Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Autophagy ; 19(11): 2912-2933, 2023 11.
Article in English | MEDLINE | ID: mdl-37459465

ABSTRACT

ABBREVIATIONS: ATG4 (autophagy related 4 cysteine peptidase); ATG4A (autophagy related 4A cysteine peptidase); ATG4B (autophagy related 4B cysteine peptidase); ATG4C (autophagy related 4C cysteine peptidase); ATG4D (autophagy related 4D cysteine peptidase); Atg8 (autophagy related 8); GABARAP (GABA type A receptor-associated protein); GABARAPL1(GABA type A receptor-associated protein like 1); GABARAPL2 (GABA type A receptor-associated protein like 2); MAP1LC3A/LC3A (microtubule associated protein 1 light chain 3 alpha); MAP1LC3B/LC3B (microtubule associated protein 1 light chain 3 beta); mATG8 (mammalian Atg8); PE (phosphatidylethanolamine); PS (phosphatydylserine); SQSTM1/p62 (sequestosome 1).


Subject(s)
Autophagy-Related Proteins , Autophagy , Animals , Autophagy-Related Proteins/metabolism , Cysteine , gamma-Aminobutyric Acid , Mammals/metabolism , Microtubule-Associated Proteins/metabolism , Mice
2.
Cell Death Dis ; 14(1): 60, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36702832

ABSTRACT

The spindle assembly checkpoint (SAC) is an essential mechanism that ensures the accurate chromosome segregation during mitosis, thus preventing genomic instability. Deubiquitinases have emerged as key regulators of the SAC, mainly by determining the fate of proteins during cell cycle progression. Here, we identify USP49 deubiquitinase as a novel regulator of the spindle checkpoint. We show that loss of USP49 in different cancer cell lines impairs proliferation and increases aneuploidy. In addition, USP49-depleted cells overcome the arrest induced by the SAC in the presence of nocodazole. Finally, we report new binding partners of USP49, including ribophorin 1, USP44, and different centrins.


Subject(s)
M Phase Cell Cycle Checkpoints , Spindle Apparatus , Humans , Spindle Apparatus/metabolism , Aneuploidy , Mitosis , Deubiquitinating Enzymes/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism
3.
EMBO J ; 41(21): e110727, 2022 11 02.
Article in English | MEDLINE | ID: mdl-36124427

ABSTRACT

Better understanding on interactions between SARS-CoV-2 and host cells should help to identify host factors that may be targetable to combat infection and COVID-19 pathology. To this end, we have conducted a genome-wide CRISPR/Cas9-based loss-of-function screen in human lung cancer cells infected with SARS-CoV-2-pseudotyped lentiviruses. Our results recapitulate many findings from previous screens that used full SARS-CoV-2 viruses, but also unveil two novel critical host factors: the lysosomal efflux transporter SPNS1 and the plasma and lysosomal membrane protein PLAC8. Functional experiments with full SARS-CoV-2 viruses confirm that loss-of-function of these genes impairs viral entry. We find that PLAC8 is a key limiting host factor, whose overexpression boosts viral infection in eight different human lung cancer cell lines. Using single-cell RNA-Seq data analyses, we demonstrate that PLAC8 is highly expressed in ciliated and secretory cells of the respiratory tract, as well as in gut enterocytes, cell types that are highly susceptible to SARS-CoV-2 infection. Proteomics and cell biology studies suggest that PLAC8 and SPNS1 regulate the autophagolysosomal compartment and affect the intracellular fate of endocytosed virions.


Subject(s)
COVID-19 , Lung Neoplasms , Humans , SARS-CoV-2 , Angiotensin-Converting Enzyme 2 , Lysosomal Membrane Proteins , Autophagy , Proteins
4.
Proc Natl Acad Sci U S A ; 119(36): e2118763119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037356

ABSTRACT

Turritopsis dohrnii is the only metazoan able to rejuvenate repeatedly after its medusae reproduce, hinting at biological immortality and challenging our understanding of aging. We present and compare whole-genome assemblies of T. dohrnii and the nonimmortal Turritopsis rubra using automatic and manual annotations, together with the transcriptome of life cycle reversal (LCR) process of T. dohrnii. We have identified variants and expansions of genes associated with replication, DNA repair, telomere maintenance, redox environment, stem cell population, and intercellular communication. Moreover, we have found silencing of polycomb repressive complex 2 targets and activation of pluripotency targets during LCR, which points to these transcription factors as pluripotency inducers in T. dohrnii. Accordingly, we propose these factors as key elements in the ability of T. dohrnii to undergo rejuvenation.


Subject(s)
Hydrozoa , Rejuvenation , Animals , Genomics , Hydrozoa/genetics , Hydrozoa/growth & development , Life Cycle Stages/genetics , Transcriptome
5.
Cell Death Differ ; 28(9): 2651-2672, 2021 09.
Article in English | MEDLINE | ID: mdl-33795848

ABSTRACT

Despite the great advances in autophagy research in the last years, the specific functions of the four mammalian Atg4 proteases (ATG4A-D) remain unclear. In yeast, Atg4 mediates both Atg8 proteolytic activation, and its delipidation. However, it is not clear how these two roles are distributed along the members of the ATG4 family of proteases. We show that these two functions are preferentially carried out by distinct ATG4 proteases, being ATG4D the main delipidating enzyme. In mammalian cells, ATG4D loss results in accumulation of membrane-bound forms of mATG8s, increased cellular autophagosome number and reduced autophagosome average size. In mice, ATG4D loss leads to cerebellar neurodegeneration and impaired motor coordination caused by alterations in trafficking/clustering of GABAA receptors. We also show that human gene variants of ATG4D associated with neurodegeneration are not able to fully restore ATG4D deficiency, highlighting the neuroprotective role of ATG4D in mammals.


Subject(s)
Autophagy-Related Protein 8 Family/metabolism , Autophagy-Related Proteins/metabolism , Cysteine Endopeptidases/metabolism , Neurodegenerative Diseases/genetics , Amino Acid Sequence , Animals , Autophagy , Disease Models, Animal , Humans , Mammals , Mice , Mice, Transgenic , Neurodegenerative Diseases/pathology
7.
Leukemia ; 34(11): 3007-3018, 2020 11.
Article in English | MEDLINE | ID: mdl-32475991

ABSTRACT

Multiple myeloma (MM) patients undergo repetitive bone marrow (BM) aspirates for genetic characterization. Circulating tumor cells (CTCs) are detectable in peripheral blood (PB) of virtually all MM cases and are prognostic, but their applicability for noninvasive screening has been poorly investigated. Here, we used next-generation flow (NGF) cytometry to isolate matched CTCs and BM tumor cells from 53 patients and compared their genetic profile. In eight cases, tumor cells from extramedullary (EM) plasmacytomas were also sorted and whole-exome sequencing was performed in the three spatially distributed tumor samples. CTCs were detectable by NGF in the PB of all patients with MM. Based on the cancer cell fraction of clonal and subclonal mutations, we found that ~22% of CTCs egressed from a BM (or EM) site distant from the matched BM aspirate. Concordance between BM tumor cells and CTCs was high for chromosome arm-level copy number alterations (≥95%) though not for translocations (39%). All high-risk genetic abnormalities except one t(4;14) were detected in CTCs whenever present in BM tumor cells. Noteworthy, ≥82% mutations present in BM and EM clones were detectable in CTCs. Altogether, these results support CTCs for noninvasive risk-stratification of MM patients based on their numbers and genetic profile.


Subject(s)
Biomarkers, Tumor , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Neoplastic Cells, Circulating/metabolism , Neoplastic Cells, Circulating/pathology , Computational Biology/methods , DNA Copy Number Variations , DNA Mutational Analysis , Female , Genetic Heterogeneity , Humans , Immunophenotyping , Liquid Biopsy , Male , Mutation , Neoplasm Staging , Prognosis , Recurrence , Whole Genome Sequencing
8.
Cell Rep ; 31(1): 107473, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32268098

ABSTRACT

Ribosomes undergo multiple conformational transitions during translation elongation. Here, we report the high-resolution cryoelectron microscopy (cryo-EM) structure of the human 80S ribosome in the post-decoding pre-translocation state (classical-PRE) at 3.3-Å resolution along with the rotated (hybrid-PRE) and the post-translocation states (POST). The classical-PRE state ribosome structure reveals a previously unobserved interaction between the C-terminal region of the conserved ribosomal protein uS19 and the A- and P-site tRNAs and the mRNA in the decoding site. In addition to changes in the inter-subunit bridges, analysis of different ribosomal conformations reveals the dynamic nature of this domain and suggests a role in tRNA accommodation and translocation during elongation. Furthermore, we show that disease-associated mutations in uS19 result in increased frameshifting. Together, this structure-function analysis provides mechanistic insights into the role of the uS19 C-terminal tail in the context of mammalian ribosomes.


Subject(s)
Peptide Chain Elongation, Translational/genetics , Ribosomal Proteins/genetics , Ribosomes/metabolism , Cryoelectron Microscopy/methods , Humans , Models, Molecular , Molecular Conformation , Peptide Chain Elongation, Translational/physiology , Protein Biosynthesis/genetics , RNA, Messenger/metabolism , Ribosomal Proteins/metabolism , Ribosomal Proteins/ultrastructure , Ribosomes/ultrastructure
9.
J Biol Chem ; 295(7): 2001-2017, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31919096

ABSTRACT

The MAX network transcriptional repressor (MNT) is an MXD family transcription factor of the basic helix-loop-helix (bHLH) family. MNT dimerizes with another transcriptional regulator, MYC-associated factor X (MAX), and down-regulates genes by binding to E-boxes. MAX also dimerizes with MYC, an oncogenic bHLH transcription factor. Upon E-box binding, the MYC-MAX dimer activates gene expression. MNT also binds to the MAX dimerization protein MLX (MLX), and MNT-MLX and MNT-MAX dimers co-exist. However, all MNT functions have been attributed to MNT-MAX dimers, and no functions of the MNT-MLX dimer have been described. MNT's biological role has been linked to its function as a MYC oncogene modulator, but little is known about its regulation. We show here that MNT localizes to the nucleus of MAX-expressing cells and that MNT-MAX dimers bind and repress the MNT promoter, an effect that depends on one of the two E-boxes on this promoter. In MAX-deficient cells, MNT was overexpressed and redistributed to the cytoplasm. Interestingly, MNT was required for cell proliferation even in the absence of MAX. We show that in MAX-deficient cells, MNT binds to MLX, but also forms homodimers. RNA-sequencing experiments revealed that MNT regulates the expression of several genes even in the absence of MAX, with many of these genes being involved in cell cycle regulation and DNA repair. Of note, MNT-MNT homodimers regulated the transcription of some genes involved in cell proliferation. The tight regulation of MNT and its functionality even without MAX suggest a major role for MNT in cell proliferation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Repressor Proteins/genetics , Transcription, Genetic , Amino Acid Sequence/genetics , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/chemistry , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Gene Regulatory Networks/genetics , Helix-Loop-Helix Motifs/genetics , Humans , Promoter Regions, Genetic , Protein Multimerization/genetics , Proto-Oncogene Proteins c-myc/chemistry , Proto-Oncogene Proteins c-myc/genetics , Repressor Proteins/chemistry
10.
Sci Rep ; 9(1): 18693, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31822694

ABSTRACT

Cell cycle stimulation is a major transforming mechanism of Myc oncoprotein. This is achieved through at least three concomitant mechanisms: upregulation of cyclins and Cdks, downregulation of the Cdk inhibitors p15 and p21 and the degradation of p27. The Myc-p27 antagonism has been shown to be relevant in human cancer. To be degraded, p27 must be phosphorylated at Thr-187 to be recognized by Skp2, a component of the ubiquitination complex. We previously described that Myc induces Skp2 expression. Here we show that not only Cdk2 but Cdk1 phosphorylates p27 at the Thr-187. Moreover, Myc induced p27 degradation in murine fibroblasts through Cdk1 activation, which was achieved by Myc-dependent cyclin A and B induction. In the absence of Cdk2, p27 phosphorylation at Thr-187 was mainly carried out by cyclin A2-Cdk1 and cyclin B1-Cdk1. We also show that Cdk1 inhibition was enough for the synthetic lethal interaction with Myc. This result is relevant because Cdk1 is the only Cdk strictly required for cell cycle and the reported synthetic lethal interaction between Cdk1 and Myc.


Subject(s)
CDC2 Protein Kinase/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Animals , CDC2 Protein Kinase/physiology , Cell Cycle , Cell Cycle Checkpoints , Cell Cycle Proteins/metabolism , Cell Division , Cell Line , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/physiology , Cyclin-Dependent Kinases/metabolism , Cyclins/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, Inbred C57BL , Phosphorylation , Proto-Oncogene Proteins c-myc/physiology , Signal Transduction
11.
Front Immunol ; 10: 2455, 2019.
Article in English | MEDLINE | ID: mdl-31681329

ABSTRACT

B-cell receptor (BCR)-dependent signaling is central for leukemia B-cell homeostasis, as underscored by the promising clinical results obtained in patients with chronic lymphocytic leukemia (CLL) treated with novel agents targeting components of this pathway. Herein, we demonstrate that the mithralog EC-7072 displays high ex vivo cytotoxic activity against leukemia cells from CLL patients independently from high-risk prognostic markers and IGHV mutational status. EC-7072 was significantly less toxic against T cells and NK cells and did not alter the production of the immune effector molecules IFN-γ and perforin. EC-7072 directly triggered caspase-3-dependent CLL cell apoptosis, which was not abrogated by microenvironment-derived factors that sustain leukemia cell survival. RNA-sequencing analyses revealed a dramatic EC-7072-driven reprograming of the transcriptome of CLL cells, including a wide downregulation of multiple components and targets of the BCR signaling pathway. Accordingly, we found decreased levels of phosphorylated signaling nodes downstream of the BCR. Crosslinking-mediated BCR activation antagonized CLL cell death triggered by EC-7072, increased the phosphorylation levels of the abovementioned signaling nodes and upregulated BCL2 expression, suggesting that the mithralog disrupts CLL cell viability by targeting the BCR signaling axis at multiple levels. EC-7072 exerted similar or higher antileukemic activity than that of several available CLL therapies and displayed additive or synergistic interaction with these drugs in killing CLL cells. Overall, our findings provide rationale for future investigation to test whether EC-7072 may be a potential therapeutic option for patients with CLL and other B-cell malignancies.


Subject(s)
Apoptosis/drug effects , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Plicamycin/analogs & derivatives , Receptors, Antigen, B-Cell/antagonists & inhibitors , Signal Transduction/drug effects , Antibiotics, Antineoplastic/pharmacology , Caspase 3/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Leukemic/drug effects , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Phosphorylation/drug effects , Plicamycin/pharmacology , Receptors, Antigen, B-Cell/genetics , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Tumor Microenvironment/drug effects
12.
Nat Ecol Evol ; 3(1): 87-95, 2019 01.
Article in English | MEDLINE | ID: mdl-30510174

ABSTRACT

Giant tortoises are among the longest-lived vertebrate animals and, as such, provide an excellent model to study traits like longevity and age-related diseases. However, genomic and molecular evolutionary information on giant tortoises is scarce. Here, we describe a global analysis of the genomes of Lonesome George-the iconic last member of Chelonoidis abingdonii-and the Aldabra giant tortoise (Aldabrachelys gigantea). Comparison of these genomes with those of related species, using both unsupervised and supervised analyses, led us to detect lineage-specific variants affecting DNA repair genes, inflammatory mediators and genes related to cancer development. Our study also hints at specific evolutionary strategies linked to increased lifespan, and expands our understanding of the genomic determinants of ageing. These new genome sequences also provide important resources to help the efforts for restoration of giant tortoise populations.


Subject(s)
Aging/genetics , Genome , Turtles/genetics , Animals , DNA Repair/genetics , Evolution, Molecular , HEK293 Cells , Humans , Inflammation Mediators , Male , Neoplasms/genetics , Phylogeny , Population Density
13.
Blood ; 132(22): 2375-2388, 2018 11 29.
Article in English | MEDLINE | ID: mdl-30181176

ABSTRACT

Genomic studies have recently identified RPS15 as a new driver gene in aggressive and chemorefractory cases of chronic lymphocytic leukemia (CLL). RPS15 encodes a ribosomal protein whose conserved C-terminal domain extends into the decoding center of the ribosome. We demonstrate that mutations in highly conserved residues of this domain affect protein stability, by increasing its ubiquitin-mediated degradation, and cell-proliferation rates. On the other hand, we show that mutated RPS15 can be loaded into the ribosomes, directly impacting on global protein synthesis and/or translational fidelity in a mutation-specific manner. Quantitative mass spectrometry analyses suggest that RPS15 variants may induce additional alterations in the translational machinery, as well as a metabolic shift at the proteome level in HEK293T and MEC-1 cells. These results indicate that CLL-related RPS15 mutations might act following patterns known for other ribosomal diseases, likely switching from a hypo- to a hyperproliferative phenotype driven by mutated ribosomes. In this scenario, loss of translational fidelity causing altered cell proteostasis can be proposed as a new molecular mechanism involved in CLL pathobiology.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Ribosomal Proteins/genetics , Ribosomes/genetics , Cell Line, Tumor , Cohort Studies , HEK293 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mutation Rate , Point Mutation , Protein Biosynthesis , Protein Domains , Ribosomal Proteins/chemistry , Ribosomes/pathology
14.
Int Immunol ; 28(3): 127-37, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26507128

ABSTRACT

Modulation of TCR signaling upon ligand binding is achieved by changes in the equilibrium between TCR degradation, recycling and synthesis; surprisingly, the molecular mechanism of such an important process is not fully understood. Here, we describe the role of a new player in the mediation of TCR degradation: the endocytic adaptor Numb. Our data show that Numb inhibition leads to abnormal intracellular distribution and defective TCR degradation in mature T lymphocytes. In addition, we find that Numb simultaneously binds to both Cbl and a site within CD3ε that overlaps with the Nck binding site. As a result, Cbl couples specifically to the CD3ε chain to mediate TCR degradation. The present study unveils a novel role of Numb that lies at the heart of TCR signaling initiation and termination.


Subject(s)
CD3 Complex/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proteolysis , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , HEK293 Cells , Humans , Membrane Proteins/genetics , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Oncogene Proteins/metabolism , Protein Binding/genetics , Protein Transport/genetics , Proto-Oncogene Proteins c-cbl/metabolism , Sequence Deletion/genetics
15.
Blood ; 126(2): 195-202, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26031915

ABSTRACT

Great progress has recently been achieved in the understanding of the genomic alterations driving chronic lymphocytic leukemia (CLL). Nevertheless, the specific molecular mechanisms governing chromatin remodeling in CLL are unknown. Here we report the genetic and functional characterization of somatic mutations affecting the chromatin remodeler CHD2, one of the most frequently mutated genes in CLL (5.3%) and in monoclonal B lymphocytosis (MBL, 7%), a B-cell expansion that can evolve to CLL. Most of the mutations affecting CHD2, identified by whole-exome sequencing of 456 CLL and 43 MBL patients, are either truncating or affect conserved residues in functional domains, thus supporting a putative role for CHD2 as a tumor suppressor gene. CHD2 mutants show altered nuclear distribution, and a chromodomain helicase DNA binding protein 2 (CHD2) mutant affected in its DNA-binding domain exhibits defective association with active chromatin. Clinicobiological analyses show that most CLL patients carrying CHD2 mutations also present mutated immunoglobulin heavy chain variable region genes (IGHVs), being the most frequently mutated gene in this prognostic subgroup. This is the first study providing functional evidence supporting CHD2 as a cancer driver and opens the way to further studies of the role of this chromatin remodeler in CLL.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Chromatin Assembly and Disassembly/genetics , Cohort Studies , HEK293 Cells , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Molecular Sequence Data , Sequence Homology, Amino Acid
16.
Int J Hematol ; 101(3): 219-28, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25630433

ABSTRACT

In spite of being the most prevalent adult leukemia in Western countries, the molecular mechanisms driving the establishment and progression of chronic lymphocytic leukemia (CLL) remain largely unknown. In recent years, the use of next-generation sequencing techniques has uncovered new and, in some cases, unexpected driver genes with prognostic and therapeutic value. The mutational landscape of CLL is characterized by high-genetic and epigenetic heterogeneity, low mutation recurrence and a long tail of cases with undefined driver genes. On the other hand, the use of deep sequencing has also revealed high intra-tumor heterogeneity and provided a detailed picture of clonal evolution processes. This phenomenon, in which aberrant DNA methylation can also participate, appears to be tightly associated to poor outcomes and chemo-refractoriness, thus providing a new subject for therapeutic intervention. Hence, and having in mind the limitations derived from the CLL complexity thus described, the application of massively parallel sequencing studies has unveiled a wealth of information that is expected to substantially improve patient staging schemes and CLL clinical management.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Animals , Chromosome Aberrations , Clonal Evolution , DNA Methylation , Epigenesis, Genetic , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Mutation
17.
Biochim Biophys Acta ; 1849(5): 506-16, 2015 May.
Article in English | MEDLINE | ID: mdl-24704206

ABSTRACT

Soon after the discovery of the Myc gene (c-Myc), it became clear that Myc expression levels tightly correlate to cell proliferation. The entry in cell cycle of quiescent cells upon Myc enforced expression has been described in many models. Also, the downregulation or inactivation of Myc results in the impairment of cell cycle progression. Given the frequent deregulation of Myc oncogene in human cancer it is important to dissect out the mechanisms underlying the role of Myc on cell cycle control. Several parallel mechanisms account for Myc-mediated stimulation of the cell cycle. First, most of the critical positive cell cycle regulators are encoded by genes induced by Myc. These Myc target genes include Cdks, cyclins and E2F transcription factors. Apart from its direct effects on the transcription, Myc is able to hyperactivate cyclin/Cdk complexes through the induction of Cdk activating kinase (CAK) and Cdc25 phosphatases. Moreover, Myc antagonizes the activity of cell cycle inhibitors as p21 and p27 through different mechanisms. Thus, Myc is able to block p21 transcription or to induce Skp2, a protein involved in p27 degradation. Finally, Myc induces DNA replication by binding to replication origins and by upregulating genes encoding proteins required for replication initiation. Myc also regulates genes involved in the mitotic control. A promising approach to treat tumors with deregulated Myc is the synthetic lethality based on the inhibition of Cdks. Thus, the knowledge of the Myc-dependent cell cycle regulatory mechanisms will help to discover new therapeutic approaches directed against malignancies with deregulated Myc. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.


Subject(s)
Cell Cycle Checkpoints/genetics , DNA Replication/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-myc/genetics , Cell Proliferation/genetics , Cyclin-Dependent Kinases/genetics , Cyclins/genetics , Gene Expression Regulation, Neoplastic , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Proto-Oncogene Proteins c-myc/biosynthesis , p21-Activated Kinases/genetics
18.
Oncotarget ; 5(13): 4694-708, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25051361

ABSTRACT

Myc (c-Myc) counteracts p27 effects, and low p27 usually correlates with high Myc expression in human cancer. However there is no information on the co-expression of both genes in chronic lymphocytic leukemia (CLL). We found a lack of correlation between RNA and protein levels of p27 and Myc in CLL cells, so we determined the protein levels by immunoblot in 107 cases of CLL. We observed a high p27 protein expression in CLL compared to normal B cells. Ectopic p27 expression in a CLL-derived cell line resulted in cell death resistance. Surprisingly, Myc expression was very low or undetectable in most CLL cases analyzed, with a clear correlation between high p27 and low Myc protein levels. This was associated with low Skp2 expression, which is consistent with the Skp2 role in p27 degradation and with SKP2 being a Myc target gene. High Myc expression did not correlate with leukemia progression, despite that cell cycle-related Myc target genes were upregulated. However, biochemical analysis showed that the high p27 levels inhibited cyclin-Cdk complexes even in Myc expressing CLL cells. Our data suggest that the combination of high p27 and low Myc is a marker of CLL cells which is mediated by Skp2.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p27/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Proto-Oncogene Proteins c-myb/metabolism , S-Phase Kinase-Associated Proteins/metabolism , Adult , Aged , Aged, 80 and over , Apoptosis/genetics , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclins/genetics , Cyclins/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Leukemic , Humans , Immunoblotting , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Microscopy, Fluorescence , Middle Aged , Proto-Oncogene Proteins c-myb/genetics , Reverse Transcriptase Polymerase Chain Reaction , S-Phase Kinase-Associated Proteins/genetics
19.
PLoS One ; 7(5): e37759, 2012.
Article in English | MEDLINE | ID: mdl-22662213

ABSTRACT

It has been previously described that p21 functions not only as a CDK inhibitor but also as a transcriptional co-repressor in some systems. To investigate the roles of p21 in transcriptional control, we studied the gene expression changes in two human cell systems. Using a human leukemia cell line (K562) with inducible p21 expression and human primary keratinocytes with adenoviral-mediated p21 expression, we carried out microarray-based gene expression profiling. We found that p21 rapidly and strongly repressed the mRNA levels of a number of genes involved in cell cycle and mitosis. One of the most strongly down-regulated genes was CCNE2 (cyclin E2 gene). Mutational analysis in K562 cells showed that the N-terminal region of p21 is required for repression of gene expression of CCNE2 and other genes. Chromatin immunoprecipitation assays indicated that p21 was bound to human CCNE2 and other p21-repressed genes gene in the vicinity of the transcription start site. Moreover, p21 repressed human CCNE2 promoter-luciferase constructs in K562 cells. Bioinformatic analysis revealed that the CDE motif is present in most of the promoters of the p21-regulated genes. Altogether, the results suggest that p21 exerts a repressive effect on a relevant number of genes controlling S phase and mitosis. Thus, p21 activity as inhibitor of cell cycle progression would be mediated not only by the inhibition of CDKs but also by the transcriptional down-regulation of key genes.


Subject(s)
Co-Repressor Proteins/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Mitosis/genetics , S Phase/genetics , Transcription, Genetic , Cell Line , Cluster Analysis , Computational Biology/methods , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p21/chemistry , Cyclins/genetics , Gene Expression Profiling , Gene Expression Regulation , Humans , K562 Cells , Keratinocytes/metabolism , Promoter Regions, Genetic , Protein Binding
20.
J Biol Chem ; 287(32): 26495-505, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22718761

ABSTRACT

Testicular germ cell tumors (TGCTs) are highly responsive to and curable by cisplatin-based chemotherapy even in advanced stages. We have studied the molecular mechanisms involved in the induction of apoptosis in response to cisplatin, and found that proapoptotic Noxa is transcriptionally up-regulated following cisplatin exposure, even in the absence of p53, in NTERA2 cisplatin-sensitive cells but not in 1411HP-resistant cells. Blockade of Noxa reduced the apoptotic response of embryonal carcinoma (EC) NTERA2 cells to cisplatin. A detailed analysis of the Noxa promoter revealed that p73 and Sp1-like factors, Sp1 and KLF6, played key roles in the transcriptional control of this gene. Overexpression of TAp73 induced Noxa whereas the dominant negative isoform ΔNp73, reduced the levels of Noxa after cisplatin exposure in NTERA2 and 2102EP. Interestingly, down-regulation of Sp1 increased Noxa expression in response to cisplatin. However, blockade of KLF6 decreased cisplatin-induced up-regulation of Noxa in EC cell lines. In addition, tissue microarray analyses of TGCTs revealed that expression of Noxa correlates with good clinical prognosis in patients with embryonal carcinoma. Thus, our data show the transcriptional network that regulates Noxa in EC cells, which is key for their apoptotic response to cisplatin-based chemotherapy, and propose Noxa as a predictive factor of therapeutic response.


Subject(s)
Apoptosis/physiology , Carcinoma, Embryonal/pathology , Cisplatin/pharmacology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic/physiology , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Sp1 Transcription Factor/physiology , Testicular Neoplasms/pathology , Tumor Suppressor Proteins/physiology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Base Sequence , Carcinoma, Embryonal/drug therapy , Carcinoma, Embryonal/genetics , Cell Line, Tumor , Cisplatin/therapeutic use , Cohort Studies , DNA Primers , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Prognosis , Promoter Regions, Genetic , Real-Time Polymerase Chain Reaction , Testicular Neoplasms/drug therapy , Testicular Neoplasms/genetics , Tumor Protein p73
SELECTION OF CITATIONS
SEARCH DETAIL