Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 848
Filter
1.
Cell Death Dis ; 15(1): 81, 2024 01 22.
Article in English | MEDLINE | ID: mdl-38253523

ABSTRACT

A core pathophysiologic feature underlying many respiratory diseases is multiciliated cell dysfunction, leading to inadequate mucociliary clearance. Due to the prevalence and highly variable etiology of mucociliary dysfunction in respiratory diseases, it is critical to understand the mechanisms controlling multiciliogenesis that may be targeted to restore functional mucociliary clearance. Multicilin, in a complex with E2F4, is necessary and sufficient to drive multiciliogenesis in airway epithelia, however this does not apply to all cell types, nor does it occur evenly across all cells in the same cell population. In this study we further investigated how co-factors regulate the ability of Multicilin to drive multiciliogenesis. Combining data in mouse embryonic fibroblasts and human bronchial epithelial cells, we identify RBL2 as a repressor of the transcriptional activity of Multicilin. Knockdown of RBL2 in submerged cultures or phosphorylation of RBL2 in response to apical air exposure, in the presence of Multicilin, allows multiciliogenesis to progress. These data demonstrate a dynamic interaction between RBL2 and Multicilin that regulates the capacity of cells to differentiate and multiciliate. Identification of this mechanism has important implications for facilitating MCC differentiation in diseases with impaired mucociliary clearance.


Subject(s)
Fibroblasts , Respiratory Tract Diseases , Animals , Mice , Humans , Cell Differentiation/genetics , Epithelial Cells , Epithelium , Retinoblastoma-Like Protein p130
2.
PLoS One ; 17(11): e0269122, 2022.
Article in English | MEDLINE | ID: mdl-36413521

ABSTRACT

RB is a well-known cell cycle regulator controlling the G1 checkpoint. Previous reports have suggested that it can influence cell fate decisions not only by regulating cell proliferation and survival but also by interacting with transcription factors and epigenetic modifiers. However, the functional redundancy of RB family proteins (RB, RBL1 and RBL2) renders it difficult to investigate their roles during early development, especially in human. Here, we address this problem by generating human embryonic stem cells lacking RB family proteins. To achieve this goal, we first introduced frameshift mutations in RBL1 and RBL2 genes using the CRISPR/Cas9 technology, and then integrated the shRNA-expression cassette to knockdown RB upon tetracycline treatment. The resulting RBL1/2_dKO+RB_iKD cells remain pluripotent and efficiently differentiate into the primary germ layers in vitro even in the absence of the RB family proteins. In contrast, we observed that subsequent differentiation into foregut endoderm was impaired without the expression of RB, RBL1 and RBL2. Thus, it is suggested that RB proteins are dispensable for the maintenance and acquisition of cell identities during early development, but they are essential to generate advanced derivatives after the formation of primary germ layers. These results also indicate that our RBL1/2_dKO+RB_iKD cell lines are useful to depict the detailed molecular roles of RB family proteins in the maintenance and generation of various cell types accessible from human pluripotent stem cells.


Subject(s)
Human Embryonic Stem Cells , Pluripotent Stem Cells , Humans , Cell Differentiation/physiology , Endoderm/metabolism , Human Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma-Like Protein p130/genetics
3.
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
4.
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
5.
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
6.
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
7.
PLoS One ; 17(4): e0266196, 2022.
Article in English | MEDLINE | ID: mdl-35385527

ABSTRACT

Retinoblastoma like protein-2 (Rbl2) is functionally regulated by phosphorylation and acetylation. Previously, we demonstrated that lysine K1083 (K1079 in human Rbl2) is a potential target for acetylation but its functional role remains elusive. We investigated alterations in human Rbl2 gene specifically targeting exons 19-22 harbouring acetylatable residues i.e. K1072, K1083 and K1115 through single stranded conformation polymorphism (SSCP) in breast cancer patients. The K1083 was found altered into arginine (R) in 51% of the cases but K1072 and K1115 remained conserved. The 'K1083R' mutation impairs the acetylation potential of this motif that may result in functional inactivation of Rbl2. These patients also showed poor survival outcome that highlights prognostic relevance of this residue. NIH3T3 cells expressing glutamine (K1083Q) mutated Rbl2 could not be arrested in G1 by serum starvation, whereas cells expressing Rbl2 with K1083R showed prolonged G1 arrest in fluorescence activated cell sorting (FACS) analysis. This suggests that K1083 acetylation is important for G1/S transition. Further, we performed molecular dynamic simulations (MDS) to analyse kinetics of residue K1083 with Cyc-D1/CDK4. Mutations at K1083 impaired this binding exposing neighbouring residues S1080, P1081, S1082 and R1084, hence enhancing the possibility of accelerated phosphorylation. S1080 has previously been reported as a promising candidate of cell cycle dependent phosphorylation in Rbl2. This highlights significance of mutations in the pocket domain of Rbl2 gene in breast cancer, and also strengthen the supposition that K1083 acetylation is pre-requisite for its phosphorylation.


Subject(s)
Breast Neoplasms , Retinoblastoma-Like Protein p130 , Acetylation , Animals , Breast Neoplasms/genetics , Female , Humans , Mice , Mutation , NIH 3T3 Cells , Retinoblastoma-Like Protein p130/genetics
8.
Cancer Lett ; 533: 215595, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35182659

ABSTRACT

Homeobox B9 (HOXB9) is involved in the occurrence and development of malignant tumors. However, the functions and underlying molecular mechanisms of HOXB9 in pancreatic cancer have yet to be identified. In this study, we find that both HOXB9 mRNA and protein levels are down-regulated in pancreatic cancer tissues and cell lines. Kaplan-Meier survival plots of 150 pancreatic cancer cases show that higher expression of HOXB9 in pancreatic cancer patients is associated with higher survival rates. We also find that over-expression of HOXB9 inhibits pancreatic cancer cell proliferation both in cell lines and the nude mouse xenograft as well as PDX models. Applying cell cycle PCR array analysis, Flow CytoMetry, ChIP-qPCR, and luciferase experiments, we observe that HOXB9 blocks cell cycle progression in the G0/G1 phase via up-regulating RBL2 and inhibiting c-Myc, and we further find that DNMT1 inhibits the expression of HOXB9 in pancreatic cancer by promoting the methylation of its promoter. Our findings highlight a novel mechanism of the DNMT1/HOXB9/RBL2/c-Myc pathway in regulating the cell cycle and proliferation of pancreatic cancer cells and provide a research basis for the prognosis and therapeutic application of HOXB9 in pancreatic cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Animals , Cell Cycle/genetics , Cell Division , Cell Line, Tumor , Cell Proliferation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Mice , Pancreatic Neoplasms/genetics , Retinoblastoma-Like Protein p130/genetics , Pancreatic Neoplasms
9.
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
10.
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
11.
Mol Biol Rep ; 48(6): 5121-5133, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34169395

ABSTRACT

The High-Risk Human Papillomaviruses (HR-HPVs) 16 and 18 are known to cause cervical cancer, which is primarily attributed to E6 and E7 oncoproteins. In addition, recent studies have focused on the vital role of the p130 pocket protein as an oncosuppressor to limit the expression of E2F transcription factors required for cell cycle progression. In view of this, the current study was conducted to investigate the mechanism by which transfection with HPV16/18 E7 leads to the deregulation of the host cell cycle, altering the localisation of p130, and expression of differentiation genes in Human Keratinocytes (HaCaT) cells. Co-immunoprecipitation, Western blot analysis, immunofluorescence microscopy, flow cytometry, quantitative-Polymerase Chain Reaction (qPCR), and the inhibition of p130 by MG132 inhibitor were employed to investigate the loss of p130 and its disruption in HPV 16/18 E7-transfected HaCaT cells. The HPV16- and HPV18-transformed cells, known as CaSki and HeLa, respectively, were also used to complement the ectopic expressions of E7 in HaCaT cells. Normal keratinocytes displayed higher level of p130 expression than HPV-transformed cells. In addition, the immunofluorescence analysis revealed that both HPV 16/18 E7-transfected HaCaT and HPV-transformed cells exhibited higher level of cytoplasmic p130 compared to nuclear p130. A significant increase in the number of S/G2 phase cells in HPV-transformed cells was also recorded since E7 has been shown to stimulate proliferation through the deactivation of Retinoblastoma Protein (pRB)-dependent G1/S checkpoint. Furthermore, the findings recorded the down-regulation of keratinocyte differentiation markers, namely p130, keratin10, and involucrin. The proteasomal degradation of the exported p130 confirmed the cellular localisation pattern of p130, which was commonly observed in cancerous cells. The findings provide strong evidence that the localisation of nuclear p130 nuclear was disrupted by HPV16/18 E7 led to the deregulation of the cell cycle and the impairment of cellular differentiation ultimately lead to cellular transformation.


Subject(s)
Crk-Associated Substrate Protein/metabolism , DNA-Binding Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/metabolism , Alphapapillomavirus/genetics , Alphapapillomavirus/pathogenicity , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Division/physiology , Cell Line , Crk-Associated Substrate Protein/genetics , DNA-Binding Proteins/genetics , Female , HeLa Cells , Human papillomavirus 16/metabolism , Human papillomavirus 16/pathogenicity , Human papillomavirus 18/metabolism , Human papillomavirus 18/pathogenicity , Humans , Keratinocytes/metabolism , Oncogene Proteins, Viral/genetics , Papillomaviridae/genetics , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/genetics , Repressor Proteins/genetics , Retinoblastoma-Like Protein p130/genetics , Transfection , Uterine Cervical Neoplasms/metabolism
12.
J Hum Genet ; 66(11): 1101-1112, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33980986

ABSTRACT

RBL2/p130, a member of the retinoblastoma family of proteins, is a key regulator of cell division and propagates irreversible senescence. RBL2/p130 is also involved in neuronal differentiation and survival, and eliminating Rbl2 in certain mouse strains leads to embryonic lethality accompanied by an abnormal central nervous system (CNS) phenotype. Conflicting reports exist regarding a role of RBL2/p130 in transcriptional regulation of DNA methyltransferases (DNMTs), as well as the control of telomere length. Here we describe the phenotype of three patients carrying bi-allelic RBL2-truncating variants. All presented with infantile hypotonia, severe developmental delay and microcephaly. Malignancies were not reported in carriers or patients. Previous studies carried out on mice and human cultured cells, associated RBL2 loss to DNA methylation and telomere length dysregulation. Here, we investigated whether patient cells lacking RBL2 display related abnormalities. The study of primary patient fibroblasts did not detect abnormalities in expression of DNMTs. Furthermore, methylation levels of whole genome DNA, and specifically of pericentromeric repeats and subtelomeric regions, were unperturbed. RBL2-null fibroblasts show no evidence for abnormal elongation by telomeric recombination. Finally, gradual telomere shortening, and normal onset of senescence were observed following continuous culturing of RBL2-mutated fibroblasts. Thus, this study resolves uncertainties regarding a potential non-redundant role for RBL2 in DNA methylation and telomere length regulation, and indicates that loss of function variants in RBL2 cause a severe autosomal recessive neurodevelopmental disorder in humans.


Subject(s)
Cognitive Dysfunction/genetics , DNA Methylation/genetics , Retinoblastoma-Like Protein p130/genetics , Telomere Shortening/genetics , Adolescent , Adult , Alleles , Animals , Child , Cognitive Dysfunction/complications , Cognitive Dysfunction/physiopathology , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Fibroblasts/metabolism , Genetic Predisposition to Disease , Humans , Male , Methyltransferases/genetics , Mice , Microcephaly/complications , Microcephaly/genetics , Microcephaly/physiopathology , Motor Activity/physiology , Muscle Hypotonia/complications , Muscle Hypotonia/genetics , Muscle Hypotonia/physiopathology , Telomere/genetics , Exome Sequencing
13.
Cancer Sci ; 112(7): 2921-2927, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33934450

ABSTRACT

Mature teratomas are usually benign tumors that rarely undergo malignant transformation. We report an advanced neuroblastoma arising in a mature teratoma of the ovary. Whole-exome sequencing identified extensive copy-neutral loss of heterozygosity (LOH) in both neuroblastoma and teratoma elements, suggesting that the neuroblastoma evolved from the teratoma. In addition, several truncating germline heterozygous variants in tumor suppressor genes, including RBL2 and FBXW12, became homozygous as a result of LOH. Collectively, we speculate that extensive LOH in teratoma cells may force heterozygous germline variants to become homozygous, which, in turn, may contribute to the development of neuroblastoma with the acquisition of additional chromosomal changes.


Subject(s)
Germ-Line Mutation , Loss of Heterozygosity , Neoplasms, Multiple Primary/genetics , Neuroblastoma/genetics , Ovarian Neoplasms/genetics , Teratoma/genetics , Adolescent , F-Box Proteins/genetics , Female , Homozygote , Humans , Neoplasms, Multiple Primary/drug therapy , Neoplasms, Multiple Primary/pathology , Neuroblastoma/drug therapy , Neuroblastoma/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Retinoblastoma-Like Protein p130/genetics , Teratoma/drug therapy , Teratoma/pathology , Exome Sequencing
14.
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
15.
Differentiation ; 113: 1-9, 2020.
Article in English | MEDLINE | ID: mdl-32120156

ABSTRACT

The functional maturation of human pancreatic ß-cells remains poorly understood. EndoC-ßH2 is a human ß-cell line with a reversible immortalized phenotype. Removal of the two oncogenes, SV40LT and hTERT introduced for its propagation, stops proliferation, triggers cell size increase and senescence, promotes mitochondrial activity and amplifies several ß-cell traits and functions. Overall, these events recapitulate several aspects of functional ß-cell maturation. We report here that selective depletion of SV40LT, but not of hTERT, is sufficient to revert EndoC-ßH2 immortalization. SV40LT inhibits the activity of the RB family members and of P53. In EndoC-ßH2 cells, the knock-down of RB itself, and, to a lesser extent, of its relative P130, precludes most events triggered by SV40LT depletion. In contrast, the knock-down of P53 does not prevent reversion of immortalization. Thus, an increase in RB and P130 activity, but not in P53 activity, is required for functional maturation of EndoC-ßH2 cells upon SV40LT-depletion. In addition, RB and/or P130 depletion in SV40LT-expressing EndoC-ßH2 cells decreases cell size, stimulates proliferation, and decreases the expression of key ß-cell genes. Thus, despite SV40LT expression, EndoC-ßH2 cells have a residual RB activity, which when suppressed reverts them to a more immature phenotype. These results show that the expression and activity levels of RB family members, especially RB itself, regulate the maturation state of EndoC-ßH2 cells.


Subject(s)
Genes, Retinoblastoma , Insulin-Secreting Cells/metabolism , Retinoblastoma Protein/physiology , Antigens, Polyomavirus Transforming/genetics , Cell Cycle , Cell Line , Cell Proliferation , Cellular Senescence , Gene Knockdown Techniques , Humans , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/cytology , Multigene Family , RNA, Small Interfering , Retinoblastoma-Like Protein p130/physiology , Telomerase/genetics , Transcription, Genetic , Tumor Suppressor Protein p53/physiology
16.
Ann Clin Transl Neurol ; 7(3): 390-396, 2020 03.
Article in English | MEDLINE | ID: mdl-32105419

ABSTRACT

The RBL2 locus has been associated with intelligence and educational attainment but not with a monogenic disorder to date. RBL2 encodes p130, a member of the retinoblastoma protein family, which is involved in mediating neuron survival and death. Previous studies on p130 knockout mice revealing embryonic death and impaired neurogenesis underscore the importance of RBL2 in brain development. Exome sequencing in two siblings with severe intellectual disability, stereotypies and dysmorphic features identified biallelic loss-of-function variants c.556C>T, p.(Arg186Ter) and a deletion of exon 13-17 in RBL2 (NM_005611.3), establishing RBL2 as a candidate gene for an autosomal recessive neurodevelopmental disorder.


Subject(s)
Neurodevelopmental Disorders/genetics , Retinoblastoma-Like Protein p130/genetics , Adolescent , Alleles , Developmental Disabilities/genetics , Face/abnormalities , Female , Humans , Intellectual Disability/genetics , Loss of Function Mutation , Magnetic Resonance Imaging , Male , Pedigree , Seizures/genetics , Siblings , Stereotypic Movement Disorder/genetics , Exome Sequencing
17.
Proc Natl Acad Sci U S A ; 117(1): 513-521, 2020 01 07.
Article in English | MEDLINE | ID: mdl-31871154

ABSTRACT

Small cell lung cancer (SCLC) is a highly aggressive subtype of lung cancer that remains among the most lethal of solid tumor malignancies. Recent genomic sequencing studies have identified many recurrently mutated genes in human SCLC tumors. However, the functional roles of most of these genes remain to be validated. Here, we have adapted the CRISPR-Cas9 system to a well-established murine model of SCLC to rapidly model loss-of-function mutations in candidate genes identified from SCLC sequencing studies. We show that loss of the gene p107 significantly accelerates tumor progression. Notably, compared with loss of the closely related gene p130, loss of p107 results in fewer but larger tumors as well as earlier metastatic spread. In addition, we observe differences in proliferation and apoptosis as well as altered distribution of initiated tumors in the lung, resulting from loss of p107 or p130 Collectively, these data demonstrate the feasibility of using the CRISPR-Cas9 system to model loss of candidate tumor suppressor genes in SCLC, and we anticipate that this approach will facilitate efforts to investigate mechanisms driving tumor progression in this deadly disease.


Subject(s)
Gene Editing/methods , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/genetics , Animals , Apoptosis/genetics , CRISPR-Cas Systems/genetics , Cell Line , Cell Proliferation/genetics , Disease Models, Animal , Disease Progression , Feasibility Studies , Humans , Loss of Function Mutation , Lung/pathology , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Staging , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p130/genetics , Small Cell Lung Carcinoma/pathology , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...