Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Hepatology ; 74(4): 1971-1993, 2021 10.
Article in English | MEDLINE | ID: mdl-33931882

ABSTRACT

BACKGROUND AND AIMS: Synthetic cyclin-dependent kinase (CDK) 4/6 inhibitors exert antitumor effects by forcing RB1 in unphosphorylated status, causing not only cell cycle arrest but also cellular senescence, apoptosis, and increased immunogenicity. These agents currently have an indication in advanced breast cancers and are in clinical trials for many other solid tumors. HCC is one of promising targets of CDK4/6 inhibitors. RB family dysfunction is often associated with the initiation of HCC; however, this is revivable, as RB family members are not frequently mutated or deleted in this malignancy. APPROACH AND RESULTS: Loss of all Rb family members in transformation related protein 53 (Trp53)-/- mouse liver resulted in liver tumor reminiscent of human HCC, and re-expression of RB1 sensitized these tumors to a CDK4/6 inhibitor, palbociclib. Introduction of an unphosphorylatable form of RB1 (RB7LP) into multiple liver tumor cell lines induced effects similar to palbociclib. By screening for compounds that enhance the efficacy of RB7LP, we identified an I kappa B kinase (IKK)ß inhibitor Bay 11-7082. Consistently, RB7LP expression and treatment with palbociclib enhanced IKKα/ß phosphorylation and NF-κB activation. Combination therapy using palbociclib with Bay 11-7082 was significantly more effective in hepatoblastoma and HCC treatment than single administration. Moreover, blockade of IKK-NF-κB or AKT pathway enhanced effects of palbociclib on RB1-intact KRAS Kirsten rat sarcoma viral oncogene homolog mutated lung and colon cancers. CONCLUSIONS: In conclusion, CDK4/6 inhibitors have a potential to treat a wide variety of RB1-intact cancers including HCC when combined with an appropriate kinase inhibitor.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Liver Neoplasms/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Aminopyridines/pharmacology , Aminopyridines/therapeutic use , Animals , Benzimidazoles/pharmacology , Benzimidazoles/therapeutic use , Carcinoma, Hepatocellular/genetics , Cell Proliferation/genetics , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Hep G2 Cells , Humans , In Vitro Techniques , Liver Neoplasms/genetics , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/genetics , Mice , Neoplasm Transplantation , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Purines/pharmacology , Purines/therapeutic use , Pyridines/therapeutic use , Retinoblastoma Protein , Tumor Suppressor Protein p53/genetics , Xenopus Proteins
2.
Oncogene ; 39(34): 5690-5707, 2020 08.
Article in English | MEDLINE | ID: mdl-32694611

ABSTRACT

RB1 gene is often homozygously deleted or mutated in prostate adenocarcinomas following acquirement of castration resistance and/or metastatic ability. We found that SUCLA2 gene is frequently involved in the deletion of the RB1 gene region in advanced prostate cancer. SUCLA2 constitutes the ß-subunit of succinate CoA ligase heterodimer that reversibly converts succinyl CoA into succinate. We sought the possibility that deletion of SUCLA2 gives rise to a metabolic vulnerability that could be targeted therapeutically. We found a significant metabolic shift in SUCLA2-deleted prostate cancer cells, including lower mitochondrial respiratory activity. By screening a number of libraries for compounds that induce cell death selectively in SUCLA2-deficient prostate cancer cells, we identified thymoquinone (2-isopropyl-5-methylbenzo-1,4-quinone) and PMA (phorbol-12-myristate-13-acetate) from a natural compound library. These findings indicate that the metabolic vulnerability in SUCLA2-deficient prostate cancer cells is pharmacologically targetable.


Subject(s)
Gene Deletion , Prostatic Neoplasms/genetics , Retinoblastoma Protein/genetics , Succinate-CoA Ligases/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Benzoquinones/pharmacology , Cell Line, Tumor , HEK293 Cells , Humans , Male , Mice, Knockout , Mice, Nude , Mice, SCID , PC-3 Cells , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Retinoblastoma Protein/deficiency , Succinate-CoA Ligases/deficiency , Tetradecanoylphorbol Acetate/analogs & derivatives , Tetradecanoylphorbol Acetate/pharmacology
3.
Cancer Res ; 79(15): 3903-3915, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31189648

ABSTRACT

Cancer cell-intrinsic properties caused by oncogenic mutations have been well characterized; however, how specific oncogenes and tumor suppressors impact the tumor microenvironment (TME) is not well understood. Here, we present a novel non-cell-autonomous function of the retinoblastoma (RB) tumor suppressor in controlling the TME. RB inactivation stimulated tumor growth and neoangiogenesis in a syngeneic and orthotropic murine soft-tissue sarcoma model, which was associated with recruitment of tumor-associated macrophages (TAM) and immunosuppressive cells such as Gr1+CD11b+ myeloid-derived suppressor cells (MDSC) or Foxp3+ regulatory T cells (Treg). Gene expression profiling and analysis of genetically engineered mouse models revealed that RB inactivation increased secretion of the chemoattractant CCL2. Furthermore, activation of the CCL2-CCR2 axis in the TME promoted tumor angiogenesis and recruitment of TAMs and MDSCs into the TME in several tumor types including sarcoma and breast cancer. Loss of RB increased fatty acid oxidation (FAO) by activating AMP-activated protein kinase that led to inactivation of acetyl-CoA carboxylase, which suppresses FAO. This promoted mitochondrial superoxide production and JNK activation, which enhanced CCL2 expression. These findings indicate that the CCL2-CCR2 axis could be an effective therapeutic target in RB-deficient tumors. SIGNIFICANCE: These findings demonstrate the cell-nonautonomous role of the tumor suppressor retinoblastoma in the tumor microenvironment, linking retinoblastoma loss to immunosuppression.


Subject(s)
Chemokine CCL2/metabolism , Retinoblastoma Protein/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Culture Techniques , Cell Line, Tumor , Chemokine CCL2/biosynthesis , Female , Gene Expression Profiling/methods , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/metabolism , Retinoblastoma Protein/deficiency , Soft Tissue Neoplasms/metabolism , Soft Tissue Neoplasms/pathology , Tumor Microenvironment , Up-Regulation
4.
Oncotarget ; 8(8): 13872-13885, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28099924

ABSTRACT

We established an in vitro cell culture system to determine novel activities of the retinoblastoma (Rb) protein during tumor progression. Rb depletion in p53-null mouse-derived soft tissue sarcoma cells induced a spherogenic phenotype. Cells retrieved from Rb-depleted spheres exhibited slower proliferation and less efficient BrdU incorporation, however, much higher spherogenic activity and aggressive behavior. We discovered six miRNAs, including mmu-miR-18a, -25, -29b, -140, -337, and -1839, whose expression levels correlated tightly with the Rb status and spherogenic activity. Among these, mmu-miR-140 appeared to be positively controlled by Rb and to antagonize the effect of Rb depletion on spherogenesis and tumorigenesis. Furthermore, among genes potentially targeted by mmu-miR-140, Il-6 was upregulated by Rb depletion and downregulated by mmu-mir-140 overexpression. Altogether, we demonstrate the possibility that mmu-mir-140 mediates the Rb function to downregulate Il-6 by targeting its 3'-untranslated region. Finally, we detected the same relationship among RB, hsa-miR-140 and IL-6 in a human breast cancer cell line MCF-7. Because IL-6 is a critical modulator of malignant features of cancer cells and the RB pathway is impaired in the majority of cancers, hsa-miR-140 might be a promising therapeutic tool that disrupts linkage between tumor suppressor inactivation and pro-inflammatory cytokine response.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Interleukin-6/metabolism , MicroRNAs/metabolism , Neoplasms/pathology , Neoplastic Stem Cells/pathology , Retinoblastoma Protein/metabolism , Animals , Cell Line, Tumor , Humans , Immunoblotting , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplasms/genetics , Oligonucleotide Array Sequence Analysis , Polymerase Chain Reaction , Retinoblastoma Protein/genetics , Transcriptome
5.
Stem Cells ; 33(5): 1657-69, 2015 May.
Article in English | MEDLINE | ID: mdl-25694388

ABSTRACT

Retinoblastoma tumor suppressor protein (RB) is inactivated more frequently during tumor progression than during tumor initiation. However, its exact role in controlling the malignant features associated with tumor progression is poorly understood. We established in vivo and in vitro models to investigate the undifferentiated state induced by Rb inactivation. Rb heterozygous mice develop well-differentiated thyroid medullary carcinoma. We found that additional deletion of Trp53, without change in lineage, converted these Rb-deficient tumors to a poorly differentiated type associated with higher self-renewal activity. Freshly prepared mouse embryonic fibroblasts (MEFs) of Rb(-/-) ; Trp53(-/-) background formed stem cell-like spheres that expressed significant levels of embryonic genes despite of lacking the ability to form colonies on soft agar or tumors in immune-deficient mice. This suggested that Rb-p53 double inactivation resulted in an undifferentiated status but without carcinogenic conversion. We next established Rb(-/-) ; N-ras(-/-) MEFs that harbored a spontaneous carcinogenic mutation in Trp53. These cells (RN6), in an Rb-dependent manner, efficiently generated spheres that expressed very high levels of embryonic genes, and appeared to be carcinogenic. We then screened an FDA-approved drug library to search for agents that suppressed the spherogenic activity of RN6 cells. Data revealed that RN6 cells were sensitive to specific agents including ones those are effective against cancer stem cells. Taken together, all these findings suggest that the genetic interaction between Rb and p53 is a critical determinant of the undifferentiated state in normal and tumor cells.


Subject(s)
Cell Differentiation , Embryo, Mammalian/cytology , Fibroblasts/cytology , Neuroendocrine Cells/cytology , Retinoblastoma Protein/metabolism , Thyroid Gland/cytology , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Animals , Behavior, Animal , Cell Line , Drug Evaluation, Preclinical , Fibroblasts/metabolism , Heterozygote , Mice, Knockout , Molecular Sequence Data , Mutation/genetics , Phenotype , Retinoblastoma Protein/deficiency , Spheroids, Cellular/metabolism , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , ras Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...