Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Curr Protoc ; 1(3): e86, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33751862

ABSTRACT

Relationships with mentors and labmates are defining aspects of a researcher's journey in science. Ideally, these interactions are outstanding opportunities to learn from others and provide the basis for lifelong collaborations. Unfortunately, sometimes interpersonal dynamics in the lab are challenging. Graduate students entering the lab can greatly benefit from advice about navigating the interpersonal aspects of doing science. This article covers essential recommendations for developing a good trainee-mentor relationship and working well with peers in the lab, or being a "good lab citizen." Lab members-especially graduate students-often spend more time with labmates than with their friends and family during their graduate career, making these relationships essential to their well-being. The guidance also covers some advice for handling a tense relationship or problematic work environment. Finally, the advice concludes by discussing how to manage the fear of failure, overcome imposter syndrome, develop self-awareness, and cope with stress. These four issues are fundamental to success in research but are not discussed with graduate students as much as may be necessary. © 2021 Wiley Periodicals LLC.


Subject(s)
Mentors , Peer Group , Humans , Students
2.
Curr Protoc ; 1(3): e87, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33740292

ABSTRACT

Researchers must conduct research responsibly for it to have an impact and to safeguard trust in science. Essential responsibilities of researchers include using rigorous, reproducible research methods, reporting findings in a trustworthy manner, and giving the researchers who contributed appropriate authorship credit. This "how-to" guide covers strategies and practices for doing reproducible research and being a responsible author. The article also covers how to utilize decision-making strategies when uncertain about the best way to proceed in a challenging situation. The advice focuses especially on graduate students, but is appropriate for undergraduates and experienced researchers. It begins with an overview of responsible conduct of research, research misconduct, and ethical behavior in the scientific workplace. The takeaway message is that responsible conduct of research requires a thoughtful approach to doing research in order to ensure trustworthy results and conclusions, and that researchers receive fair credit. © 2021 Wiley Periodicals LLC.


Subject(s)
Authorship , Scientific Misconduct , Humans , Morals , Research Personnel , Students
3.
Cell Rep ; 27(7): 2063-2074.e5, 2019 05 14.
Article in English | MEDLINE | ID: mdl-31091446

ABSTRACT

Competition for nutrients like glucose can metabolically restrict T cells and contribute to their hyporesponsiveness during cancer. Metabolic adaptation to the surrounding microenvironment is therefore key for maintaining appropriate cell function. For instance, cancer cells use acetate as a substrate alternative to glucose to fuel metabolism and growth. Here, we show that acetate rescues effector function in glucose-restricted CD8+ T cells. Mechanistically, acetate promotes histone acetylation and chromatin accessibility and enhances IFN-γ gene transcription and cytokine production in an acetyl-CoA synthetase (ACSS)-dependent manner. Ex vivo acetate treatment increases IFN-γ production by exhausted T cells, whereas reducing ACSS expression in T cells impairs IFN-γ production by tumor-infiltrating lymphocytes and tumor clearance. Thus, hyporesponsive T cells can be epigenetically remodeled and reactivated by acetate, suggesting that pathways regulating the use of substrates alternative to glucose could be therapeutically targeted to promote T cell function during cancer.


Subject(s)
Acetate-CoA Ligase/immunology , Acetates/immunology , CD8-Positive T-Lymphocytes/immunology , Glucose/immunology , Interferon-gamma/immunology , Neoplasm Proteins/immunology , Neoplasms, Experimental/immunology , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Line, Tumor , Humans , Mice , Neoplasms, Experimental/pathology
4.
FASEB Bioadv ; 1(7): 404-414, 2019 Jul.
Article in English | MEDLINE | ID: mdl-32095781

ABSTRACT

The orphan small nucleolar RNA (snoRNA) ACA11 is overexpressed as a result of the t(4;14) chromosomal translocation in multiple myeloma (MM), increases reactive oxygen species, and drives cell proliferation. Like other snoRNAs, ACA11 is predominantly localized to a sub-nuclear organelle, the nucleolus. We hypothesized that increased ACA11 expression would increase ribosome biogenesis and protein synthesis. We found that ACA11 overexpression in MM cells increased nucleolar area and number as well as silver-binding nucleolar organizing regions (AgNORs). Supporting these data, samples from t(4;14)-positive patients had higher AgNORs scores than t(4;14)-negative samples. ACA11 also upregulated ribosome production, pre-47S rRNA synthesis, and protein synthesis in a ROS-dependent manner. Lastly, ACA11 overexpression enhanced the response to proteasome inhibitor in MM cells, while no effect was found in response to high doses of melphalan. Together, these data demonstrate that ACA11 stimulates ribosome biogenesis and influences responses to chemotherapy. ACA11 may be a useful target to individualize the treatment for t(4;14)-positive myeloma patients.

5.
Nat Commun ; 9(1): 3787, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224629

ABSTRACT

Nearly all patients with small cell lung cancer (SCLC) eventually relapse with chemoresistant disease. The molecular mechanisms driving chemoresistance in SCLC remain un-characterized. Here, we describe whole-exome sequencing of paired SCLC tumor samples procured at diagnosis and relapse from 12 patients, and unpaired relapse samples from 18 additional patients. Multiple somatic copy number alterations, including gains in ABCC1 and deletions in MYCL, MSH2, and MSH6, are identifiable in relapsed samples. Relapse samples also exhibit recurrent mutations and loss of heterozygosity in regulators of WNT signaling, including CHD8 and APC. Analysis of RNA-sequencing data shows enrichment for an ASCL1-low expression subtype and WNT activation in relapse samples. Activation of WNT signaling in chemosensitive human SCLC cell lines through APC knockdown induces chemoresistance. Additionally, in vitro-derived chemoresistant cell lines demonstrate increased WNT activity. Overall, our results suggest WNT signaling activation as a mechanism of chemoresistance in relapsed SCLC.


Subject(s)
Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Wnt Signaling Pathway/genetics , Adenomatous Polyposis Coli Protein/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cadherins/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Loss of Heterozygosity , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mutation , Neoplasm Recurrence, Local , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Exome Sequencing , Wnt Signaling Pathway/drug effects
6.
Article in English | MEDLINE | ID: mdl-30619092

ABSTRACT

Numerous human diseases arise from alterations of genetic information, most notably DNA mutations. Thought to be merely the intermediate between DNA and protein, changes in RNA sequence were an afterthought until the discovery of RNA editing 30 years ago. RNA editing alters RNA sequence without altering the sequence or integrity of genomic DNA. The most common RNA editing events are A-to-I changes mediated by adenosine deaminase acting on RNA (ADAR), and C-to-U editing mediated by apolipoprotein B mRNA editing enzyme, catalytic polypeptide 1 (APOBEC1). Both A-to-I and C-to-U editing were first identified in the context of embryonic development and physiological homeostasis. The role of RNA editing in human disease has only recently started to be understood. In this review, the impact of RNA editing on the development of cancer and metabolic disorders will be examined. Distinctive functions of each RNA editase that regulate either A-to-I or C-to-U editing will be highlighted in addition to pointing out important regulatory mechanisms governing these processes. The potential of developing novel therapeutic approaches through intervention of RNA editing will be explored. As the role of RNA editing in human disease is elucidated, the clinical utility of RNA editing targeted therapies will be needed. This review aims to serve as a bridge of information between past findings and future directions of RNA editing in the context of cancer and metabolic disease.

7.
FASEB J ; 31(2): 482-490, 2017 02.
Article in English | MEDLINE | ID: mdl-28148777

ABSTRACT

Overexpression of the multiple myeloma set domain (MMSET) Wolf-Hirschhorn syndrome candidate 1 gene, which contains an orphan box H/ACA class small nucleolar RNA, ACA11, in an intron, is associated with several cancer types, including multiple myeloma (MM). ACA11 and MMSET are overexpressed cotranscriptionally as a result of the t(4;14) chromosomal translocation in a subset of patients with MM. RNA sequencing of CD138+ tumor cells from t(4;14)-positive and -negative MM patient bone marrow samples revealed an enhanced oxidative phosphorylation mRNA signature. Supporting these data, ACA11 overexpression in a t(4;14)-negative MM cell line, MM1.S, demonstrated enhanced reactive oxygen species (ROS) levels. In addition, an enhancement of cell proliferation, increased soft agar colony size, and elevated ERK1/2 phosphorylation were observed. This ACA11-driven hyperproliferative phenotype depended on increased ROS levels as exogenously added antioxidants attenuate the increased proliferation. A major transcriptional regulator of the cellular antioxidant response, nuclear factor (erythroid-derived 2)-like 2 (NRF2), shuttled to the nucleus, as expected, in response to ACA11-driven increases in ROS; however, transcriptional up-regulation of some of NRF2's antioxidant target genes was abrogated in the presence of ACA11 overexpression. These data show for the first time that ACA11 promotes proliferation through inhibition of NRF2 function resulting in sustained ROS levels driving cancer cell proliferation.-Mahajan, N., Wu, H.-J., Bennett, R. L., Troche, C., Licht, J. D., Weber, J. D., Maggi, L. B., Jr., Tomasson, M. H. Sabotaging of the oxidative stress response by an oncogenic noncoding RNA.


Subject(s)
Fibroblasts/physiology , Gene Expression Regulation/physiology , Oncogenes/physiology , RNA, Untranslated/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Humans , Mice , Multiple Myeloma/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress , RNA, Untranslated/genetics , Reactive Oxygen Species
8.
Breast Cancer Res ; 17: 51, 2015 Apr 08.
Article in English | MEDLINE | ID: mdl-25888162

ABSTRACT

With tremendous advances in sequencing and analysis in recent years, a wealth of genetic information has become available to identify and classify breast cancer into five main subtypes - luminal A, luminal B, claudin-low, human epidermal growth factor receptor 2-enriched, and basal-like. Current treatment decisions are often based on these classifications, and while more beneficial than any single treatment for all breast cancers, targeted therapeutics have exhibited limited success with most of the subtypes. Luminal B breast cancers are associated with early relapse following endocrine therapy and often exhibit a poor prognosis that is similar to that of the aggressive basal-like breast cancers. Identifying genetic components that contribute to the luminal B endocrine resistant phenotype has become imperative. To this end, numerous groups have identified activation of the phosphatidylinositol 3-kinase (PI3K) pathway as a common recurring event in luminal B cancers with poor outcome. Examining the pathways downstream of PI3K, Fu and colleagues have recreated a human model of the luminal B subtype of breast cancer. The authors were able to reduce expression of phosphatase and tensin homolog (PTEN), the negative regulator of PI3K, using inducible short hairpin RNAs. By varying the expression of PTEN, the authors effectively conferred endocrine resistance and recapitulated the luminal B gene expression signature. Using this system in vitro and in vivo, they then tested the ability of selective kinase inhibitors downstream of PI3K to enhance current endocrine therapies. A combination of fulvestrant, which blocks ligand-dependent and -independent estrogen receptor signaling, with protein kinase B inhibition was found to overcome endocrine resistance. These findings squarely place PTEN expression levels at the nexus of luminal B breast cancers and indicates that patients with PTEN-low estrogen receptor-positive tumors might benefit from combined endocrine and PI3K pathway therapies.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Breast Neoplasms/drug therapy , PTEN Phosphohydrolase/metabolism , Sirolimus/pharmacology , Animals , Female , Humans
9.
Cell Rep ; 7(2): 514-526, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24726362

ABSTRACT

The ARF and p53 tumor suppressors are thought to act in a linear pathway to prevent cellular transformation in response to various oncogenic signals. Here, we show that loss of p53 leads to an increase in ARF protein levels, which function to limit the proliferation and tumorigenicity of p53-deficient cells by inhibiting an IFN-ß-STAT1-ISG15 signaling axis. Human triple-negative breast cancer (TNBC) tumor samples with coinactivation of p53 and ARF exhibit high expression of both STAT1 and ISG15, and TNBC cell lines are sensitive to STAT1 depletion. We propose that loss of p53 function and subsequent ARF induction creates a selective pressure to inactivate ARF and propose that tumors harboring coinactivation of ARF and p53 would benefit from therapies targeted against STAT1 and ISG15 activation.


Subject(s)
ADP-Ribosylation Factors/metabolism , Breast Neoplasms/metabolism , Cytokines/metabolism , Interferon-beta/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitins/metabolism , ADP-Ribosylation Factors/genetics , Animals , Cell Line , Cell Proliferation , Cells, Cultured , Female , Humans , Mice , Mutation , STAT1 Transcription Factor/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics
10.
Biochim Biophys Acta ; 1842(6): 831-9, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24525025

ABSTRACT

Since its discovery close to twenty years ago, the ARF tumor suppressor has played a pivotal role in the field of cancer biology. Elucidating ARF's basal physiological function in the cell has been the focal interest of numerous laboratories throughout the world for many years. Our current understanding of ARF is constantly evolving to include novel frameworks for conceptualizing the regulation of this critical tumor suppressor. As a result of this complexity, there is great need to broaden our understanding of the intricacies governing the biology of the ARF tumor suppressor. The ARF tumor suppressor is a key sensor of signals that instruct a cell to grow and proliferate and is appropriately localized in nucleoli to limit these processes. This article is part of a Special Issue entitled: Role of the Nucleolus in Human Disease.


Subject(s)
Cell Nucleolus/metabolism , Ribosomes/metabolism , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Cycle Checkpoints/genetics , Cell Nucleolus/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Protein Binding , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Ribosomes/genetics , Tumor Suppressor Protein p14ARF/genetics , Tumor Suppressor Protein p53/genetics
11.
Cell ; 153(6): 1239-51, 2013 Jun 06.
Article in English | MEDLINE | ID: mdl-23746840

ABSTRACT

A "switch" from oxidative phosphorylation (OXPHOS) to aerobic glycolysis is a hallmark of T cell activation and is thought to be required to meet the metabolic demands of proliferation. However, why proliferating cells adopt this less efficient metabolism, especially in an oxygen-replete environment, remains incompletely understood. We show here that aerobic glycolysis is specifically required for effector function in T cells but that this pathway is not necessary for proliferation or survival. When activated T cells are provided with costimulation and growth factors but are blocked from engaging glycolysis, their ability to produce IFN-γ is markedly compromised. This defect is translational and is regulated by the binding of the glycolysis enzyme GAPDH to AU-rich elements within the 3' UTR of IFN-γ mRNA. GAPDH, by engaging/disengaging glycolysis and through fluctuations in its expression, controls effector cytokine production. Thus, aerobic glycolysis is a metabolically regulated signaling mechanism needed to control cellular function.


Subject(s)
Glycolysis , Lymphocyte Activation , Oxidative Phosphorylation , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , 3' Untranslated Regions , Animals , Cell Proliferation , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Interferon-gamma/genetics , Listeria monocytogenes , Listeriosis/immunology , Mice , Mice, Inbred C57BL , Protein Biosynthesis , T-Lymphocytes/immunology
13.
PLoS One ; 7(7): e42005, 2012.
Article in English | MEDLINE | ID: mdl-22860046

ABSTRACT

Osteoclasts are terminally differentiated cells that attach to bone and secrete proteases to degrade the bone matrix. The primary protease responsible for the degradation of the organic component of the bone matrix is Cathepsin K, which was largely thought to be unique to osteoclasts. Given its apparent selective expression in osteoclasts, the Cathepsin K promoter has been engineered to drive the expression of Cre recombinase in mice and has been the most relevant tool for generating osteoclast-specific gene loss. In an effort to understand the role of the ARF tumor suppressor in osteoclasts, we crossed Arf (fl/fl) mice to Ctsk(Cre/+) mice, which unexpectedly resulted in the germline loss of Arf. We subsequently confirmed Cre activity in gametes by generating Ctsk(Cre/+); Rosa(+) mice. These results raise significant concerns regarding in vivo bone phenotypes created using Ctsk(Cre/+) mice and warrant further investigation into the role of Cathepsin K in gametes as well as alternative tools for studying osteoclast-specific gene loss in vivo.


Subject(s)
Cathepsin K/genetics , Gene Deletion , Germ Cells , Integrases/genetics , Animals , Base Sequence , DNA Primers , Female , Male , Mice , Real-Time Polymerase Chain Reaction
14.
J Clin Invest ; 122(8): 2793-806, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22751105

ABSTRACT

The histone methyltransferase WHSC1 (also known as MMSET) is overexpressed in multiple myeloma (MM) as a result of the t(4;14) chromosomal translocation and in a broad variety of other cancers by unclear mechanisms. Overexpression of WHSC1 did not transform wild-type or tumor-prone primary hematopoietic cells. We found that ACA11, an orphan box H/ACA class small nucleolar RNA (snoRNA) encoded within an intron of WHSC1, was highly expressed in t(4;14)-positive MM and other cancers. ACA11 localized to nucleoli and bound what we believe to be a novel small nuclear ribonucleoprotein (snRNP) complex composed of several proteins involved in postsplicing intron complexes. RNA targets of this uncharacterized snRNP included snoRNA intermediates hosted within ribosomal protein (RP) genes, and an RP gene signature was strongly associated with t(4;14) in patients with MM. Expression of ACA11 was sufficient to downregulate RP genes and other snoRNAs implicated in the control of oxidative stress. ACA11 suppressed oxidative stress, afforded resistance to chemotherapy, and increased the proliferation of MM cells, demonstrating that ACA11 is a critical target of the t(4;14) translocation in MM and suggesting an oncogenic role in other cancers as well.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , RNA, Small Nucleolar/genetics , RNA, Small Nucleolar/metabolism , Repressor Proteins/metabolism , Translocation, Genetic , Animals , Base Sequence , Cell Proliferation , Cell Transformation, Neoplastic , Chromosomes, Human, Pair 14/genetics , Chromosomes, Human, Pair 4/genetics , DNA, Complementary/genetics , DNA, Neoplasm/genetics , Gene Expression Profiling , Humans , Introns , Mice , Molecular Sequence Data , Multiple Myeloma/pathology , Nucleic Acid Conformation , Oxidative Stress , RNA, Neoplasm/chemistry , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Small Nucleolar/chemistry , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism
15.
Cancer Res ; 69(16): 6454-62, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19638589

ABSTRACT

Cellular survival from radiation-induced DNA damage requires access to sites of damage for the assembly of repair complexes and the subsequent repair, particularly the repair of DNA double strand breaks (DSB). Hyperthermia causes changes in protein-protein/DNA interactions in the nucleus that block access to sites of DNA damage. Studies presented here indicate that the nucleolar protein, nucleophosmin (NPM), redistributes from the nucleolus following hyperthermia, increases its association with DNA, and blocks access to DNA DSBs. Reduction of NPM significantly reduces heat-induced radiosensitization, but reduced NPM level does not alter radiation sensitivity per se. NPM knockdown reduces heat-induced inhibition of DNA DSB repair. Also, these results suggest that NPM associates with nuclear matrix attachment region DNA in heat-shocked cells.


Subject(s)
Heat-Shock Response/physiology , Hot Temperature , Nuclear Proteins/metabolism , Radiation Tolerance/physiology , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/genetics , HeLa Cells , Heat-Shock Response/drug effects , Heat-Shock Response/genetics , Humans , Models, Biological , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nucleophosmin , RNA, Small Interfering/pharmacology , Radiation Dosage , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Tissue Distribution/drug effects , Tumor Cells, Cultured
16.
Mol Cell Biol ; 28(23): 7050-65, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18809582

ABSTRACT

Nucleophosmin (NPM) (B23) is an essential protein in mouse development and cell growth; however, it has been assigned numerous roles in very diverse cellular processes. Here, we present a unified mechanism for NPM's role in cell growth; NPM directs the nuclear export of both 40S and 60S ribosomal subunits. NPM interacts with rRNA and large and small ribosomal subunit proteins and also colocalizes with large and small ribosomal subunit proteins in the nucleolus, nucleus, and cytoplasm. The transduction of NPM shuttling-defective mutants or the loss of Npm1 inhibited the nuclear export of both the 40S and 60S ribosomal subunits, reduced the available pool of cytoplasmic polysomes, and diminished overall protein synthesis without affecting rRNA processing or ribosome assembly. While the inhibition of NPM shuttling can block cellular proliferation, the dramatic effects on ribosome export occur prior to cell cycle inhibition. Modest increases in NPM expression amplified the export of newly synthesized rRNAs, resulting in increased rates of protein synthesis and indicating that NPM is rate limiting in this pathway. These results support the idea that NPM-regulated ribosome export is a fundamental process in cell growth.


Subject(s)
Active Transport, Cell Nucleus , Cell Proliferation , Nuclear Proteins/physiology , Ribosomes/metabolism , Animals , Cells, Cultured , Cytoplasm , Humans , Kinetics , Mice , Mice, Knockout , Molecular Chaperones , Mutation , Nuclear Proteins/genetics , Nucleophosmin , Polyribosomes , Protein Biosynthesis , RNA, Ribosomal/metabolism
17.
Mol Cell Biol ; 28(3): 1068-80, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18070929

ABSTRACT

The nucleolus is the center of ribosome synthesis, with the nucleophosmin (NPM) and p19(ARF) proteins antagonizing one another to either promote or inhibit growth. However, basal NPM and ARF proteins form nucleolar complexes whose functions remain unknown. Nucleoli from Arf(-/)(-) cells displayed increased nucleolar area, suggesting that basal ARF might regulate key nucleolar functions. Concordantly, ribosome biogenesis and protein synthesis were dramatically elevated in the absence of Arf, causing these cells to exhibit tremendous gains in protein amounts and increases in cell volume. The transcription of ribosomal DNA (rDNA), the processing of nascent rRNA molecules, and the nuclear export of ribosomes were all increased in the absence of ARF. Similar results were obtained using targeted lentiviral RNA interference of ARF in wild-type MEFs. Postmitotic osteoclasts from Arf-null mice exhibited hyperactivity in vitro and in vivo, demonstrating a physiological function for basal ARF. Moreover, the knockdown of NPM blocked the increases in Arf(-/-) ribosome output and osteoclast activity, demonstrating that these gains require NPM. Thus, basal ARF proteins act as a monitor of steady-state ribosome biogenesis and growth independent of their ability to prevent unwarranted hyperproliferation.


Subject(s)
Cell Nucleolus/ultrastructure , Cyclin-Dependent Kinase Inhibitor p16/physiology , Nuclear Proteins/physiology , Animals , DNA, Ribosomal/genetics , Mice , Nucleophosmin , Protein Biosynthesis , RNA, Ribosomal/genetics , Ribosomes/genetics , Ribosomes/metabolism , Transcription, Genetic , Tumor Suppressor Proteins
18.
Curr Med Chem ; 14(17): 1815-27, 2007.
Article in English | MEDLINE | ID: mdl-17627519

ABSTRACT

One of the outstanding fundamental questions in cancer cell biology concerns how cells coordinate cellular growth (or macromolecular synthesis) with cell cycle progression and mitosis. Intuitively, rapidly dividing cells must have some control over these processes; otherwise cells would continue to shrink in volume with every passing cycle, similar to the cytoreductive divisions seen in the very early stages of embryogenesis. The problem is easily solved in unicellular organisms, such as yeast, as their growth rates are entirely dependent on nutrient availability. Multicellular organisms such as mammals, however, must have acquired additional levels of control, as nutrient availability is seldom an issue and the organism has a prodigious capacity to store necessary metabolites in the form of glycogen, lipids, and protein. Furthermore, the specific needs and specialized architecture of tissues must constrain growth for growth's sake; if not, the necessary function of the organ could be lost. While certainly a myriad of mechanisms for preventing this exist via initiating cell death (e.g. apoptosis, autophagy, necrosis), these all depend on some external cue, such as death signals, hypoxia, lack of nutrients or survival signals. However there must also be some cell autonomous method for surveying against inappropriate growth signals (such as oncogenic stress) that occur in a stochastic fashion, possibly as a result of random mutations. The ARF tumor suppressor seems to fulfill that role, as its expression is near undetectable in normal tissues, yet is potently induced by oncogenic stress (such as overexpression of oncogenic Ras or myc). As a result of induced expression of ARF, the tumor suppressor protein p53 is stabilized and promotes cell cycle arrest. Mutations or epigenetic alterations of the INK4a/Arf locus are second only to p53 mutations in cancer cells, and in some cancers, alterations in both Arf and p53 observed, suggesting that these two tumor suppressors act coordinately to prevent unwarranted cell growth and proliferation. The aim of this review is to characterize the current knowledge in the field about both p53-dependent and independent functions of ARF as well as to summarize the present models for how ARF might control rates of cell proliferation and/or macromolecular synthesis. We will discuss potential therapeutic targets in the ARF pathway, and some preliminary attempts at enhancing or restoring the activity of this important tumor suppressor.


Subject(s)
ADP-Ribosylation Factor 1/drug effects , ADP-Ribosylation Factor 1/physiology , Antineoplastic Agents/pharmacology , Genes, Tumor Suppressor/drug effects , Genes, Tumor Suppressor/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , ADP-Ribosylation Factor 1/genetics , Animals , Cell Nucleolus/genetics , Cell Nucleolus/physiology , Genes, p53/physiology , Humans , Neoplasms/genetics , Neoplasms/physiopathology , Peptides/chemical synthesis , Peptides/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Ribosomes/physiology , Signal Transduction/genetics
19.
Cancer Res ; 67(4): 1609-17, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17308101

ABSTRACT

Nucleophosmin (B23) is a nucleolar phosphoprotein that has been implicated in numerous cellular processes. In particular, nucleophosmin interacts with nucleolar components of newly synthesized ribosomes to promote ribosome nuclear export. Nucleophosmin is a classic mitogen-induced protein, with changes in its expression correlating with growth factor stimulation. In this study, we examined the underlying mechanism of nucleophosmin induction and showed that hyperproliferative signals emanating from oncogenic H-Ras(V12) cause tremendous increases in nucleophosmin protein expression. Nucleophosmin protein accumulation was dependent on mammalian target of rapamycin (mTOR) activation, as rapamycin completely prevented nucleophosmin induction. Consistent with this finding, genetic ablation of Tsc1, a major upstream inhibitor of mTOR, resulted in nucleophosmin protein induction through increased translation of existing nucleophosmin mRNAs. Increases in nucleophosmin protein accumulation were suppressed by reintroduction of TSC1. Induction of nucleophosmin through Tsc1 loss resulted in a greater pool of actively translating ribosomes in the cytoplasm, higher overall rates of protein synthesis, and increased cell proliferation, all of which were dependent on efficient nucleophosmin nuclear export. Nucleophosmin protein accumulation in the absence of Tsc1 promoted the nuclear export of maturing ribosome subunits, providing a mechanistic link between TSC1/mTOR signaling, nucleophosmin-mediated nuclear export of ribosome subunits, protein synthesis levels, and cell growth.


Subject(s)
Nuclear Proteins/metabolism , Ribosomes/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Chromones/pharmacology , Humans , Mice , Morpholines/pharmacology , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nucleophosmin , Platelet-Derived Growth Factor/pharmacology , Protein Biosynthesis , Protein Kinases/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , ras Proteins/metabolism
20.
J Immunol ; 177(5): 3413-20, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16920983

ABSTRACT

In response to virus infection or treatment with dsRNA, macrophages express the inducible form of cyclooxygenase-2 (COX-2) and produce proinflammatory prostaglandins. Recently, we have shown that NF-kappaB is required for encephalomyocarditis virus (EMCV)- and dsRNA-stimulated COX-2 expression in mouse macrophages. The dsRNA-dependent protein kinase R is not required for EMCV-stimulated COX-2 expression, suggesting the presence of protein kinase R-independent pathways in the regulation of this antiviral gene. In this study, the role of MAPK in the regulation of macrophage expression of cyclooxygenase-2 (COX)-2 in response to EMCV infection was examined. Treatment of mouse macrophages or RAW-264.7 cells with dsRNA or infection with EMCV stimulates the rapid activation of the MAPKs p38, JNK, and ERK. Inhibition of p38 and JNK activity results in attenuation while ERK inhibition does not modulate dsRNA- and EMCV-induced COX-2 expression and PGE2 production by macrophages. JNK and p38 appear to selectively regulate COX-2 expression, as inhibition of either kinase fails to prevent dsRNA- or EMCV-stimulated inducible NO synthase expression by macrophages. Using macrophages isolated from TLR3-deficient mice, we show that p38 and JNK activation and COX-2 expression in response to EMCV or poly(IC) does not require the presence the dsRNA receptor TLR3. These findings support a role for p38 and JNK in the selective regulation of COX-2 expression by macrophages in response to virus infection.


Subject(s)
Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Cyclooxygenase 2/metabolism , Encephalomyocarditis virus/physiology , Macrophages/enzymology , Mitogen-Activated Protein Kinases/metabolism , Animals , Cardiovirus Infections/virology , Cells, Cultured , Cyclooxygenase 2/genetics , Dinoprostone/biosynthesis , Enzyme Activation , Gene Expression Regulation, Enzymologic/genetics , MAP Kinase Signaling System , Macrophages/drug effects , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/metabolism , Nitric Oxide Synthase Type II/metabolism , Phosphorylation , Poly I-C/pharmacology , RNA, Double-Stranded/genetics , Toll-Like Receptor 3/deficiency , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...