Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 154
Filter
1.
J Am Heart Assoc ; 11(19): e024764, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36129061

ABSTRACT

Background The postmitotic state of adult cardiomyocytes, maintained by the cell cycle repressor Rbl2 (retinoblastoma-like 2), is associated with considerable resistance to apoptosis. However, whether Rbl2 regulates cardiomyocyte apoptosis remains unknown. Methods and Results Here, we show that ablation of Rbl2 increased cardiomyocyte apoptosis following acute myocardial ischemia/reperfusion injury, leading to diminished cardiac function and exaggerated ventricular remodeling in the long term. Mechanistically, ischemia/reperfusion induced expression of the proapoptotic protein BCL2 interacting protein 3 (Bnip3), which was augmented by deletion of Rbl2. Because the Bnip3 promoter contains an adenoviral early region 2 binding factor (E2F)-binding site, we further showed that loss of Rbl2 upregulated the transcriptional activator E2F1 but downregulated the transcriptional repressor E2F4. In cultured cardiomyocytes, treatment with H2O2 markedly increased the levels of E2F1 and Bnip3, resulting in mitochondrial depolarization and apoptosis. Depletion of Rbl2 significantly augmented H2O2-induced mitochondrial damage and apoptosis in vitro. Conclusions Rbl2 deficiency enhanced E2F1-mediated Bnip3 expression, resulting in aggravated cardiomyocyte apoptosis and ischemia/reperfusion injury. Our results uncover a novel antiapoptotic role for Rbl2 in cardiomyocytes, suggesting that the cell cycle machinery may directly regulate apoptosis in postmitotic cardiomyocytes. These findings may be exploited to develop new strategies to limit ischemia/reperfusion injury in the treatment of acute myocardial infarction.


Subject(s)
Myocardial Reperfusion Injury , Retinal Neoplasms , Retinoblastoma , Apoptosis/physiology , Humans , Hydrogen Peroxide/metabolism , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Retinal Neoplasms/metabolism , Retinoblastoma/metabolism , Retinoblastoma-Like Protein p130/metabolism
2.
Chem Biol Interact ; 367: 110133, 2022 Nov 01.
Article in English | MEDLINE | ID: mdl-36030841

ABSTRACT

Nuclear receptor pregnane X receptor (PXR) can induce significant liver enlargement through hepatocyte hypertrophy and proliferation. A previous report showed that during the process of PXR-induced liver enlargement, hepatocyte hypertrophy occurs around the central vein (CV) area while hepatocyte proliferation occurs around the portal vein (PV) area. However, the features of this spatial change remain unclear. Therefore, this study aims to explore the features of the spatial changes in hepatocytes in PXR-induced liver enlargement. PXR-induced spatial changes in hepatocyte hypertrophy and proliferation were confirmed in C57BL/6 mice. The liver was perfused with digitonin to destroy the hepatocytes around the CV or PV areas, and then the regional expression of proteins related to hepatocyte hypertrophy and proliferation was further measured. The results showed that the expression of PXR downstream proteins, such as cytochrome P450 (CYP) 3A11, CYP2B10, P-glycoprotein (P-gp) and organ anion transporting polypeptide 4 (OATP4) was upregulated around the CV area, while the expression of proliferation-related proteins such as cyclin B1 (CCNB1), cyclin D1 (CCND1) and serine/threonine NIMA-related kinase 2 (NEK2) was upregulated around the PV area. At the same time, the expression of cyclin-dependent kinase inhibitors such as retinoblastoma-like protein 2 (RBL2), cyclin-dependent kinase inhibitor 1B (CDKN1B) and CDKN1A was downregulated around the PV area. This study demonstrated that the spatial change in PXR-induced hepatocyte hypertrophy and proliferation is associated with the regional expression of PXR downstream targets and proliferation-related proteins and the regional distribution of triglycerides (TGs). These findings provide new insight into the understanding of PXR-induced hepatomegaly.


Subject(s)
Cyclin D1 , Receptors, Steroid , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Anions/metabolism , Cell Proliferation , Cyclin B1/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cyclin-Dependent Kinases/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme System/metabolism , Digitonin/metabolism , Hepatocytes/metabolism , Hepatomegaly/chemically induced , Hepatomegaly/metabolism , Hypertrophy/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , NIMA-Related Kinases/metabolism , Pregnane X Receptor/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Retinoblastoma-Like Protein p130/metabolism , Serine/metabolism , Threonine/metabolism , Triglycerides/metabolism
3.
Structure ; 30(9): 1340-1353.e3, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35716663

ABSTRACT

The retinoblastoma protein (Rb) and its homologs p107 and p130 are critical regulators of gene expression during the cell cycle and are commonly inactivated in cancer. Rb proteins use their "pocket domain" to bind an LxCxE sequence motif in other proteins, many of which function with Rb proteins to co-regulate transcription. Here, we present binding data and crystal structures of the p107 pocket domain in complex with LxCxE peptides from the transcriptional co-repressor proteins HDAC1, ARID4A, and EID1. Our results explain why Rb and p107 have weaker affinity for cellular LxCxE proteins compared with the E7 protein from human papillomavirus, which has been used as the primary model for understanding LxCxE motif interactions. Our structural and mutagenesis data also identify and explain differences in Rb and p107 affinities for some LxCxE-containing sequences. Our study provides new insights into how Rb proteins bind their cell partners with varying affinity and specificity.


Subject(s)
Repressor Proteins , Retinoblastoma Protein , Cell Cycle , Humans , Repressor Proteins/genetics , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p130/metabolism
4.
Eur J Med Genet ; 65(6): 104513, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35487417

ABSTRACT

RBL2/p130 is one of three highly conserved members of the retinoblastoma (RB) protein family. It is strongly upregulated during neuronal differentiation and brain development, and is critical for survival of post-mitotic neurons. Similar to RB1, it has been implicated as a tumor suppressor gene and has been shown to be dysregulated in various types of cancer. Recent publications describe biallelic, germline loss of function variants in RBL2 in individuals with profound developmental delay. We report a child with profound developmental delay, microcephaly, and hypotonia, who developed fulminant exophthalmos at age 6 years. Brain MRI followed by a biopsy of an intra-orbital mass revealed a mesenchymal tumor. Post-surgical histopathologic examination of the resected tumor was compatible with diagnosis of nodular fasciitis. Exome sequencing from peripheral blood identified a biallelic frameshift variant (c.901dupT) in RBL2. Notably, no malignancies were reported in previous cases with RBL2 variants. This case provides a possible association between RBL2 and orbital tumors.


Subject(s)
Fasciitis , Retinal Neoplasms , Retinoblastoma , Child , Humans , Retinoblastoma/genetics , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism
5.
Sci Rep ; 12(1): 1049, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058503

ABSTRACT

Wild-type p53 is a stress-responsive transcription factor and potent tumor suppressor. P53 activates or represses genes involved in cell cycle progression or apoptosis in order to arrest the cell cycle or induce cell death. Transcription repression by p53 is indirect and requires repressive members of the RB-family (RB1, RBL1, RBL2) and formation of repressor complexes of RB1-E2F and RBL1/RBL2-DREAM. Many aurora kinase A/B (AURKA/B) pathway genes are repressed in a p53-DREAM-dependent manner. We found heightened expression of RBL2 and reduced expression of AURKA/B pathway genes is associated with improved outcomes in p53 wild-type but not p53 mutant non-small cell lung cancer (NSCLC) patients. Knockdown of p53, RBL2, or the DREAM component LIN37 increased AURKA/B pathway gene expression and reduced paclitaxel and radiation toxicity in NSCLC cells. In contrast, pharmacologic inhibition of AURKA/B or knockdown of AURKA/B pathway components increased paclitaxel and IR sensitivity. The results support a model in which p53-RBL2-DREAM-mediated repression of the AURKA/B pathway contributes to tumor suppression, improved tumor therapy responses, and better outcomes in p53 wild-type NSCLCs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Gene Expression Regulation, Neoplastic , Retinoblastoma-Like Protein p130/metabolism , Tumor Suppressor Protein p53/metabolism , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Aurora Kinase B/genetics , Aurora Kinase B/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Humans , Paclitaxel/therapeutic use , Retinoblastoma-Like Protein p130/genetics , Tumor Suppressor Protein p53/genetics
6.
Elife ; 102021 12 01.
Article in English | MEDLINE | ID: mdl-34851822

ABSTRACT

Cell cycle gene expression programs fuel proliferation and are universally dysregulated in cancer. The retinoblastoma (RB)-family of proteins, RB1, RBL1/p107, and RBL2/p130, coordinately represses cell cycle gene expression, inhibiting proliferation, and suppressing tumorigenesis. Phosphorylation of RB-family proteins by cyclin-dependent kinases is firmly established. Like phosphorylation, ubiquitination is essential to cell cycle control, and numerous proliferative regulators, tumor suppressors, and oncoproteins are ubiquitinated. However, little is known about the role of ubiquitin signaling in controlling RB-family proteins. A systems genetics analysis of CRISPR/Cas9 screens suggested the potential regulation of the RB-network by cyclin F, a substrate recognition receptor for the SCF family of E3 ligases. We demonstrate that RBL2/p130 is a direct substrate of SCFcyclin F. We map a cyclin F regulatory site to a flexible linker in the p130 pocket domain, and show that this site mediates binding, stability, and ubiquitination. Expression of a mutant version of p130, which cannot be ubiquitinated, severely impaired proliferative capacity and cell cycle progression. Consistently, we observed reduced expression of cell cycle gene transcripts, as well a reduced abundance of cell cycle proteins, analyzed by quantitative, iterative immunofluorescent imaging. These data suggest a key role for SCFcyclin F in the CDK-RB network and raise the possibility that aberrant p130 degradation could dysregulate the cell cycle in human cancers.


Subject(s)
Cyclins/genetics , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma/genetics , Stem Cell Factor/genetics , Cyclins/metabolism , HEK293 Cells , HeLa Cells , Humans , MCF-7 Cells , Retinoblastoma-Like Protein p130/metabolism , Stem Cell Factor/metabolism
7.
Cancer Res ; 80(22): 5051-5062, 2020 11 15.
Article in English | MEDLINE | ID: mdl-32973083

ABSTRACT

Small cell lung cancer (SCLC) remains a recalcitrant disease where limited therapeutic options have not improved overall survival, and approved targeted therapies are lacking. Amplification of the tyrosine kinase receptor FGFR1 (fibroblast growth factor receptor 1) is one of the few actionable alterations found in the SCLC genome. However, efforts to develop targeted therapies for FGFR1-amplified SCLC are hindered by critical gaps in knowledge around the molecular origins and mediators of FGFR1-driven signaling as well as the physiologic impact of targeting FGFR1. Here we show that increased FGFR1 promotes tumorigenic progression in precancerous neuroendocrine cells and is required for SCLC development in vivo. Notably, Fgfr1 knockout suppressed tumor development in a mouse model lacking the retinoblastoma-like protein 2 (Rbl2) tumor suppressor gene but did not affect a model with wild-type Rbl2. In support of a functional interaction between these two genes, loss of RBL2 induced FGFR1 expression and restoration of RBL2 repressed it, suggesting a novel role for RBL2 as a regulator of FGFR1 in SCLC. Additionally, FGFR1 activated phospholipase C gamma 1 (PLCG1), whereas chemical inhibition of PLCG1 suppressed SCLC growth, implicating PLCG1 as an effector of FGFR1 signaling in SCLC. Collectively, this study uncovers mechanisms underlying FGFR1-driven SCLC that involve RBL2 upstream and PLCG1 downstream, thus providing potential biomarkers for anti-FGFR1 therapy. SIGNIFICANCE: This study identifies RBL2 and PLCG1 as critical components of amplified FGFR1 signaling in SCLC, thus representing potential targets for biomarker analysis and therapeutic development in this disease.


Subject(s)
Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Phospholipase C gamma/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Retinoblastoma-Like Protein p130/metabolism , Small Cell Lung Carcinoma/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Disease Progression , Female , Gene Deletion , Genes, Regulator , Genes, Retinoblastoma , Humans , Lung Neoplasms/etiology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/pathology , Phospholipase C gamma/antagonists & inhibitors , Precancerous Conditions/metabolism , Precancerous Conditions/pathology , Receptor, Fibroblast Growth Factor, Type 1/genetics , Retinoblastoma-Like Protein p130/genetics , Small Cell Lung Carcinoma/etiology , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism
8.
Nucleic Acids Res ; 47(21): 11197-11208, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31667499

ABSTRACT

Cell cycle gene expression occurs in two waves. The G1/S genes encode factors required for DNA synthesis and the G2/M genes contribute to mitosis. The Retinoblastoma protein (RB) and DREAM complex (DP, RB-like, E2F4 and MuvB) cooperate to repress all cell cycle genes during G1 and inhibit entry into the cell cycle. DNA damage activates p53 leading to increased levels of p21 and inhibition of cell cycle progression. Whether the G1/S and G2/M genes are differentially repressed by RB and the RB-like proteins p130 and p107 in response to DNA damage is not known. We performed gene expression profiling of primary human fibroblasts upon DNA damage and assessed the effects on G1/S and G2/M genes. Upon p53 activation, p130 and RB cooperated to repress the G1/S genes. In addition, in the absence of RB and p130, p107 contributed to repression of G1/S genes. In contrast, G2/M genes were repressed by p130 and p107 after p53 activation. Furthermore, repression of G2/M genes by p107 and p130 led to reduced entry into mitosis. Our data demonstrates specific roles for RB, p130-DREAM, and p107-DREAM in p53 and p21 mediated repression of cell cycle genes.


Subject(s)
G1 Phase Cell Cycle Checkpoints/genetics , G2 Phase Cell Cycle Checkpoints/genetics , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/genetics , Tumor Suppressor Protein p53/metabolism , Cell Cycle/genetics , Cells, Cultured , Down-Regulation/genetics , Genes, cdc/genetics , Humans , Retinoblastoma Protein/metabolism , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/metabolism , Tumor Suppressor Protein p53/genetics
9.
Cell Death Dis ; 10(10): 721, 2019 09 26.
Article in English | MEDLINE | ID: mdl-31558708

ABSTRACT

Exencephaly/anencephaly is one of the leading causes of neonatal mortality and the most extreme open neural tube defect with no current treatments and limited mechanistic understanding. We hypothesized that exencephaly leads to a local neurodegenerative process in the brain exposed to the amniotic fluid as well as diffuse degeneration in other encephalic areas and the spinal cord. To evaluate the consequences of in utero neural tissue exposure, brain and spinal cord samples from E17 exencephalic murine fetuses (maternal intraperitoneal administration of valproic acid at E8) were analyzed and compared to controls and saline-injected shams (n = 11/group). Expression of apoptosis and senescence genes (p53, p21, p16, Rbl2, Casp3, Casp9) was determined by qRT-PCR and protein expression analyzed by western blot. Apoptosis was measured by TUNEL assay and PI/AV flow cytometry. Valproic acid at E8 induced exencephaly in 22% of fetuses. At E17 the fetuses exhibited the characteristic absence of cranial bones. The brain structures from exencephalic fetuses demonstrated a loss of layers in cortical regions and a complete loss of structural organization in the olfactory bulb, hippocampus, dental gyrus and septal cortex. E17 fetuses had reduced expression of NeuN, GFAP and Oligodendrocytes in the brain with primed microglia. Intrinsic apoptotic activation (p53, Caspase9 and 3) was upregulated and active Caspase3 localized to the layer of brain exposed to the amniotic fluid. Senescence via p21-Rbl2 was increased in the brain and in the spinal cord at the lamina I-II of the somatosensory dorsal horn. The current study characterizes CNS alterations in murine exencephaly and demonstrates that degeneration due to intrinsic apoptosis and senescence occurs in the directly exposed brain but also remotely in the spinal cord.


Subject(s)
Anencephaly/pathology , Apoptosis , Brain/pathology , Necrosis/pathology , Neural Tube Defects/pathology , Spinal Cord/pathology , Amniotic Fluid/metabolism , Anencephaly/chemically induced , Anencephaly/embryology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Brain/cytology , Brain/embryology , Caspase 3/metabolism , Caspase 9/metabolism , Cellular Senescence/drug effects , Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Disease Progression , Female , Mice , Microglia/cytology , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , Necrosis/embryology , Necrosis/metabolism , Neurons/cytology , Neurons/pathology , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Spinal Cord/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation , Valproic Acid
10.
Cell Cycle ; 18(19): 2566-2579, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31432742

ABSTRACT

This study is aimed to clarify the potential role of lncRNA LINC00899 in invasion and migration of spinal ependymoma cells through the FoxO pathway via RBL2. Spinal ependymoma related chip data (GSE50161 and GSE66354) was initially downloaded and differentially expressed lncRNAs were screened out. Fifty-eight cases of spinal ependymoma and normal ependymal tissues were collected. The effects of LINC00899 and RBL2 on the spinal ependymoma cell migration and invasion were determined using the third generation spinal ependymoma cells and transfection with LINC00899 vector, siRNA-LINC00899 and siRNA-RBL2. The expression of LINC00899, pathway and cell proliferation- and apoptosis-related factors was determined. Finally, we also detected cell proliferation, migration, invasion, cycle and apoptosis after transfection. Our results showed that LINC00899 was up-regulated in spinal ependymoma and RBL2 was confirmed as a target gene of LINC00899 and found to be involved in regulation of FoxO pathway. LINC00899 expression increased in spinal ependymoma tissues whereas RBL2 expression decreased. Moreover, we found that siRNA-LINC00899 could elevate RBL2, p21, p27 and Bax levels, decrease FoxO, Bcl-2, Vimentin, Annexin levels, reduced cell proliferation, migration and invasion and enhanced apoptosis. Taken together, our study suggests that down-regulated LINC00899 exerts anti-oncogenic effects on spinal ependymoma via RBL2-dependent FoxO, which provides a novel therapeutic target for the treatment of spinal ependymomas.


Subject(s)
Ependymoma/metabolism , Forkhead Box Protein O1/metabolism , RNA, Long Noncoding/metabolism , Retinoblastoma-Like Protein p130/metabolism , Spinal Cord Neoplasms/metabolism , Adolescent , Adult , Annexins/genetics , Annexins/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Child , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Databases, Genetic , Ependymoma/genetics , Ependymoma/pathology , Female , Forkhead Box Protein O1/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Neoplasm Invasiveness/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Long Noncoding/genetics , Retinoblastoma-Like Protein p130/genetics , Spinal Cord Neoplasms/genetics , Spinal Cord Neoplasms/pathology , Vimentin/genetics , Vimentin/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
11.
J Physiol Biochem ; 75(2): 209-215, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31020623

ABSTRACT

The mammary gland increases energy requirements during pregnancy and lactation to support epithelial proliferation and milk nutrients synthesis. Lactose, the principal carbohydrate of the milk, is synthetized in the Golgi of mammary epithelial cells by lactose synthase from glucose and UPD galactose. We studied the temporal changes in the expression of GLUT1 and GLUT8 in mammary gland and their association with lactose synthesis and proliferation in BALB/c mice. Six groups were used: virgin, pregnant at 2 and 17 days, lactating at 2 and 10 days, and weaning at 2 days. Temporal expression of GLUT1 and GLUT8 transporters by qPCR, western blot and immunohistochemistry, and its association with lactalbumin, Ki67, and cytokeratin 18 within mammary tissue was studied, along with subcellular localization. GLUT1 and GLUT8 transporters increased their expression during mammary gland progression, reaching 20-fold increasing in GLUT1 mRNA at lactation (p < 0.05) and 2-fold at protein level for GLUT1 and GLUT8 (p < 0.05 and 0.01, respectively). The temporal expression pattern was shared with cytokeratin 18 and Ki67 (p < 0.01). Endogenous GLUT8 partially co-localized with 58 K protein and α-lactalbumin in mammary tissue and with Golgi membrane-associated protein 130 in isolated epithelial cells. The spatial-temporal synchrony between expression of GLUT8/GLUT1 and alveolar cell proliferation, and its localization in cis-Golgi associated to lactose synthase complex, suggest that both transporters are involved in glucose uptake into this organelle, supporting lactose synthesis.


Subject(s)
Epithelial Cells/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 1/metabolism , Golgi Apparatus/metabolism , Mammary Glands, Animal/metabolism , Animals , Epithelial Cells/immunology , Female , Glucose/metabolism , Glucose Transport Proteins, Facilitative/genetics , Glucose Transporter Type 1/genetics , Keratin-18/metabolism , Lactalbumin/metabolism , Lactation , Lactose/biosynthesis , Lactose Synthase/metabolism , Mice , Mice, Inbred BALB C , Peptides/metabolism , Pregnancy , RNA, Messenger/metabolism , Retinoblastoma-Like Protein p130/metabolism , Time Factors , Weaning
12.
Redox Biol ; 24: 101169, 2019 06.
Article in English | MEDLINE | ID: mdl-30889466

ABSTRACT

Recently, clinical development of PARP inhibitors (PARPi) expanded from using them as a single agent to combining them with DNA-damaging therapy to derive additional therapeutic benefit from stimulated DNA damage. Furthermore, inhibiting PARP in cancers with BRCA1/2 mutations has been shown to be an effective synthetic lethality approach either as a single agent or in combination with the different DNA damaging agents: chemotherapy or ionizing radiation (IR). However, inherited BRCA1/2 mutations account only for 5-10% of breast cancers, 10-15% of ovarian cancers, and lesser for the other cancers. Hence, for most of the cancer patients with BRCA1/2-proficient tumors, sensitization to DNA-damaging agents with PARPi is significantly less effective. We recently demonstrated that moderate, non-toxic concentrations of NO-donors inhibited BRCA1 expression, with subsequent inhibition of error-free HRR and increase of error-prone non-homologous end joining (NHEJ). We also demonstrated that the effect of NO-dependent block of BRCA1 expression can only be achieved in the presence of oxidative stress, a condition that characterizes the tumor microenvironment and is also a potential effect of IR. Hence, NO-donors in combination with PARPi, with effects limited by tumor microenvironment and irradiated area, suggest a precise tumor-targeted approach for radio-sensitization of BRCA1/2-proficient tumors. The combination with NO-donors allows PARPi to be successfully applied to a wider variety of tumors. The present work demonstrates a new drug combination (NO-donors and PARP-inhibitors) which demonstrated a high potency in sensitization of wide variety of tumors to ionizing radiation treatment.


Subject(s)
Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/chemistry , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Cell Line, Tumor , DNA Damage , DNA Repair , Edetic Acid/chemistry , Humans , Radiation Tolerance/drug effects , Radiation Tolerance/radiation effects , Radiation, Ionizing , Retinoblastoma-Like Protein p130/metabolism , Signal Transduction , Synthetic Lethal Mutations/drug effects , Synthetic Lethal Mutations/genetics
13.
PLoS One ; 13(12): e0208110, 2018.
Article in English | MEDLINE | ID: mdl-30540809

ABSTRACT

The propensity for differentiation varies substantially across human pluripotent stem cell (hPSC) lines, greatly restricting the use of hPSCs for cell replacement therapy or disease modeling. Here, we investigate the underlying mechanisms and demonstrate that activation of the retinoblastoma (Rb) pathway in a transient manner is important for differentiation. In prior work, we demonstrated that pre-treating hPSCs with dimethylsulfoxide (DMSO) before directed differentiation enhanced differentiation potential across all three germ layers. Here, we show that exposure to DMSO improves the efficiency of hPSC differentiation through Rb and by repressing downstream E2F-target genes. While transient inactivation of the Rb family members (including Rb, p107, and p130) suppresses DMSO's capacity to enhance differentiation across all germ layers, transient expression of a constitutively active (non-phosphorylatable) form of Rb increases the differentiation efficiency similar to DMSO. Inhibition of downstream targets of Rb, such as E2F signaling, also promotes differentiation of hPSCs. More generally, we demonstrate that the duration of Rb activation plays an important role in regulating differentiation capacity.


Subject(s)
Cell Differentiation/drug effects , Dimethyl Sulfoxide/pharmacology , Pluripotent Stem Cells/drug effects , Retinoblastoma Protein/metabolism , Signal Transduction/drug effects , Aminopyridines/pharmacology , Cell Culture Techniques , Cell Differentiation/genetics , Cell Line , E2F Transcription Factors/antagonists & inhibitors , E2F Transcription Factors/metabolism , Gene Knockdown Techniques , Germ Layers/cytology , Germ Layers/drug effects , Germ Layers/physiology , Humans , Hydroxyquinolines/pharmacology , Pluripotent Stem Cells/physiology , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism , Signal Transduction/genetics , Time Factors
14.
J Biol Chem ; 293(41): 15815-15826, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30166342

ABSTRACT

Heat shock protein 27 (HSP27) protects cells under stress. Here, we demonstrate that HSP27 also promotes cell cycle progression of MRC-5 human lung fibroblast cells. Serum starvation for 24 h induced G1 arrest in these cells, and upon serum refeeding, the cells initiated cell cycle progression accompanied by an increase in HSP27 protein levels. HSP27 levels peaked at 12 h, and transcriptional up-regulation of six G2/M-related genes (CCNA2, CCNB1, CCNB2, CDC25C, CDCA3, and CDK1) peaked at 24-48 h. siRNA-mediated HSP27 silencing in proliferating MRC-5 cells induced G2 arrest coinciding with down-regulation of these six genes. Of note, the promoters of all of these genes have the cell cycle-dependent element and/or the cell cycle gene-homology region. These promoter regions are known to be bound by the E2F family proteins (E2F-1 to E2F-8) and retinoblastoma (RB) family proteins (RB1, p107, and p130), among which E2F-4 and p130 were strongly up-regulated in HSP27-knockdown cells. E2F-4 or p130 knockdown concomitant with the HSP27 knockdown rescued MRC-5 cells from G2 arrest and up-regulated the six cell cycle genes. Moreover, we observed cellular senescence in MRC-5 cells on day 3 after the HSP27 knockdown, as evidenced by increased senescence-associated ß-gal activity and up-regulated inflammatory cytokines. The cellular senescence was also suppressed by the concomitant knockdown of E2F-4/HSP27 or p130/HSP27. Our findings indicate that HSP27 promotes cell cycle progression of MRC-5 cells by suppressing expression of the transcriptional repressors E2F-4 and p130.


Subject(s)
Cell Cycle , E2F4 Transcription Factor/metabolism , HSP27 Heat-Shock Proteins/metabolism , Retinoblastoma-Like Protein p130/metabolism , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Down-Regulation , Fibroblasts/metabolism , G2 Phase , Gene Silencing , Heat-Shock Proteins , Humans , Lung/metabolism , Molecular Chaperones , Oxygen/chemistry , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism , Up-Regulation
15.
Eur Rev Med Pharmacol Sci ; 22(8): 2343-2350, 2018 04.
Article in English | MEDLINE | ID: mdl-29762838

ABSTRACT

OBJECTIVE: Glioma is a tumor of the brain. Although the clinical regimens and surgical techniques for glioma have improved, therapies of advanced glioma remain challenging, carrying dismal overall survival and therapeutic success rates. Evidence has shown that miRNAs played important roles in glioma development. The current study aimed at investigating the function of a novel cancerogenic miRNA, miR-93, in glioma progression by investigating the expression and mechanism of it. PATIENTS AND METHODS: qRT-PCR was conducted to assess the miR-93 expression and the mRNA expression of target gene in glioma tissues and cells. The invasion and migration abilities of the glioma cells were determined by transwell assays. Luciferase reporter assay was performed to confirm the target of miR-93. RESULTS: The results indicated that miR-93 expression in glioma tissues and cells was increased significantly than that in normal brain tissues and cells. Furthermore, miR-93 promoted glioma cell migration and invasion. RBL2 was recognized as a direct target of miR-93 in glioma cells, and overexpression of RBL2 could reverse the stimulative effect of miR-93 in glioma cell. CONCLUSIONS: The above findings suggested that miR-93 together with RBL2 could be diagnostic targets and novel prognostic markers for glioma.


Subject(s)
MicroRNAs/metabolism , Retinoblastoma-Like Protein p130/metabolism , 3' Untranslated Regions , Antagomirs/metabolism , Base Sequence , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mutagenesis , Retinoblastoma-Like Protein p130/chemistry , Retinoblastoma-Like Protein p130/genetics , Sequence Alignment
16.
Oncogene ; 37(27): 3657-3671, 2018 07.
Article in English | MEDLINE | ID: mdl-29606701

ABSTRACT

The retinoblastoma (RB) protein family includes RB1/p105, RBL1/p107, and RBL2/p130, which are key factors in cell-cycle regulation and stand at the crossroads of multiple pathways dictating cell fate decisions. The role of RB proteins in apoptosis is controversial because they can inhibit or promote apoptosis depending on the context, on the apoptotic stimuli and on their intrinsic status, impacting on the response to antitumoral treatments. Here we identified RBL2/p130 as a direct substrate of the AKT kinase, a key antiapoptotic factor hyperactive in multiple cancer types. We showed that RBL2/p130 and AKT1 physically interact and AKT phosphorylates RBL2/p130 Ser941, located in the pocket domain, but not when this residue is mutated into Ala. We found that pharmacological inhibition of AKT, through the highly selective AKT inhibitor VIII (AKTiVIII), impairs RBL2/p130 Ser941 phosphorylation and increases RBL2/p130 stability, mRNA expression and nuclear levels in both lung cancer and mesothelioma cell lines, mirroring the more extensively studied effects on the p27 cell-cycle inhibitor. Consistently, AKT inhibition reduced cell viability, induced cell accumulation in G0/G1, and triggered apoptosis, which proved to be largely dependent on RBL2/p130 itself, as shown upon RBL2/p130 silencing. AKT inhibition induced RBL2/p130-dependent apoptosis also in HEK-293 cells, in which re-expression of a short hairpin-resistant RBL2/p130 was able to rescue AKTiVIII-induced apoptosis upon RBL2/p130 silencing. Our data also showed that the combination of AKT and cyclin-dependent kinases (CDK) inhibitors, which converge on the re-activation of RBL2/p130 antitumoral potential, could be a promising anticancer strategy.


Subject(s)
Apoptosis/physiology , Lung Neoplasms/pathology , Mesothelioma/pathology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Retinoblastoma-Like Protein p130/metabolism , A549 Cells , Benzimidazoles/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Survival , HEK293 Cells , Humans , Lung Neoplasms/genetics , Mesothelioma/genetics , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/genetics , Quinoxalines/pharmacology , RNA, Messenger/biosynthesis , Retinoblastoma-Like Protein p130/genetics
17.
Mol Biol Rep ; 45(2): 185-194, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29417345

ABSTRACT

Dynamic positioning of nucleosomes is pivotal in determining level of genes expression especially on or around transcription start site (TSS) of a gene. Purpose of the current study was to determine nucleosome position around TSS of Rbl2/p130. We investigated Rbl2/p130 expression in connection to nucleosome positions around its TSS among breast tumors and their adjacent normal control tissues (ANCT) using micrococcal nuclease (MNAse) digestion assay and ChIP-PCR analysis. Three fold reduced Rbl2/p130 expression in these tumor tissues were noticed compared to their control tissues. DNA obtained from MNAse digested chromatin was used as PCR template. Region between - 137 to + 140 around TSS was scanned using 3 primer pairs (P1 = - 137 to + 69; P2 = - 90 to + 69; P3 = - 33 to + 140). ~ 66% breast tumors and ~ 26% ANCT samples were positive for P1. The difference was found statistically significant (p = 0.000) with an odd ratio (OD) of 9.143, suggesting that nucleosome formation in this region is ~ 9 times more probable in tumor samples. ~ 73% of the tumor and 60% ANCT were positive for P2, which although is significant (p = 0.035) with OD = 3.250, but less preferable than P1. However, P3 was not found to be a preferred area for nucleosome occupancy (p = 0.670; OD = 1.2). Negative correlations for nucleosome positions were observed especially for P1. Our results indicate that nucleosome are present slightly downstream of TSS in routine, while in case of breast carcinogenesis nucleosomes slides 55 bases upstream of the TSS, aligning + 1 position at the center of nucleosome, hence hindering access to the transcriptional machinery.


Subject(s)
Breast Neoplasms/genetics , Nucleosomes/metabolism , Retinoblastoma-Like Protein p130/genetics , Transcription Initiation Site , Breast Neoplasms/metabolism , Chromatin Assembly and Disassembly , Chromosome Mapping/methods , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Female , Genes, Tumor Suppressor , Humans , Immunoprecipitation/methods , Micrococcal Nuclease/metabolism , Nucleosomes/genetics , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Retinoblastoma-Like Protein p130/metabolism
18.
J Cell Physiol ; 233(9): 6965-6974, 2018 09.
Article in English | MEDLINE | ID: mdl-29336491

ABSTRACT

Gender-based differences may influence the occurrence of several ocular conditions suggesting the possibility that fluctuations in sex steroid homeostasis may have direct effects on the eye physiology. Here, we evaluated the effect of sex steroid hormone fluctuations in male retinal pigment epithelial cells, RPEs (ARPE-19). To mimic hormonal fluctuations occurring during aging, we exposed ARPE-19 to acute, prolonged or chronic estradiol, and progesterone challenges. We found that chronic estradiol treatment promotes a remarkable necrosis of RPE cells, and does not affect pRb2/p130 or PAI-2 sub-cellular localization. In contrast, chronic progesterone exposure induces nuclear subcellular rearrangement of pRb2/p130, co-immunolocalization of pRb2/p130 with PAI-2, and accumulation of cells in G2/M phase, which is accompanied by a remarkable reduction of necrosis in favour of apoptosis activation. This study has a high clinical significance since it considers sex steroid fluctuations as inducers of milieu change in the retina able to influence pathological situations occurring with aging in non-reproductive systems such as the eye. Exogenous administration of physiologically significant amounts of sex hormones for long periods of time is a common clinical practice for transgender patients seeking sex reassignment. In particular, our study offers the unique opportunity to unravel the effects of sex hormones, not only in determining gender differences but also in affecting the physiology of non-reproductive systems, such as the eye, in the underserved transgender community.


Subject(s)
Epithelial Cells/metabolism , Gonadal Steroid Hormones/pharmacology , Retinal Pigment Epithelium/pathology , Adult , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Epithelial Cells/drug effects , Epithelial Cells/pathology , Estradiol/pharmacology , Humans , Male , Necrosis , Plasminogen Activator Inhibitor 2/metabolism , Progesterone/pharmacology , Retinoblastoma-Like Protein p130/metabolism
19.
Nucleic Acids Res ; 46(1): 174-188, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29059406

ABSTRACT

The Histone 3 lysine 4 methylation (H3K4me3) mark closely correlates with active transcription. E2F-responsive promoters display dynamic changes in H3K4 methylation during the course of cell cycle progression. However, how and when these marks are reset, is not known. Here we show that the retinoblastoma binding protein RBP2/KDM5A, capable of removing tri-methylation marks on H3K4, associates with the E2F4 transcription factor via the pocket protein-p130-in a cell-cycle-stage specific manner. The association of RBP2 with p130 is LxCxE motif dependent. RNAi experiments reveal that p130 recruits RBP2 to E2F-responsive promoters in early G1 phase to bring about H3K4 demethylation and gene repression. A point mutation in LxCxE motif of RBP2 renders it incapable of p130-interaction and hence, repression of E2F-regulated gene promoters. We also examine how RBP2 may be recruited to non-E2F responsive promoters. Our studies provide insight into how the chromatin landscape needs to be adjusted rapidly and periodically during cell-cycle progression, concomitantly with temporal transcription, to bring about expression/repression of specific gene sets.


Subject(s)
E2F Transcription Factors/metabolism , Histones/metabolism , Promoter Regions, Genetic/genetics , Retinoblastoma-Binding Protein 2/metabolism , Retinoblastoma-Like Protein p130/metabolism , Retinol-Binding Proteins, Cellular/metabolism , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Cell Line , Cells, Cultured , E2F4 Transcription Factor/metabolism , G1 Phase/genetics , HeLa Cells , Humans , Methylation , Mice , Mutation , Protein Binding , RNA Interference , Retinoblastoma-Binding Protein 2/genetics , Retinoblastoma-Like Protein p130/genetics , Retinol-Binding Proteins, Cellular/genetics
20.
Int J Mol Sci ; 18(8)2017 Aug 16.
Article in English | MEDLINE | ID: mdl-28812991

ABSTRACT

The retinoblastoma (RB) tumor suppressor is known as a master regulator of the cell cycle. RB is mutated or functionally inactivated in the majority of human cancers. This transcriptional regulator exerts its function in cell cycle control through its interaction with the E2F family of transcription factors and with chromatin remodelers and modifiers that contribute to the repression of genes important for cell cycle progression. Over the years, studies have shown that RB participates in multiple processes in addition to cell cycle control. Indeed, RB is known to interact with over 200 different proteins and likely exists in multiple complexes. RB, in some cases, acts through its interaction with E2F1, other members of the pocket protein family (p107 and p130), and/or chromatin remodelers and modifiers. RB is a tumor suppressor with important chromatin regulatory functions that affect genomic stability. These functions include the role of RB in DNA repair, telomere maintenance, chromosome condensation and cohesion, and silencing of repetitive regions. In this review we will discuss recent advances in RB biology related to RB, partner proteins, and their non-transcriptional functions fighting back against genomic instability.


Subject(s)
DNA Repair , Gene Silencing , Genomic Instability , Retinoblastoma Protein/metabolism , Telomere Homeostasis , Animals , E2F Transcription Factors/genetics , E2F Transcription Factors/metabolism , Humans , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...