Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Gastrointest Cancer ; 52(4): 1356-1369, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34738187

ABSTRACT

PURPOSE: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide with lack of effective systemic chemotherapy. In this study, we aimed to evaluate the value of ATPase family AAA domain-containing protein 2 (ATAD2) as a biomarker and potential therapeutic target for HCC. METHODS: The expression of ATAD2 was tested in different HCC patient cohorts by immunohistochemistry and comparative transcriptional analysis. The co-expression of ATAD2 and proliferation markers was compared during liver regeneration and malignancy with different bioinformatics tools. The cellular effects of ATAD2 inactivation in liver malignancy was tested on cell cycle, apoptosis, and colony formation ability as well as tumor formation using RNA interference. The genes affected by ATAD2 inactivation in three different HCC cell lines were identified by global gene expression profiling and bioinformatics tools. RESULTS: ATAD2 overexpression is closely correlated with HCC tumor stage. There was gradual increase from dysplasia, well-differentiated and poorly-differentiated HCC, respectively. We also observed transient upregulation of ATAD2 expression during rat liver regeneration in parallel to changes in Ki-67 expression. ATAD2 knockdown resulted in apoptosis and decreased cell survival in vitro and decreased tumor formation in some HCC cell lines. However, three other HCC cell lines tested were not affected. Similarly, gene expression response to ATAD2 inactivation in different HCC cell lines was highly heterogeneous. CONCLUSIONS: ATAD2 is a potential proliferation marker for liver regeneration and HCC. It may also serve as a therapeutic target despite heterogeneous response of malignant cells.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , ATPases Associated with Diverse Cellular Activities/metabolism , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Ki-67 Antigen/metabolism , Liver Neoplasms/pathology , Rats
2.
Cancer Cell ; 31(4): 576-590.e8, 2017 04 10.
Article in English | MEDLINE | ID: mdl-28399412

ABSTRACT

Cyclins and cyclin-dependent kinases (CDKs) are hyperactivated in numerous human tumors. To identify means of interfering with cyclins/CDKs, we performed nine genome-wide screens for human microRNAs (miRNAs) directly regulating cell-cycle proteins. We uncovered a distinct class of miRNAs that target nearly all cyclins/CDKs, which are very effective in inhibiting cancer cell proliferation. By profiling the response of over 120 human cancer cell lines, we derived an expression-based algorithm that can predict the response of tumors to cell-cycle-targeting miRNAs. Using systemic administration of nanoparticle-formulated miRNAs, we inhibited tumor progression in seven mouse xenograft models, including three treatment-refractory patient-derived tumors, without affecting normal tissues. Our results highlight the utility of using cell-cycle-targeting miRNAs for treatment of refractory cancer types.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , 3' Untranslated Regions , Algorithms , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Line, Tumor , Drug Delivery Systems/methods , Female , Genome-Wide Association Study , Humans , Mice, Inbred Strains , MicroRNAs/administration & dosage , MicroRNAs/pharmacology , Mutation , Nanoparticles , Proto-Oncogene Proteins p21(ras)/genetics , Xenograft Model Antitumor Assays
3.
Cell Discov ; 2: 16030, 2016.
Article in English | MEDLINE | ID: mdl-27672444

ABSTRACT

Loss of PTEN, a negative regulator of the phosphoinositide 3-kinase signaling pathway, is a frequent event in T-cell acute lymphoblastic leukemia, suggesting the importance of phosphoinositide 3-kinase activity in this disease. Indeed, hyperactivation of the phosphoinositide 3-kinase pathway is associated with the disease aggressiveness, poor prognosis and resistance to current therapies. To identify a molecular pathway capable of cooperating with PTEN deficiency to drive oncogenic transformation of leukocytes, we performed an unbiased transformation screen with a library of tyrosine kinases. We found that activation of NTRK2 is able to confer a full growth phenotype of Ba/F3 cells in an IL3-independent manner in the PTEN-null setting. NTRK2 activation cooperates with PTEN deficiency through engaging both phosphoinositide3-kinase/AKT and JAK/STAT3 pathway activation in leukocytes. Notably, pharmacological inhibition demonstrated that p110α and p110δ are the major isoforms mediating the phosphoinositide 3-kinase/AKT signaling driven by NTRK2 activation in PTEN-deficient leukemia cells. Furthermore, combined inhibition of phosphoinositide 3-kinase and STAT3 significantly suppressed proliferation of PTEN-mutant T-cell acute lymphoblastic leukemia both in culture and in mouse xenografts. Together, our data suggest that a unique conjunction of PTEN deficiency and NTRK2 activation in T-cell acute lymphoblastic leukemia, and combined pharmacologic inhibition of phosphoinositide 3-kinase and STAT3 signaling may serve as an effective and durable therapeutic strategy for T-cell acute lymphoblastic leukemia.

4.
Cancer Cell ; 29(3): 255-269, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26977878

ABSTRACT

Using transgenic mouse models, cell line-based functional studies, and clinical specimens, we show that cyclin D1/CDK4 mediate resistance to targeted therapy for HER2-positive breast cancer. This is overcome using CDK4/6 inhibitors. Inhibition of CDK4/6 not only suppresses Rb phosphorylation, but also reduces TSC2 phosphorylation and thus partially attenuates mTORC1 activity. This relieves feedback inhibition of upstream EGFR family kinases, resensitizing tumors to EGFR/HER2 blockade. Consequently, dual inhibition of EGFR/HER2 and CDK4/6 invokes a more potent suppression of TSC2 phosphorylation and hence mTORC1/S6K/S6RP activity. The suppression of both Rb and S6RP enhances G1 arrest and a phenotype resembling cellular senescence. In vivo, CDK4/6 inhibitors sensitize patient-derived xenograft tumors to HER2-targeted therapies and delay tumor recurrence in a transgenic model of HER2-positive breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Disease Models, Animal , ErbB Receptors/metabolism , Female , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Nude , Mice, Transgenic , Multiprotein Complexes/metabolism , Neoplasm Recurrence, Local/mortality , Phosphorylation/drug effects , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism
5.
Nat Commun ; 6: 8501, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26442967

ABSTRACT

The tumour suppressor PTEN, which antagonizes PI3K signalling, is frequently inactivated in haematologic malignancies. In mice, deletion of PTEN in haematopoietic stem cells (HSCs) causes perturbed haematopoiesis, myeloproliferative neoplasia (MPN) and leukaemia. Although the roles of the PI3K isoforms have been studied in PTEN-deficient tumours, their individual roles in PTEN-deficient HSCs are unknown. Here we show that when we delete PTEN in HSCs using the Mx1-Cre system, p110ß ablation prevents MPN, improves HSC function and suppresses leukaemia initiation. Pharmacologic inhibition of p110ß in PTEN-deficient mice recapitulates these genetic findings, but suggests involvement of both Akt-dependent and -independent pathways. Further investigation reveals that a p110ß-Rac signalling loop plays a critical role in PTEN-deficient HSCs. Together, these data suggest that myeloid neoplasia driven by PTEN loss is dependent on p110ß via p110ß-Rac-positive-feedback loop, and that disruption of this loop may offer a new and effective therapeutic strategy for PTEN-deficient leukaemia.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/genetics , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Leukemia/genetics , Myeloproliferative Disorders/genetics , PTEN Phosphohydrolase/genetics , Animals , Blotting, Western , Bone Marrow/metabolism , Carcinogenesis/genetics , Chemotaxis , Flow Cytometry , Immunohistochemistry , Mice , rac GTP-Binding Proteins
6.
Cell ; 163(1): 174-86, 2015 Sep 24.
Article in English | MEDLINE | ID: mdl-26406377

ABSTRACT

Triple-negative breast cancer (TNBC) is a highly aggressive form of breast cancer that exhibits extremely high levels of genetic complexity and yet a relatively uniform transcriptional program. We postulate that TNBC might be highly dependent on uninterrupted transcription of a key set of genes within this gene expression program and might therefore be exceptionally sensitive to inhibitors of transcription. Utilizing kinase inhibitors and CRISPR/Cas9-mediated gene editing, we show here that triple-negative but not hormone receptor-positive breast cancer cells are exceptionally dependent on CDK7, a transcriptional cyclin-dependent kinase. TNBC cells are unique in their dependence on this transcriptional CDK and suffer apoptotic cell death upon CDK7 inhibition. An "Achilles cluster" of TNBC-specific genes is especially sensitive to CDK7 inhibition and frequently associated with super-enhancers. We conclude that CDK7 mediates transcriptional addiction to a vital cluster of genes in TNBC and CDK7 inhibition may be a useful therapy for this challenging cancer.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic , Transcription, Genetic , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cyclin-Dependent Kinases/antagonists & inhibitors , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Cyclin-Dependent Kinase-Activating Kinase
7.
Nat Rev Cancer ; 15(1): 7-24, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25533673

ABSTRACT

Phosphatidylinositol 3-kinases (PI3Ks) are crucial coordinators of intracellular signalling in response to extracellular stimuli. Hyperactivation of PI3K signalling cascades is one of the most common events in human cancers. In this Review, we discuss recent advances in our knowledge of the roles of specific PI3K isoforms in normal and oncogenic signalling, the different ways in which PI3K can be upregulated, and the current state and future potential of targeting this pathway in the clinic.


Subject(s)
Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/physiology , Animals , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Molecular Targeted Therapy , Neoplasms/drug therapy , Phosphatidylinositol Phosphates/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
8.
J Clin Invest ; 124(4): 1794-809, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569456

ABSTRACT

The genes encoding RAS family members are frequently mutated in juvenile myelomonocytic leukemia (JMML) and acute myeloid leukemia (AML). RAS proteins are difficult to target pharmacologically; therefore, targeting the downstream PI3K and RAF/MEK/ERK pathways represents a promising approach to treat RAS-addicted tumors. The p110α isoform of PI3K (encoded by Pik3ca) is an essential effector of oncogenic KRAS in murine lung tumors, but it is unknown whether p110α contributes to leukemia. To specifically examine the role of p110α in murine hematopoiesis and in leukemia, we conditionally deleted p110α in HSCs using the Cre-loxP system. Postnatal deletion of p110α resulted in mild anemia without affecting HSC self-renewal; however, deletion of p110α in mice with KRASG12D-associated JMML markedly delayed their death. Furthermore, the p110α-selective inhibitor BYL719 inhibited growth factor-independent KRASG12D BM colony formation and sensitized cells to a low dose of the MEK inhibitor MEK162. Furthermore, combined inhibition of p110α and MEK effectively reduced proliferation of RAS-mutated AML cell lines and disease in an AML murine xenograft model. Together, our data indicate that RAS-mutated myeloid leukemias are dependent on the PI3K isoform p110α, and combined pharmacologic inhibition of p110α and MEK could be an effective therapeutic strategy for JMML and AML.


Subject(s)
Genes, ras , Hematopoiesis/genetics , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Juvenile/enzymology , Leukemia, Myelomonocytic, Juvenile/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Animals , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Erythropoiesis/genetics , Erythropoiesis/physiology , Heterografts , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myelomonocytic, Juvenile/pathology , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphatidylinositol 3-Kinases/deficiency , Signal Transduction
9.
PLoS One ; 8(5): e64016, 2013.
Article in English | MEDLINE | ID: mdl-23691139

ABSTRACT

Senescence is a permanent proliferation arrest in response to cell stress such as DNA damage. It contributes strongly to tissue aging and serves as a major barrier against tumor development. Most tumor cells are believed to bypass the senescence barrier (become "immortal") by inactivating growth control genes such as TP53 and CDKN2A. They also reactivate telomerase reverse transcriptase. Senescence-to-immortality transition is accompanied by major phenotypic and biochemical changes mediated by genome-wide transcriptional modifications. This appears to happen during hepatocellular carcinoma (HCC) development in patients with liver cirrhosis, however, the accompanying transcriptional changes are virtually unknown. We investigated genome-wide transcriptional changes related to the senescence-to-immortality switch during hepatocellular carcinogenesis. Initially, we performed transcriptome analysis of senescent and immortal clones of Huh7 HCC cell line, and identified genes with significant differential expression to establish a senescence-related gene list. Through the analysis of senescence-related gene expression in different liver tissues we showed that cirrhosis and HCC display expression patterns compatible with senescent and immortal phenotypes, respectively; dysplasia being a transitional state. Gene set enrichment analysis revealed that cirrhosis/senescence-associated genes were preferentially expressed in non-tumor tissues, less malignant tumors, and differentiated or senescent cells. In contrast, HCC/immortality genes were up-regulated in tumor tissues, or more malignant tumors and progenitor cells. In HCC tumors and immortal cells genes involved in DNA repair, cell cycle, telomere extension and branched chain amino acid metabolism were up-regulated, whereas genes involved in cell signaling, as well as in drug, lipid, retinoid and glycolytic metabolism were down-regulated. Based on these distinctive gene expression features we developed a 15-gene hepatocellular immortality signature test that discriminated HCC from cirrhosis with high accuracy. Our findings demonstrate that senescence bypass plays a central role in hepatocellular carcinogenesis engendering systematic changes in the transcription of genes regulating DNA repair, proliferation, differentiation and metabolism.


Subject(s)
Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Cellular Senescence/genetics , Genome, Human , Liver Neoplasms/pathology , Transcription, Genetic , Base Sequence , Carcinoma, Hepatocellular/genetics , DNA Primers , Gene Expression Profiling , Humans , Liver Neoplasms/genetics , Polymerase Chain Reaction
10.
Liver Int ; 31(4): 561-71, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21382167

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a leading cause of cancer deaths. Aflatoxins, which may play a causative role in 5-28% of HCCs worldwide, are activated in liver cells and induce principally G→T mutations, including the TP53 codon 249(G→T) hotspot mutation. The DNA damage checkpoint response acts as an antitumour mechanism against genotoxic agents, but its role in aflatoxin-induced DNA damage is unknown. AIM: We studied the DNA damage checkpoint response of human cells to aflatoxin B1 (AFB1). METHODS AND RESULTS: The treatment of HepG2 hepatoma cells with mutation-inducing doses (3-5 µmol/l) of AFB1 induced DNA adducts, 8-hydroxyguanine lesions and DNA strand breaks that lasted several days. Persistent phospho-H2AX and 53BP1 foci were also detected, but cell growth was not affected. AFB1-exposed HepG2 cells formed phospho-H2AX and 53BP1 foci, but failed to phosphorylate both Chk1 and Chk2. Huh7 hepatoma and HCT116 colorectal cancer cell lines also exhibited a similarly incomplete checkpoint response. p53 phosphorylation also failed, and AFB1-exposed cells did not show p53-dependent G1 arrest or a sustained G2/M arrest. These observations contrasted sharply with the fully functional DNA damage response of cells to Adriamycin. Cotreatment of cells with AFB1 did not inhibit p53 and p21(Cip1) accumulation induced by Adriamycin. Thus, the deficient checkpoint response to AFB1 was not due to an inhibitory effect, but could be explained by an inefficient activation. CONCLUSION: Genotoxic doses of AFB1 induce an incomplete and inefficient checkpoint response in human cells. This defective response may contribute to the mutagenic and carcinogenic potencies of aflatoxins.


Subject(s)
Aflatoxin B1/toxicity , Carcinoma, Hepatocellular/chemically induced , DNA Damage/drug effects , Liver Neoplasms/chemically induced , Mutation/drug effects , Tumor Suppressor Protein p53/genetics , Blotting, Western , Bromodeoxyuridine , Cell Line, Tumor , Comet Assay , Doxorubicin , Fluorescent Antibody Technique, Indirect , Humans , Immunoenzyme Techniques , Mutagenicity Tests , Phosphorylation , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase
11.
PLoS One ; 5(6): e11288, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20585577

ABSTRACT

BACKGROUND: Breast cancer is a remarkably heterogeneous disease. Luminal, basal-like, "normal-like", and ERBB2+ subgroups were identified and were shown to have different prognoses. The mechanisms underlying this heterogeneity are poorly understood. In our study, we explored the role of cellular differentiation and senescence as a potential cause of heterogeneity. METHODOLOGY/PRINCIPAL FINDINGS: A panel of breast cancer cell lines, isogenic clones, and breast tumors were used. Based on their ability to generate senescent progeny under low-density clonogenic conditions, we classified breast cancer cell lines as senescent cell progenitor (SCP) and immortal cell progenitor (ICP) subtypes. All SCP cell lines expressed estrogen receptor (ER). Loss of ER expression combined with the accumulation of p21(Cip1) correlated with senescence in these cell lines. p21(Cip1) knockdown, estrogen-mediated ER activation or ectopic ER overexpression protected cells against senescence. In contrast, tamoxifen triggered a robust senescence response. As ER expression has been linked to luminal differentiation, we compared the differentiation status of SCP and ICP cell lines using stem/progenitor, luminal, and myoepithelial markers. The SCP cells produced CD24+ or ER+ luminal-like and ASMA+ myoepithelial-like progeny, in addition to CD44+ stem/progenitor-like cells. In contrast, ICP cell lines acted as differentiation-defective stem/progenitor cells. Some ICP cell lines generated only CD44+/CD24-/ER-/ASMA- progenitor/stem-like cells, and others also produced CD24+/ER- luminal-like, but not ASMA+ myoepithelial-like cells. Furthermore, gene expression profiles clustered SCP cell lines with luminal A and "normal-like" tumors, and ICP cell lines with luminal B and basal-like tumors. The ICP cells displayed higher tumorigenicity in immunodeficient mice. CONCLUSIONS/SIGNIFICANCE: Luminal A and "normal-like" breast cancer cell lines were able to generate luminal-like and myoepithelial-like progeny undergoing senescence arrest. In contrast, luminal B/basal-like cell lines acted as stem/progenitor cells with defective differentiation capacities. Our findings suggest that the malignancy of breast tumors is directly correlated with stem/progenitor phenotypes and poor differentiation potential.


Subject(s)
Breast Neoplasms/pathology , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , Cluster Analysis , Female , Humans , Immunohistochemistry , Receptors, Estrogen/metabolism
12.
Mol Cancer ; 8: 90, 2009 Oct 22.
Article in English | MEDLINE | ID: mdl-19849855

ABSTRACT

BACKGROUND: beta-catenin mutations that constitutively activate the canonical Wnt signaling have been observed in a subset of hepatocellular carcinomas (HCCs). These mutations are associated with chromosomal stability, low histological grade, low tumor invasion and better patient survival. We hypothesized that canonical Wnt signaling is selectively activated in well-differentiated, but repressed in poorly differentiated HCCs. To this aim, we characterized differentiation status of HCC cell lines and compared their expression status of Wnt pathway genes, and explored their activity of canonical Wnt signaling. RESULTS: We classified human HCC cell lines into "well-differentiated" and "poorly differentiated" subtypes, based on the expression of hepatocyte lineage, epithelial and mesenchymal markers. Poorly differentiated cell lines lost epithelial and hepatocyte lineage markers, and overexpressed mesenchymal markers. Also, they were highly motile and invasive. We compared the expression of 45 Wnt pathway genes between two subtypes. TCF1 and TCF4 factors, and LRP5 and LRP6 co-receptors were ubiquitously expressed. Likewise, six Frizzled receptors, and canonical Wnt3 ligand were expressed in both subtypes. In contrast, canonical ligand Wnt8b and noncanonical ligands Wnt4, Wnt5a, Wnt5b and Wnt7b were expressed selectively in well- and poorly differentiated cell lines, respectively. Canonical Wnt signaling activity, as tested by a TCF reporter assay was detected in 80% of well-differentiated, contrary to 14% of poorly differentiated cell lines. TCF activity generated by ectopic mutant beta-catenin was weak in poorly differentiated SNU449 cell line, suggesting a repressive mechanism. We tested Wnt5a as a candidate antagonist. It strongly inhibited canonical Wnt signaling that is activated by mutant beta-catenin in HCC cell lines. CONCLUSION: Differential expression of Wnt ligands in HCC cells is associated with selective activation of canonical Wnt signaling in well-differentiated, and its repression in poorly differentiated cell lines. One potential mechanism of repression involved Wnt5a, acting as an antagonist of canonical Wnt signaling. Our observations support the hypothesis that Wnt pathway is selectively activated or repressed depending on differentiation status of HCC cells. We propose that canonical and noncanonical Wnt pathways have complementary roles in HCC, where the canonical signaling contributes to tumor initiation, and noncanonical signaling to tumor progression.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Wnt Proteins/metabolism , Autocrine Communication , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/classification , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Differentiation , Cell Line, Tumor , Cell Nucleus/metabolism , Frizzled Receptors/metabolism , Gene Expression Regulation, Neoplastic , Genes, Reporter , Humans , Immunohistochemistry , Ligands , Liver Neoplasms/classification , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mutation/genetics , Neoplasm Proteins/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , TCF Transcription Factors/genetics , TCF Transcription Factors/metabolism , Wnt-5a Protein
13.
Cancer Lett ; 286(1): 103-13, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19070423

ABSTRACT

Cellular senescence is a process leading to terminal growth arrest with characteristic morphological features. This process is mediated by telomere-dependent, oncogene-induced and ROS-induced pathways, but persistent DNA damage is the most common cause. Senescence arrest is mediated by p16(INK4a)- and p21(Cip1)-dependent pathways both leading to retinoblastoma protein (pRb) activation. p53 plays a relay role between DNA damage sensing and p21(Cip1) activation. pRb arrests the cell cycle by recruiting proliferation genes to facultative heterochromatin for permanent silencing. Replicative senescence that occurs in hepatocytes in culture and in liver cirrhosis is associated with lack of telomerase activity and results in telomere shortening. Hepatocellular carcinoma (HCC) cells display inactivating mutations of p53 and epigenetic silencing of p16(INK4a). Moreover, they re-express telomerase reverse transcriptase required for telomere maintenance. Thus, senescence bypass and cellular immortality is likely to contribute significantly to HCC development. Oncogene-induced senescence in premalignant lesions and reversible immortality of cancer cells including HCC offer new potentials for tumor prevention and treatment.


Subject(s)
Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/genetics , Cellular Senescence , Liver Neoplasms/pathology , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Activation , Hepatocytes/pathology , Humans , Protein Kinase Inhibitors , Telomerase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...