae ,Oncogenes ,Cell Biology ,Zebrafish Proteins ,biology.organism_classification ,Biological Evolution ,DNA-Binding Proteins ,Basic-Leucine Zipper Transcription Factors ,Multigene Family ,Vertebrates ,Research Article ,Transcription Factors - Abstract
To gain insight into the role of Myc family oncoproteins and their associated protein Max in vertebrate growth and development, we sought to identify homologs in the zebra fish (Brachydanio rerio). A combination of a polymerase chain reaction-based cloning strategy and low-stringency hybridization screening allowed for the isolation of zebra fish c-, N-, and L-myc and max genes; subsequent structural characterization showed a high degree of conservation in regions that encode motifs of known functional significance. On the functional level, zebra fish Max, like its mammalian counterpart, served to suppress the transformation activity of mouse c-Myc in rat embryo fibroblasts. In addition, the zebra fish c-myc gene proved capable of cooperating with an activated H-ras to effect the malignant transformation of mammalian cells, albeit with diminished potency compared with mouse c-myc. With respect to their roles in normal developing tissues, the differential temporal and spatial patterns of steady-state mRNA expression observed for each zebra fish myc family member suggest unique functions for L-myc in early embryogenesis, for N-myc in establishment and growth of early organ systems, and for c-myc in increasingly differentiated tissues. Furthermore, significant alterations in the steady-state expression of zebra fish myc family genes concomitant with relatively constant max expression support the emerging model of regulation of Myc function in cellular growth and differentiation.
- Published
- 1993
- Full Text
- View/download PDF
377. Comparative analysis of the expression and oncogenic activities of Xenopus c-, N-, and L-myc homologs
- Author
-
Jim Horner, Milan Jamrich, Ronald A. DePinho, Anna Lau, Nicole Schreiber-Agus, and Richard Torres
- Subjects
Male ,Molecular Sequence Data ,Restriction Mapping ,Genes, myc ,Xenopus ,Gene Expression ,Sequence alignment ,In Vitro Techniques ,Biology ,Homology (biology) ,Proto-Oncogene Proteins c-myc ,Mice ,Structure-Activity Relationship ,Xenopus laevis ,Transactivation ,Gene expression ,Animals ,Humans ,Gene family ,Amino Acid Sequence ,RNA, Messenger ,Molecular Biology ,Gene ,Cells, Cultured ,Base Sequence ,Fishes ,DNA ,Cell Biology ,biology.organism_classification ,Molecular biology ,Rats ,Cell Transformation, Neoplastic ,Female ,Chickens ,Sequence Alignment ,Research Article - Abstract
A polymerase chain reaction-based cloning strategy allowed for the isolation of two distinct Xenopus L-myc genes, as well as previously isolated xc- and xN-myc genes, thus demonstrating that these three well-defined members of the mammalian myc gene family are present in lower vertebrates as well. Comparison of the Xenopus and mammalian Myc families revealed a high degree of structural relatedness at the gene and protein levels; this homology was consistent with the ability of the xc-myc1 and xN-myc1 genes to function as oncogenes in primary mammalian cells. In contrast, the xL-myc1 gene was found to be incapable of transforming rat embryo fibroblast cells, and this inactivity may relate to localized but significant differences in its putative transactivation domain. Analysis of xc-, xN-, and xL-myc gene expression demonstrated that (i) all three genes were highly expressed during oogenesis and their transcripts accumulated as abundant maternal mRNAs, (ii) each gene exhibited a distinctive pattern of expression during embryogenesis and in adult tissues, and (iii) the xL-myc1 and xL-myc2 genes were coordinately expressed in the maternal and zygotic genomes. The markedly high expression of the Xenopus myc gene family in differentiated tissues, such as the central nervous system and kidney, contrasts sharply with the low levels observed in mammalian adult tissues. These differences may reflect unique functions of the Myc family proteins in processes specific to amphibians, such as tissue regeneration.
- Published
- 1993
- Full Text
- View/download PDF
378. Cancer signaling: when phosphorylation meets methylation
- Author
-
Haoqiang Ying and Ronald A. DePinho
- Subjects
MAP kinase kinase kinase ,Cancer ,Cell Biology ,Methylation ,Biology ,medicine.disease_cause ,medicine.disease ,Histone-Lysine N-Methyltransferase ,Cell biology ,Anti-apoptotic Ras signalling cascade ,medicine ,bacteria ,Phosphorylation ,Oncogene Protein p21(ras) ,KRAS ,Molecular Biology - Abstract
The propagation of kinase-mediated phosphorylation signals is central to the oncogenic activity of the RAS-MAPK pathway in human cancers. A recent study shows that protein lysine methylation controls the phosphorylation status of a key component of the RAS-MAPK pathway to enable oncogenic KRAS in cancer progression.
- Published
- 2014
- Full Text
- View/download PDF
379. FoxOs enforce a progression checkpoint to constrain mTORC1-activated renal tumorigenesis
- Author
-
Boyi Gan, Eliot Fletcher-Sananikone, Ronald A. DePinho, Hongwu Zheng, Gerald C. Chu, Li Zhuang, Sabina Signoretti, Michelle Chang, William G. Kaelin, Shan Jiang, Sujun Hua, Zhihu Ding, David J. Kwiatkowski, Carol Lim, Michael Collins, Y. Alan Wang, and Jian Hu
- Subjects
Transcriptional Activation ,Cancer Research ,Apoptosis ,mTORC1 ,Biology ,Mechanistic Target of Rapamycin Complex 1 ,medicine.disease_cause ,urologic and male genital diseases ,Article ,Tuberous Sclerosis Complex 1 Protein ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,Renal cell carcinoma ,microRNA ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Tumor Cells, Cultured ,Animals ,Humans ,Carcinoma, Renal Cell ,030304 developmental biology ,0303 health sciences ,TOR Serine-Threonine Kinases ,Tumor Suppressor Proteins ,fungi ,Cancer ,Proteins ,Forkhead Transcription Factors ,Cell Biology ,medicine.disease ,Kidney Neoplasms ,Gene Expression Regulation, Neoplastic ,MicroRNAs ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Multiprotein Complexes ,embryonic structures ,Cancer research ,TSC1 ,biological phenomena, cell phenomena, and immunity ,Carcinogenesis ,Clear cell ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
SummarymTORC1 is a validated therapeutic target for renal cell carcinoma (RCC). Here, analysis of Tsc1-deficient (mTORC1 hyperactivation) mice uncovered a FoxO-dependent negative feedback circuit constraining mTORC1-mediated renal tumorigenesis. We document robust FoxO activation in Tsc1-deficient benign polycystic kidneys and FoxO extinction on progression to murine renal tumors; murine renal tumor progression on genetic deletion of both Tsc1 and FoxOs; and downregulated FoxO expression in most human renal clear cell and papillary carcinomas, yet continued expression in less aggressive RCCs and benign renal tumor subtypes. Mechanistically, integrated analyses revealed that FoxO-mediated block operates via suppression of Myc through upregulation of the Myc antagonists, Mxi1-SRα and mir-145, establishing a FoxO-Mxi1-SRα/mir-145 axis as a major progression block in renal tumor development.
- Published
- 2010
380. Insulin signaling in osteoblasts integrates bone remodeling and energy metabolism
- Author
-
Ronald A. DePinho, Anna Teti, Patricia Ducy, Gerard Karsenty, Andrea Del Fattore, Mathieu Ferron, Tatsuya Yoshizawa, and Jianwen Wei
- Subjects
musculoskeletal diseases ,medicine.medical_specialty ,medicine.medical_treatment ,Osteocalcin ,HUMDISEASE ,GPRC6A ,General Biochemistry, Genetics and Molecular Biology ,Bone resorption ,Article ,Bone remodeling ,Mice ,Internal medicine ,medicine ,Glucose homeostasis ,Animals ,Humans ,Insulin ,Cells, Cultured ,Osteoblasts ,biology ,Biochemistry, Genetics and Molecular Biology(all) ,Osteoblast ,Extracellular Matrix ,Mice, Inbred C57BL ,Insulin receptor ,Endocrinology ,medicine.anatomical_structure ,Glucose ,SIGNALING ,biology.protein ,Bone Remodeling ,Energy Metabolism ,Signal Transduction - Abstract
SummaryThe broad expression of the insulin receptor suggests that the spectrum of insulin function has not been fully described. A cell type expressing this receptor is the osteoblast, a bone-specific cell favoring glucose metabolism through a hormone, osteocalcin, that becomes active once uncarboxylated. We show here that insulin signaling in osteoblasts is necessary for whole-body glucose homeostasis because it increases osteocalcin activity. To achieve this function insulin signaling in osteoblasts takes advantage of the regulation of osteoclastic bone resorption exerted by osteoblasts. Indeed, since bone resorption occurs at a pH acidic enough to decarboxylate proteins, osteoclasts determine the carboxylation status and function of osteocalcin. Accordingly, increasing or decreasing insulin signaling in osteoblasts promotes or hampers glucose metabolism in a bone resorption-dependent manner in mice and humans. Hence, in a feed-forward loop, insulin signals in osteoblasts activate a hormone, osteocalcin, that promotes glucose metabolism.
- Published
- 2010
381. Mig-6 controls EGFR trafficking and suppresses gliomagenesis
- Author
-
Y. Alan Wang, Oreste Segatto, Hongwu Zheng, Hailei Zhang, Jian Hu, Yonghong Xiao, An Jou Chen, Ruprecht Wiedemeyer, Elena Ivanova, Michelle A. Lee, Cameron Brennan, Kenneth L. Scott, Carol Lim, Lisa A. Elferink, Haiyan Yan, Lynda Chin, Jihye Paik, Ronald A. DePinho, Haoqiang Ying, Joseph Huang, Sabin Dhakal, and Jayne M. Stommel
- Subjects
Tumor suppressor gene ,Endosome ,Biology ,Mice ,Growth factor receptor ,Cell Line, Tumor ,Two-Hybrid System Techniques ,Cell Adhesion ,Animals ,Humans ,Neoplasm Invasiveness ,Epidermal growth factor receptor ,Late endosome ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Regulation of gene expression ,Multidisciplinary ,Brain Neoplasms ,Tumor Suppressor Proteins ,Intracellular Signaling Peptides and Proteins ,Signal transducing adaptor protein ,Glioma ,Biological Sciences ,Cell biology ,ErbB Receptors ,Gene Expression Regulation, Neoplastic ,Cancer research ,biology.protein ,Cyclin-dependent kinase 8 ,Lysosomes - Abstract
Glioblastoma multiforme (GBM) is the most common and lethal primary brain cancer that is driven by aberrant signaling of growth factor receptors, particularly the epidermal growth factor receptor (EGFR). EGFR signaling is tightly regulated by receptor endocytosis and lysosome-mediated degradation, although the molecular mechanisms governing such regulation, particularly in the context of cancer, remain poorly delineated. Here, high-resolution genomic profiles of GBM identified a highly recurrent focal 1p36 deletion encompassing the putative tumor suppressor gene, Mig-6. We show that Mig-6 quells the malignant potential of GBM cells and dampens EGFR signaling by driving EGFR into late endosomes and lysosome-mediated degradation upon ligand stimulation. Mechanistically, this effect is mediated by the binding of Mig-6 to a SNARE protein STX8, a protein known to be required for late endosome trafficking. Thus, Mig-6 functions to ensure recruitment of internalized receptor to late endosomes and subsequently the lysosomal degradation compartment through its ability to specifically link EGFR and STX8 during ligand-stimulated EGFR trafficking. In GBM, the highly frequent loss of Mig-6 would therefore serve to sustain aberrant EGFR-mediated oncogenic signaling. Together, these data uncover a unique tumor suppression mechanism involving the regulation of receptor trafficking.
- Published
- 2010
382. Linking functional decline of telomeres, mitochondria and stem cells during ageing
- Author
-
Ronald A. DePinho and Ergun Sahin
- Subjects
Telomerase ,Aging ,Multidisciplinary ,Genome ,DNA damage ,Hematopoietic System ,Stem Cells ,Mitochondrion ,Biology ,Telomere ,Article ,Cell biology ,Mitochondria ,Haematopoiesis ,Phenotype ,Ageing ,Animals ,Homeostasis ,Humans ,Stem cell ,Tumor Suppressor Protein p53 - Abstract
The study of human genetic disorders and mutant mouse models has provided evidence that genome maintenance mechanisms, DNA damage signalling and metabolic regulation cooperate to drive the ageing process. In particular, age-associated telomere damage, diminution of telomere 'capping' function and associated p53 activation have emerged as prime instigators of a functional decline of tissue stem cells and of mitochondrial dysfunction that adversely affect renewal and bioenergetic support in diverse tissues. Constructing a model of how telomeres, stem cells and mitochondria interact with key molecules governing genome integrity, 'stemness' and metabolism provides a framework for how diverse factors contribute to ageing and age-related disorders.
- Published
- 2010
383. FoxO1 is a Positive Regulator of Bone Formation by Favoring Protein Synthesis and Resistance to Oxidative Stress in Osteoblasts
- Author
-
Jihye Paik, Stavroula Kousteni, Yoshihiro Yoshikawa, Lili Xu, Marie Therese Rached, Aruna Kode, and Ronald A. DePinho
- Subjects
medicine.medical_specialty ,endocrine system ,Physiology ,HUMDISEASE ,FOXO1 ,Biology ,medicine.disease_cause ,Article ,Bone remodeling ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Osteogenesis ,Internal medicine ,medicine ,Animals ,Transcription factor ,Molecular Biology ,Cells, Cultured ,030304 developmental biology ,Cell Proliferation ,0303 health sciences ,Osteoblasts ,Amino acid import ,Forkhead Box Protein O1 ,ATF4 ,FOXO Family ,Osteoblast ,Forkhead Transcription Factors ,Cell Biology ,Activating Transcription Factor 4 ,Oxidative Stress ,Endocrinology ,medicine.anatomical_structure ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Protein Biosynthesis ,Mutation ,Oxidative stress - Abstract
SummaryOsteoporosis, a disease of low bone mass, is associated with decreased osteoblast numbers and increased levels of oxidative stress within osteoblasts. Since transcription factors of the FoxO family confer stress resistance, we investigated their potential impact on skeletal integrity. Here we employ cell-specific deletion and molecular analyses to show that, among the three FoxO proteins, only FoxO1 is required for proliferation and redox balance in osteoblasts and thereby controls bone formation. FoxO1 regulation of osteoblast proliferation occurs through its interaction with ATF4, a transcription factor regulating amino acid import, as well as through its regulation of a stress-dependent pathway influencing p53 signaling. Accordingly, decreasing oxidative stress levels or increasing protein intake normalizes bone formation and bone mass in mice lacking FoxO1 specifically in osteoblasts. These results identify FoxO1 as a crucial regulator of osteoblast physiology and provide a direct mechanistic link between oxidative stress and the regulation of bone remodeling.
- Published
- 2010
384. Overlapping functions of Hdac1 and Hdac2 in cell cycle regulation and haematopoiesis
- Author
-
James W. Horner, Heinz Jacobs, Jaco Van Der Torre, Roel H. Wilting, Marinus R. Heideman, Jan Hermen Dannenberg, Eva Yanover, and Ronald A. DePinho
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,animal structures ,Histone Deacetylase 2 ,Apoptosis ,Histone Deacetylase 1 ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Mice ,Animals ,Humans ,Molecular Biology ,Cells, Cultured ,Mice, Knockout ,General Immunology and Microbiology ,Histone deacetylase 2 ,Kinase ,General Neuroscience ,Cell Cycle ,Anemia ,Cell cycle ,Thrombocytopenia ,Hematopoiesis ,Mice, Inbred C57BL ,Haematopoiesis ,enzymes and coenzymes (carbohydrates) ,Histone ,Acetylation ,embryonic structures ,Cancer research ,biology.protein ,Biocatalysis ,biological phenomena, cell phenomena, and immunity ,Tumor Suppressor Protein p53 ,G1 phase ,Deacetylase activity - Abstract
Histone deacetylases (HDACs) counterbalance acetylation of lysine residues, a protein modification involved in numerous biological processes. Here, Hdac1 and Hdac2 conditional knock-out alleles were used to study the function of class I Hdac1 and Hdac2 in cell cycle progression and haematopoietic differentiation. Combined deletion of Hdac1 and Hdac2, or inactivation of their deacetylase activity in primary or oncogenic-transformed fibroblasts, results in a senescence-like G(1) cell cycle arrest, accompanied by up-regulation of the cyclin-dependent kinase inhibitor p21(Cip). Notably, concomitant genetic inactivation of p53 or p21(Cip) indicates that Hdac1 and Hdac2 regulate p53-p21(Cip)-independent pathways critical for maintaining cell cycle progression. In vivo, we show that Hdac1 and Hdac2 are not essential for liver homeostasis. In contrast, total levels of Hdac1 and Hdac2 in the haematopoietic system are critical for erythrocyte-megakaryocyte differentiation. Dual inactivation of Hdac1 and Hdac2 results in apoptosis of megakaryocytes and thrombocytopenia. Together, these data indicate that Hdac1 and Hdac2 have overlapping functions in cell cycle regulation and haematopoiesis. In addition, this work provides insights into mechanism-based toxicities observed in patients treated with HDAC inhibitors.
- Published
- 2010
385. The FoxO3/type 2 deiodinase pathway is required for normal mouse myogenesis and muscle regeneration
- Author
-
Monica Dentice, Domenico Salvatore, P. Reed Larsen, Ronald A. DePinho, Ombretta Guardiola, Raffaele Ambrosio, Gabriella Minchiotti, Alessandro Marsili, Annarita Sibilio, Gianfranco Fenzi, and Jihye Paik
- Subjects
medicine.medical_specialty ,Molecular Sequence Data ,Deiodinase ,DIO2 ,THYROID-HORMONE ,TYPE-2 IODOTHYRONINE DEIODINASE ,Muscle Development ,MyoD ,Iodide Peroxidase ,Cell Line ,Mice ,Internal medicine ,SATELLITE CELLS ,medicine ,Animals ,Humans ,Regeneration ,Myocyte ,Muscle, Skeletal ,Mice, Knockout ,Triiodothyronine ,Base Sequence ,biology ,Myogenesis ,Stem Cells ,Forkhead Box Protein O3 ,Infant ,Skeletal muscle ,Cell Differentiation ,Forkhead Transcription Factors ,STEM-CELL FUNCTION ,General Medicine ,medicine.anatomical_structure ,Endocrinology ,biology.protein ,FOXO3 ,SKELETAL-MUSCLE ,Sequence Alignment ,Research Article - Abstract
The active thyroid hormone 3,5,3' triiodothyronine (T3) is a major regulator of skeletal muscle function. The deiodinase family of enzymes controls the tissue-specific activation and inactivation of the prohorrnone thyroxine (T4). Here we show that type 2 deiodinase (D2) is essential for normal mouse myogenesis and muscle regeneration. Indeed, D2-mediated increases in T3 were essential for the enhanced transcription of myogenic differentiation 1 (MyoD) and for execution of the myogenic program. Conversely, the expression of T3-dependent genes was reduced and after injury regeneration markedly delayed in muscles of mice null for the gene encoding D2 (Dio2), despite normal circulating T3 concentrations. Forkhead box 03 (Fox03) was identified as a key molecule inducing D2 expression and thereby increasing intracellular T3 production. Accordingly, Fox03-depleted primary myoblasts also had a differentiation deficit that could be rescued by high levels of T3. In conclusion, the Fox03/D2 pathway selectively enhances intracellular active thyroid hormone concentrations in muscle, providing a striking example of how a circulating hormone can be tissue-specifically activated to influence development locally.
- Published
- 2010
- Full Text
- View/download PDF
386. Myc family oncoproteins function through a common pathway to transform normal cells in culture: cross-interference by Max and trans-acting dominant mutants
- Author
-
Sharon D. Morgenbesser, Ronald A. DePinho, and Bhaskar Mukherjee
- Subjects
Leucine zipper ,Genes, myc ,Repressor ,Context (language use) ,Biology ,medicine.disease_cause ,Proto-Oncogene Proteins c-myc ,Cotransformation ,Genetics ,medicine ,Transcription factor ,Regulation of gene expression ,Mutation ,Blotting, Northern ,Precipitin Tests ,Molecular biology ,DNA-Binding Proteins ,Gene Expression Regulation, Neoplastic ,Basic-Leucine Zipper Transcription Factors ,Cell Transformation, Neoplastic ,Genes, ras ,Trans-acting ,Plasmids ,Transcription Factors ,Developmental Biology - Abstract
The myc family of cellular oncogenes encodes three highly related nuclear phosphoproteins (c-Myc, N-Myc, and L-Myc) that are believed to function as sequence-specific transcription factors capable of regulating genes important in cellular growth and differentiation. Current evidence indicates that Myc family proteins exist as biologically active heterodimeric complexes in association with another helix-loop-helix leucine zipper phosphoprotein, Max. We have investigated the common and unique properties among the Myc family, as well as the physiological role of Max in the regulation of Myc family function. We demonstrate that trans-activation-incompetent mutants of one Myc family member can act in trans to dominantly suppress the cotransformation activities of all three Myc oncoproteins, indicating that the Myc family functions through common genetic elements in its cellular transformation pathways. Employing co-immunoprecipitation with either anti-Myc or anti-Max antibodies, we show that the transfected normal c-Myc, N-Myc, and L-Myc oncoproteins associate with the endogenous Max protein in REF transformants, indicating that the Max interaction represents at least one component common to Myc family function. In addition, we observed a striking reduction in Myc cotransformation activity when a Max expression construct was added to myc/ras co-transfections. We discuss these biological findings in the context of a proposed model for Myc/Max function and regulation in which Max serves as either an obligate partner in the Myc/Max transcriptional complex or as a repressor in the form of a transcriptionally inert Max/Max homodimer capable of occupying Myc/Max-responsive gene targets.
- Published
- 1992
- Full Text
- View/download PDF
387. Myc and Max: a putative transcriptional complex in search of a cellular target
- Author
-
Richard Torres, Sharon D. Morgenbesser, Nicole Schreiber-Agus, and Ronald A. DePinho
- Subjects
Genetics ,Candidate gene ,Base Sequence ,Transcription, Genetic ,Cell growth ,Molecular Sequence Data ,Genetic observations ,DNA ,Cell Biology ,Biology ,Models, Biological ,Cellular protein ,Cell biology ,DNA-Binding Proteins ,Proto-Oncogene Proteins c-myc ,Transactivation ,Basic-Leucine Zipper Transcription Factors ,Transcription (biology) ,Animals ,Transcription factor ,Gene ,Transcription Factors - Abstract
Biochemical and genetic observations have supported the hypothesis that Myc family proteins function to regulate genes important in cellular growth and differentiation. The recent findings that Myc proteins can associate with other cellular proteins, possess sequence-specific DNA-binding activity and may directly transactivate transcription of several candidate genes have provided an experimental framework in which to test the transcription factor model. Based on principles established for several well characterized viral oncoproteins, a model is presented in which the regulation of Myc function is controlled by specific cellular protein interactions that serve to activate or repress transactivation activity or deny access of the Myc complex to its target sequences.
- Published
- 1992
- Full Text
- View/download PDF
388. A novel POU homeodomain gene specifically expressed in cells of the developing mammalian nervous system
- Author
-
Frederick W. Alt, Scott Mellis, Milton W. Datta, Peter E. Fisher, Carol J. Thiele, Robert G. Collum, Carlo M. Croce, Tarik Morory, Mark A. Israel, Kay Huebner, Thomas Theil, and Ronald A. DePinho
- Subjects
Transcription, Genetic ,Placenta ,Cellular differentiation ,Molecular Sequence Data ,Restriction Mapping ,Genes, myc ,Sarcoma, Ewing ,Biology ,Exon ,Pregnancy ,Neoplasms ,Gene expression ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Neuroectodermal Tumors, Primitive, Peripheral ,Cloning, Molecular ,Gene ,Peptide sequence ,Genomic Library ,Transcription Factor Brn-3A ,Chromosomes, Human, Pair 13 ,Sequence Homology, Amino Acid ,POU domain ,Genes, Homeobox ,Chromosome Mapping ,DNA ,Exons ,Chromosome Banding ,Rats ,embryonic structures ,Homeobox ,Drosophila ,Female ,DNA Probes - Abstract
We report the isolation of a novel human POU domain encoding gene named RDC-1. The POU domain of the RDC-1 encoded protein is highly related to the POU domain potentially encoded by the rat brain-3 sequence and to that of the Drosophila I-POU protein; outside of the POU region, RDC-1 is unrelated to any previously characterized protein. The RDC-1 gene is expressed almost exclusively in normal tissues and transformed cells of neural origin. In the developing mouse and human fetus, RDC-1 is expressed in a spatially and temporally restricted pattern that suggests a critical role in the differentiation of neuronal tissues. In addition, RDC-1 is expressed in a unique subset of tumors of the peripheral nervous system including neuroepitheliomas and Ewing's sarcomas but not neuroblastomas. Based on its unique structural characteristics and expression pattern, we discuss potential functions for the RDC-1 protein.
- Published
- 1992
- Full Text
- View/download PDF
389. SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction
- Author
-
Steven P. Gygi, Alejandro Gutierrez, Keiichi I. Nakayama, Lixin Wan, Jeffrey Settleman, Jon C. Aster, Ichiro Onoyama, Hiroyuki Inuzuka, Shavali Shaik, Alan Tseng, Bo Zhai, Wenyi Wei, Ronald A. DePinho, Amanda L. Christie, Thomas Look, Alan W. Lau, Darning Gao, Andrew L. Kung, Yonghong Xiao, and Richard S. Maser
- Subjects
Niacinamide ,F-Box-WD Repeat-Containing Protein 7 ,Tumor suppressor gene ,Pyridines ,Ubiquitin-Protein Ligases ,Molecular Sequence Data ,Apoptosis ,Cell Cycle Proteins ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Piperazines ,Article ,Nitrophenols ,Glycogen Synthase Kinase 3 ,Mice ,GSK-3 ,Cell Line, Tumor ,medicine ,Animals ,Humans ,MCL1 ,Amino Acid Sequence ,Phosphorylation ,Sulfonamides ,Multidisciplinary ,SKP Cullin F-Box Protein Ligases ,biology ,Apoptosis Regulator ,F-Box Proteins ,Phenylurea Compounds ,Tumor Suppressor Proteins ,Benzenesulfonates ,Biphenyl Compounds ,Ubiquitination ,food and beverages ,Cancer ,Sorafenib ,medicine.disease ,Ubiquitin ligase ,Biphenyl compound ,Proto-Oncogene Proteins c-bcl-2 ,biology.protein ,Cancer research ,Myeloid Cell Leukemia Sequence 1 Protein ,Ovarian cancer ,Protein Binding - Abstract
The effective use of targeted therapy is highly dependent on the identification of responder patient populations. Loss of FBW7, which encodes a tumour-suppressor protein, is frequently found in various types of human cancer, including breast cancer, colon cancer and T-cell acute lymphoblastic leukaemia (T-ALL). In line with these genomic data, engineered deletion of Fbw7 in mouse T cells results in T-ALL, validating FBW7 as a T-ALL tumour suppressor. Determining the precise molecular mechanisms by which FBW7 exerts antitumour activity is an area of intensive investigation. These mechanisms are thought to relate in part to FBW7-mediated destruction of key proteins relevant to cancer, including Jun, Myc, cyclin E and notch 1 (ref. 9), all of which have oncoprotein activity and are overexpressed in various human cancers, including leukaemia. In addition to accelerating cell growth, overexpression of Jun, Myc or notch 1 can also induce programmed cell death. Thus, considerable uncertainty surrounds how FBW7-deficient cells evade cell death in the setting of upregulated Jun, Myc and/or notch 1. Here we show that the E3 ubiquitin ligase SCF(FBW7) (a SKP1-cullin-1-F-box complex that contains FBW7 as the F-box protein) governs cellular apoptosis by targeting MCL1, a pro-survival BCL2 family member, for ubiquitylation and destruction in a manner that depends on phosphorylation by glycogen synthase kinase 3. Human T-ALL cell lines showed a close relationship between FBW7 loss and MCL1 overexpression. Correspondingly, T-ALL cell lines with defective FBW7 are particularly sensitive to the multi-kinase inhibitor sorafenib but resistant to the BCL2 antagonist ABT-737. On the genetic level, FBW7 reconstitution or MCL1 depletion restores sensitivity to ABT-737, establishing MCL1 as a therapeutically relevant bypass survival mechanism that enables FBW7-deficient cells to evade apoptosis. Therefore, our work provides insight into the molecular mechanism of direct tumour suppression by FBW7 and has implications for the targeted treatment of patients with FBW7-deficient T-ALL.
- Published
- 2009
390. FoxO4 Inhibits NF-κB and Protects Mice Against Colonic Injury and Inflammation
- Author
-
Ronald A. DePinho, Wen Zhou, Qian Cao, Qing Jun Zhang, Diego H. Castrillon, Zhi Ping Liu, and Yan Peng
- Subjects
CD4-Positive T-Lymphocytes ,Chemokine ,Time Factors ,Transcription, Genetic ,Colon ,medicine.medical_treatment ,Inflammation ,Cell Cycle Proteins ,Transfection ,CCL5 ,Article ,Permeability ,Tight Junctions ,Mice ,medicine ,Animals ,Humans ,RNA, Messenger ,Transcription factor ,Immunity, Mucosal ,Mice, Knockout ,Hepatology ,biology ,Gene Expression Profiling ,Gastroenterology ,NF-kappa B ,Forkhead Transcription Factors ,Colitis ,Inflammatory Bowel Diseases ,Molecular biology ,Up-Regulation ,Reverse transcription polymerase chain reaction ,Disease Models, Animal ,Cytokine ,Trinitrobenzenesulfonic Acid ,biology.protein ,Intraepithelial lymphocyte ,Cytokines ,Tumor necrosis factor alpha ,medicine.symptom ,Caco-2 Cells ,Signal Transduction ,Transcription Factors - Abstract
Background & Aims FoxO4 is a member of the forkhead box transcription factor O (FoxO) subfamily. FoxO proteins are involved in diverse biological processes. In this study, we examine the role of FoxO4 in intestinal mucosal immunity and inflammatory bowel disease (IBD). Methods Foxo4 -null mice were subjected to trinitrobenzene sulfonic acid (TNBS) treatment. Microarray analysis and quantitative reverse transcription polymerase chain reaction were used to identify the cytokine transcripts that were altered by Foxo4 deletion. The effects of Foxo4 deficiency on the intestinal epithelial permeability and levels of tight junction proteins were examined by permeable fluorescent dye and Western blot. The molecular and cellular mechanisms by which FoxO4 regulates the mucosal immunity were explored through immunologic and biochemical analyses. The expression level of FoxO4 in intestinal epithelial cells of patients with IBD was examined with immunohistochemistry. Results Foxo4 -null mice were more susceptible to TNBS injury–induced colitis. The chemokine CCL5 is significantly up-regulated in the colonic epithelial cells of Foxo4 -null mice, with increased recruitment of CD4 + intraepithelial T cells and up-regulation of cytokines interferon-γ and tumor necrosis factor-α in the colon. Foxo4 deficiency also resulted in an increase in intestinal epithelial permeability and down-regulation of the tight junction proteins ZO-1 and claudin-1. Mechanistically, FoxO4 inhibited the transcriptional activity of nuclear factor-κB (NF-κB), and Foxo4 deficiency is associated with increased NF-κB activity in vivo. FoxO4 transcription is transiently repressed in response to TNBS treatment and in patients with IBD. Conclusion These results indicate that FoxO4 is an endogenous inhibitor of NF-κB and identify a novel function of FoxO4 in the regulation of NF-κB–mediated mucosal immunity.
- Published
- 2009
391. Chimeric mouse tumor models reveal differences in pathway activation between ERBB family- and KRAS-dependent lung adenocarcinomas
- Author
-
Lynda Chin, Joerg Heyer, Marcus Bosenberg, William M. Rideout, Tong Zi, M. Isabel Chiu, Ronald A. DePinho, James W. Horner, Shailaja Reddypalli, Rebecca Rancourt, Steven C. Clark, Yinghui Zhou, Tyler Jacks, Angela Bressel, Murray O. Robinson, and Jin Kyeung Woo
- Subjects
Lung Neoplasms ,Biomedical Engineering ,Bioengineering ,Mice, Transgenic ,Biology ,Adenocarcinoma ,medicine.disease_cause ,Applied Microbiology and Biotechnology ,Piperazines ,Transcriptome ,Proto-Oncogene Proteins p21(ras) ,Chimera (genetics) ,Mice ,medicine ,Animals ,Lung cancer ,PI3K/AKT/mTOR pathway ,Embryonic Stem Cells ,EGFR inhibitors ,Chimera ,medicine.disease ,ErbB Receptors ,Disease Models, Animal ,Phenotype ,Immunology ,Mutation ,Cancer research ,Quinazolines ,Molecular Medicine ,Immunohistochemistry ,KRAS ,Respiratory Insufficiency ,Biotechnology ,Signal Transduction - Abstract
To recapitulate the stochastic nature of human cancer development, we have devised a strategy for generating mouse tumor models that involves stepwise genetic manipulation of embryonic stem (ES) cells and chimera generation. Tumors in the chimeric animals develop from engineered cells in the context of normal tissue. Adenocarcinomas arising in an allelic series of lung cancer models containing HER2 (also known as ERBB2), KRAS or EGFR oncogenes exhibit features of advanced malignancies. Treatment of EGFR(L858R) and KRAS(G12V) chimeric models with an EGFR inhibitor resulted in near complete tumor regression and no response to the treatment, respectively, accurately reflecting previous clinical observations. Transcriptome and immunohistochemical analyses reveal that PI3K pathway activation is unique to ERBB family tumors whereas KRAS-driven tumors show activation of the JNK/SAP pathway, suggesting points of therapeutic intervention for this difficult-to-treat tumor category.
- Published
- 2009
392. mTORC1 signaling governs hematopoietic stem cell quiescence
- Author
-
Boyi Gan and Ronald A. DePinho
- Subjects
F-Box-WD Repeat-Containing Protein 7 ,Ubiquitin-Protein Ligases ,mTORC1 ,Promyelocytic Leukemia Protein ,F-box protein ,Article ,Tuberous Sclerosis Complex 1 Protein ,Mice ,medicine ,PTEN ,Animals ,Nuclear protein ,Molecular Biology ,Transcription factor ,Cell Proliferation ,biology ,Cell growth ,F-Box Proteins ,Tumor Suppressor Proteins ,PTEN Phosphohydrolase ,Hematopoietic stem cell ,Nuclear Proteins ,Cell Biology ,Hematopoietic Stem Cells ,Cell biology ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,Developmental Biology ,Signal Transduction ,Transcription Factors - Abstract
The stringent regulation of hematopoietic stem cell (HSC) quiescence versus cell cycle progression is essential for the preservation of a pool of long-term self-renewing cells and vital for sustaining an adequate supply of all blood lineages throughout life. Cell growth, the process that is mass increase, serves as a trigger for cell cycle progression and is regulated predominantly by mammalian target of rapamycin complex 1 (mTORC1) signaling. Emerging data from various mice models show deletion of several mTORC1 negative regulators, including PTEN, TSC1, PML and Fbxw7 result in similar HSC phenotypes characterized as HSC hyper-proliferation and subsequent exhaustion, and defective repopulating potential. Further pharmacological approaches show that PTEN, TSC1 and PML regulate HSC maintenance through mTORC1. mTORC1-mediated cell growth regulatory circuits thus plays a critical role in the regulation of HSC quiescence.
- Published
- 2009
393. IL-7 engages multiple mechanisms to overcome chronic viral infection and limit organ pathology
- Author
-
Katharina Lahl, Arda Shahinian, Thomas F. Tedder, Marc Pellegrini, Philipp A. Lang, Brigitte Assouline, Sameh Basta, Alisha R. Elford, Simon P. Preston, Tak W. Mak, Amy E. Lin, Michel Morre, Jihye Paik, Ronald A. DePinho, Jesse G. Toe, Thomas Calzascia, Karl S. Lang, Tim Sparwasser, and Pamela S. Ohashi
- Subjects
Naive T cell ,medicine.medical_treatment ,T cell ,T-Lymphocytes ,Programmed Cell Death 1 Receptor ,Medizin ,Down-Regulation ,Suppressor of Cytokine Signaling Proteins ,Biology ,Lymphocytic Choriomeningitis ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,Immunity ,medicine ,Animals ,Humans ,Lymphocytic choriomeningitis virus ,SOCS3 ,030304 developmental biology ,0303 health sciences ,Innate immune system ,Biochemistry, Genetics and Molecular Biology(all) ,Interleukin-6 ,Interleukin-7 ,Forkhead Transcription Factors ,Antigens, Differentiation ,Recombinant Proteins ,3. Good health ,Cytokine ,medicine.anatomical_structure ,Suppressor of Cytokine Signaling 3 Protein ,Immunology ,030215 immunology - Abstract
SummaryUnderstanding the factors that impede immune responses to persistent viruses is essential in designing therapies for HIV infection. Mice infected with LCMV clone-13 have persistent high-level viremia and a dysfunctional immune response. Interleukin-7, a cytokine that is critical for immune development and homeostasis, was used here to promote immunity toward clone-13, enabling elucidation of the inhibitory pathways underlying impaired antiviral immune response. Mechanistically, IL-7 downregulated a critical repressor of cytokine signaling, Socs3, resulting in amplified cytokine production, increased T cell effector function and numbers, and viral clearance. IL-7 enhanced thymic output to expand the naive T cell pool, including T cells that were not LCMV specific. Additionally, IL-7 promoted production of cytoprotective IL-22 that abrogated liver pathology. The IL-7-mediated effects were dependent on endogenous IL-6. These attributes of IL-7 have profound implications for its use as a therapeutic in the treatment of chronic viral diseases.
- Published
- 2009
394. Identification of Novel Alternative Splice Isoforms of Circulating Proteins in a Mouse Model of Human Pancreatic Cancer
- Author
-
Chunxia Lu, Gilbert S. Omenn, Samir M. Hanash, Rajasree Menon, David J. States, Yan Zhang, Nabeel Bardeesy, Qing Zhang, Ronald A. DePinho, and Damian Fermin
- Subjects
Male ,Cancer Research ,Molecular Sequence Data ,Mouse Protein ,Article ,Proteoglycan 4 ,Animals ,Humans ,Protein Isoforms ,splice ,Amino Acid Sequence ,Peptide sequence ,Gene ,Genetics ,biology ,Reverse Transcriptase Polymerase Chain Reaction ,Alternative splicing ,Malate dehydrogenase 1 ,Blood Proteins ,Molecular biology ,Neoplasm Proteins ,Pancreatic Neoplasms ,Alternative Splicing ,Disease Models, Animal ,Oncology ,Proteome ,biology.protein - Abstract
To assess the potential of tumor-associated, alternatively spliced gene products as a source of biomarkers in biological fluids, we have analyzed a large data set of mass spectra derived from the plasma proteome of a mouse model of human pancreatic ductal adenocarcinoma. MS/MS spectra were interrogated for novel splice isoforms using a nonredundant database containing an exhaustive three-frame translation of Ensembl transcripts and gene models from ECgene. This integrated analysis identified 420 distinct splice isoforms, of which 92 did not match any previously annotated mouse protein sequence. We chose seven of those novel variants for validation by reverse transcription–PCR. The results were concordant with the proteomic analysis. All seven novel peptides were successfully amplified in pancreas specimens from both wild-type and mutant mice. Isotopic labeling of cysteine-containing peptides from tumor-bearing mice and wild-type controls enabled relative quantification of the proteins. Differential expression between tumor-bearing and control mice was notable for peptides from novel variants of muscle pyruvate kinase, malate dehydrogenase 1, glyceraldehyde-3-phosphate dehydrogenase, proteoglycan 4, minichromosome maintenance, complex component 9, high mobility group box 2, and hepatocyte growth factor activator. Our results show that, in a mouse model for human pancreatic cancer, novel and differentially expressed alternative splice isoforms are detectable in plasma and may be a source of candidate biomarkers. [Cancer Res 2009;69(1):300–9]
- Published
- 2009
395. Complex Transcriptional Regulation of myc Family Gene Expression in the Developing Mouse Brain and Liver
- Author
-
Ronald A. DePinho, Lin Xu, and Sharon D. Morgenbesser
- Subjects
Transcription, Genetic ,Molecular Sequence Data ,Restriction Mapping ,Genes, myc ,RNA polymerase II ,Biology ,Mice ,Exon ,Gene expression ,Tumor Cells, Cultured ,Transcriptional regulation ,Animals ,RNA, Messenger ,Transcriptional attenuation ,Promoter Regions, Genetic ,Gene ,Molecular Biology ,MYC Family Gene ,Regulation of gene expression ,Base Sequence ,Brain ,Exons ,Cell Biology ,Molecular biology ,Animals, Newborn ,Gene Expression Regulation ,Liver ,Organ Specificity ,Multigene Family ,biology.protein ,RNA ,RNA Polymerase II ,Research Article - Abstract
myc family genes (c-, N-, and L-myc) have been shown to be differentially expressed with respect to tissue type and developmental stage. To define and compare the regulatory mechanisms governing their differential developmental expression, we examined the transcriptional regulation of each myc family member during murine postnatal brain and liver development. Nuclear run-on transcription assays demonstrated that both the rate of transcriptional initiation and the degree of transcriptional blocking contribute in a complex manner to the regulation of all three genes. During postnatal brain development, the relative contribution of each transcriptional control mechanism to the regulation of myc family gene expression was found to be different for each gene. For instance, while modulation of transcriptional attenuation did not appear to contribute to the down-regulation of L-myc expression, attenuation was found to be the dominant mechanism by which steady-state N-myc mRNA levels were down-regulated. Different transcriptional strategies were found to be employed in newborn versus adult developing liver for repression of N- and L-myc expression. Undetectable steady-state N- and L-myc mRNA levels in newborn liver were associated with a very low rate of transcriptional initiation, whereas the lack of N- and L-myc expression at the adult stage was accompanied by a high rate of initiation and a striking degree of transcriptional attenuation. Transcriptional attenuation in the N-myc gene was found to map to a region encoding a potential stem-loop structure followed by a thymine tract within the first exon and was not dependent on the use of a specific transcriptional start site.
- Published
- 1991
- Full Text
- View/download PDF
396. Instability of immunoglobulin genes in S107 cell line
- Author
-
Seung Uon Shin, Donald J. Zack, Ronald A. DePinho, Stuart Rudikoff, and Matthew D. Scharff
- Subjects
Molecular Sequence Data ,Gene Rearrangement, B-Lymphocyte, Heavy Chain ,Immunoglobulin Variable Region ,Somatic hypermutation ,Biology ,medicine.disease_cause ,Mice ,Germline mutation ,Gene duplication ,Tumor Cells, Cultured ,Genetics ,medicine ,Animals ,Crossing Over, Genetic ,RNA, Messenger ,Gene ,Mice, Inbred BALB C ,Mutation ,Base Sequence ,Genes, Immunoglobulin ,Cell Biology ,General Medicine ,Molecular biology ,Phenotype ,Cell culture ,Multigene Family ,Immunoglobulin heavy chain ,Immunoglobulin Constant Regions ,Immunoglobulin Heavy Chains - Abstract
Somatic mutation occurs frequently in rearranged and expressed immunoglobulin variable region genes in vivo. In contrast, V region hypermutation seldom occurs in antibody-forming cells in culture. The S107 mouse myeloma cell line is one of the few cell lines that has been observed to generate V region mutations frequently and spontaneously in vitro. Detailed examination reveals that both the S107 tumor and the cell line derived from it contain and express a duplicated heavy-chain gene. In culture, only one of the two heavy-chain genes undergoes both V and C region mutation, and variants with complex phenotypes and genotypes arise as a result of mutation and segregation of these duplicated genes.
- Published
- 1991
- Full Text
- View/download PDF
397. Structural and functional analysis of the mouse mdr1b gene promoter
- Author
-
Ronald A. DePinho, Dalia Cohen, Stephen I Hong Hsu, Nancy Carrasco, Richard L. Piekarz, and Susan Band Horwitz
- Subjects
Chloramphenicol acetyltransferase ,Gene isoform ,Expression vector ,5' flanking region ,Gene expression ,Transcriptional regulation ,Promoter ,Cell Biology ,Biology ,Molecular Biology ,Biochemistry ,Gene ,Molecular biology - Abstract
The overproduction of P-glycoprotein, an integral membrane protein thought to function as a drug efflux pump, is the hallmark of the multidrug resistance phenotype. In murine multidrug resistant J774.2 cell lines, distinct mdr genes, mdr1a and mdr1b, encode unique P-glycoprotein isoforms. To examine the transcriptional regulation of the mdr1b gene, its promoter was isolated and characterized. The transcription initiation site was mapped by primer extension, and the 5'-flanking region was sequenced. Several potential regulatory elements were identified in this region. A transient expression vector was constructed by fusion of 540 base pairs of 5'-flanking sequence and part of the first untranslated exon to the chloramphenicol acetyltransferase (CAT) gene. When transfected into monkey kidney COS-1, rat pituitary GH3 or T47D human breast cells, the mdr1b 5'-flanking sequences were capable of driving CAT expression. Transient transfection studies using deletion subclones of the mdr1b-CAT construct were done to locate potential cis-acting sequences. The studies indicate the presence of cis-acting elements in the 5'-flanking region of the mdr1b gene. The implications of these findings for expression and regulation of the mdr1b gene are discussed.
- Published
- 1991
- Full Text
- View/download PDF
398. Unlimited Mileage from Telomerase?
- Author
-
Ronald A. DePinho and Titia de Lange
- Subjects
Genetics ,Telomerase ,Multidisciplinary ,Cell division ,Cellular senescence ,Telomere ,Biology ,medicine.disease_cause ,Retinoblastoma Protein ,Cell biology ,Proto-Oncogene Proteins c-myc ,Cell Transformation, Neoplastic ,Neoplasms ,ras Proteins ,medicine ,Animals ,Humans ,Carcinogenesis ,human activities ,Cell Division ,Cellular Senescence ,Cyclin-Dependent Kinase Inhibitor p16 ,Signal Transduction - Abstract
Last year, scientists from the company Geron reported that inserting the gene for telomerase--the enzyme that makes the protective ends of chromosomes and is turned on in many tumors--into fibroblasts can prolong their ability to replicate, up to 100 generations. But how is this different from what happens in cancer cells? In their Perspective, de Lange and DePinho explore new evidence that begins to sort out the controls of cellular replication and oncogenesis. They liken the function of telomerase to that of a gas tank in a car, with oncogenes as the gas pedal and tumor suppressors as the brake.
- Published
- 1999
- Full Text
- View/download PDF
399. Genomic alterations link Rho family of GTPases to the highly invasive phenotype of pancreas cancer
- Author
-
Hongwu Zheng, Ming-Sound Tsao, Marina Protopopova, Alec C. Kimmelman, Scott Deroo, John P. McGrath, Lynda Chin, Xiaoxu Wang, Aditya H. Ponugoti, Gerald C. Chu, Giminna Yeo, Bin Feng, Aram F. Hezel, David S. Klimstra, Ergun Sahin, Haoqiang Ying, Jianhua Zhang, Shenghong Yang, Roustem Nabioullin, Mark Redston, Alexei Protopopov, Jean X. Zhang, Andrew J. Aguirre, P. Andrew Futreal, Yonghong Xiao, Jihye Paik, Ronald A. DePinho, Elena Ivanova, and William C. Hahn
- Subjects
Genome instability ,rho GTP-Binding Proteins ,Mice, Nude ,Rho family of GTPases ,Protein Serine-Threonine Kinases ,Mice ,Cell Movement ,Pancreatic cancer ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,p21-activated kinases ,Cell Line, Transformed ,Regulation of gene expression ,Multidisciplinary ,biology ,Pancreatic Ducts ,Cancer ,Genomics ,Biological Sciences ,medicine.disease ,Phenotype ,Cell biology ,Gene Expression Regulation, Neoplastic ,Pancreatic Neoplasms ,p21-Activated Kinases ,biology.protein ,Cancer research ,Signal transduction ,Proto-Oncogene Proteins c-akt ,Carcinoma, Pancreatic Ductal ,Signal Transduction - Abstract
Pancreas ductal adenocarcinoma (PDAC) is a highly lethal cancer that typically presents as advanced, unresectable disease. This invasive tendency, coupled with intrinsic resistance to standard therapies and genome instability, are major contributors to poor long-term survival. The genetic elements governing the invasive propensity of PDAC have not been well elucidated. Here, in the course of validating resident genes in highly recurrent and focal amplifications in PDAC, we have identified Rio Kinase 3 (RIOK3) as an amplified gene that alters cytoskeletal architecture as well as promotes pancreatic ductal cell migration and invasion. We determined that RIOK3 promotes its invasive activities through activation of the small G protein, Rac. This genomic and functional link to Rac signaling prompted a genome wide survey of other components of the Rho family network, revealing p21 Activated Kinase 4 (PAK4) as another amplified gene in PDAC tumors and cell lines. Like RIOK3, PAK4 promotes pancreas ductal cell motility and invasion. Together, the genomic and functional profiles establish the Rho family GTP-binding proteins as integral to the hallmark invasive nature of this lethal disease.
- Published
- 2008
400. HDAC2 negatively regulates memory formation and synaptic plasticity
- Author
-
Nadine F. Joseph, Ralph Mazitschek, Emanuela Giacometti, Jan Hermen Dannenberg, James E. Bradner, Stephen J. Haggarty, Ying Zhou, Rudolf Jaenisch, Li-Huei Tsai, Thomas J.F. Nieland, Jun Gao, X. L. Wang, Ronald A. DePinho, and Ji-Song Guan
- Subjects
Male ,Dendritic spine ,Epigenetics in learning and memory ,Dendritic Spines ,Hippocampus ,Histone Deacetylase 2 ,Histone Deacetylase 1 ,Biology ,Bioinformatics ,Hydroxamic Acids ,Histone Deacetylases ,Article ,Synapse ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Electrical Synapses ,Memory ,medicine ,Memory impairment ,Animals ,Learning ,Promoter Regions, Genetic ,030304 developmental biology ,Mice, Knockout ,Neurons ,0303 health sciences ,Vorinostat ,Multidisciplinary ,Memoria ,Neurodegeneration ,Sodium ,medicine.disease ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,Repressor Proteins ,Butyrates ,Gene Expression Regulation ,Synaptic plasticity ,Female ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Chromatin modifications, especially histone-tail acetylation, have been implicated in memory formation. Increased histone-tail acetylation induced by inhibitors of histone deacetylases (HDACis) facilitates learning and memory in wild-type mice as well as in mouse models of neurodegeneration. Harnessing the therapeutic potential of HDACis requires knowledge of the specific HDAC family member(s) linked to cognitive enhancement. Here we show that neuron-specific overexpression of HDAC2, but not that of HDAC1, decreased dendritic spine density, synapse number, synaptic plasticity and memory formation. Conversely, Hdac2 deficiency resulted in increased synapse number and memory facilitation, similar to chronic treatment with HDACis in mice. Notably, reduced synapse number and learning impairment of HDAC2-overexpressing mice were ameliorated by chronic treatment with HDACis. Correspondingly, treatment with HDACis failed to further facilitate memory formation in Hdac2-deficient mice. Furthermore, analysis of promoter occupancy revealed an association of HDAC2 with the promoters of genes implicated in synaptic plasticity and memory formation. Taken together, our results suggest that HDAC2 functions in modulating synaptic plasticity and long-lasting changes of neural circuits, which in turn negatively regulates learning and memory. These observations encourage the development and testing of HDAC2-selective inhibitors for human diseases associated with memory impairment.
- Published
- 2008
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.