Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Biol Psychiatry ; 94(4): 341-351, 2023 08 15.
Article in English | MEDLINE | ID: mdl-36958377

ABSTRACT

BACKGROUND: Schizophrenia (SCZ) is caused by an interplay of polygenic risk and environmental factors, which may alter regulators of gene expression leading to pathogenic misexpression of SCZ risk genes. The CPEB family of RNA-binding proteins (CPEB1-4) regulates translation of target RNAs (approximately 40% of overall genes). We previously identified CPEB4 as a key dysregulated translational regulator in autism spectrum disorder (ASD) because its neuronal-specific microexon (exon 4) is mis-spliced in ASD brains, causing underexpression of numerous ASD risk genes. The genetic factors and pathogenic mechanisms shared between SCZ and ASD led us to hypothesize CPEB4 mis-splicing in SCZ leading to underexpression of multiple SCZ-related genes. METHODS: We performed MAGMA-enrichment analysis on Psychiatric Genomics Consortium genome-wide association study data and analyzed RNA sequencing data from the PsychENCODE Consortium. Reverse transcriptase polymerase chain reaction and Western blot were performed on postmortem brain tissue, and the presence/absence of antipsychotics was assessed through toxicological analysis. Finally, mice with mild overexpression of exon 4-lacking CPEB4 (CPEB4Δ4) were generated and analyzed biochemically and behaviorally. RESULTS: First, we found enrichment of SCZ-associated genes for CPEB4-binder transcripts. We also found decreased usage of CPEB4 microexon in SCZ probands, which was correlated with decreased protein levels of CPEB4-target SCZ-associated genes only in antipsychotic-free individuals. Interestingly, differentially expressed genes fit those reported for SCZ, specifically in the SCZ probands with decreased CPEB4-microexon inclusion. Finally, we demonstrated that mice with mild overexpression of CPEB4Δ4 showed decreased protein levels of CPEB4-target SCZ genes and SCZ-linked behaviors. CONCLUSIONS: We identified aberrant CPEB4 splicing and downstream misexpression of SCZ risk genes as a novel etiological mechanism in SCZ.


Subject(s)
Antipsychotic Agents , Autism Spectrum Disorder , Schizophrenia , Animals , Mice , Antipsychotic Agents/therapeutic use , Autism Spectrum Disorder/genetics , Brain/metabolism , Genetic Predisposition to Disease , Genome-Wide Association Study , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Schizophrenia/genetics , Schizophrenia/drug therapy
2.
iScience ; 25(2): 103790, 2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35243213

ABSTRACT

Upon tissue injury, cytokine expression reprogramming transiently remodels the inflammatory immune microenvironment to activate repair processes and subsequently return to homeostasis. However, chronic inflammation induces permanent changes in cytokine production which exacerbate tissue damage and may even favor tumor development. Here, we address the contribution of post-transcriptional regulation, by the RNA-binding protein CPEB4, to intestinal immune homeostasis and its role in inflammatory bowel diseases (IBD) and colorectal cancer (CRC) development. We found that intestinal damage induces CPEB4 expression in adaptive and innate immune cells, which is required for the translation of cytokine mRNA(s) such as the one encoding interleukin-22. Accordingly, CPEB4 is required for the development of gut-associated lymphoid tissues and to maintain intestinal immune homeostasis, mediating repair and remodeling after acute inflammatory tissue damage and promoting the resolution of intestinal inflammation. CPEB4 is chronically overexpressed in inflammatory cells in patients with IBD and in CRC, favoring tumor development.

3.
Sci Adv ; 6(20): eaax3868, 2020 05.
Article in English | MEDLINE | ID: mdl-32440535

ABSTRACT

Organogenesis is directed by coordinated cell proliferation and differentiation programs. The hierarchical networks of transcription factors driving mammary gland development and function have been widely studied. However, the contribution of posttranscriptional gene expression reprogramming remains largely unexplored. The 3' untranslated regions of messenger RNAs (mRNAs) contain combinatorial ensembles of cis-regulatory elements that define transcript-specific regulation of protein synthesis through their cognate RNA binding proteins. We analyze the contribution of the RNA binding cytoplasmic polyadenylation element-binding (CPEB) protein family, which collectively regulate mRNA translation for about 30% of the genome. We find that CPEB2 is required for the integration of hormonal signaling by controlling the protein expression from a subset of ER/PR- regulated transcripts. Furthermore, CPEB2 is critical for the development of ER-positive breast tumors. This work uncovers a previously unknown gene expression regulation level in breast morphogenesis and tumorigenesis, coordinating sequential transcriptional and posttranscriptional layers of gene expression regulation.


Subject(s)
Breast Neoplasms , Mammary Glands, Human , 3' Untranslated Regions , Breast Neoplasms/genetics , Female , Hormones , Humans , Mammary Glands, Human/metabolism , Organogenesis , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
4.
Sci Rep ; 9(1): 14065, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31575908

ABSTRACT

CD98 heavy chain (CD98hc) forms heteromeric amino acid (AA) transporters by interacting with different light chains. Cancer cells overexpress CD98hc-transporters in order to meet their increased nutritional and antioxidant demands, since they provide branched-chain AA (BCAA) and aromatic AA (AAA) availability while protecting cells from oxidative stress. Here we show that BCAA and AAA shortage phenocopies the inhibition of mTORC1 signalling, protein synthesis and cell proliferation caused by CD98hc ablation. Furthermore, our data indicate that CD98hc sustains glucose uptake and glycolysis, and, as a consequence, the pentose phosphate pathway (PPP). Thus, loss of CD98hc triggers a dramatic reduction in the nucleotide pool, which leads to replicative stress in these cells, as evidenced by the enhanced DNA Damage Response (DDR), S-phase delay and diminished rate of mitosis, all recovered by nucleoside supplementation. In addition, proper BCAA and AAA availability sustains the expression of the enzyme ribonucleotide reductase. In this regard, BCAA and AAA shortage results in decreased content of deoxynucleotides that triggers replicative stress, also recovered by nucleoside supplementation. On the basis of our findings, we conclude that CD98hc plays a central role in AA and glucose cellular nutrition, redox homeostasis and nucleotide availability, all key for cell proliferation.


Subject(s)
Amino Acids/metabolism , Cell Cycle , Fusion Regulatory Protein 1, Heavy Chain/metabolism , Nucleotides/metabolism , Amino Acids, Aromatic/metabolism , Amino Acids, Branched-Chain/metabolism , Cell Division , DNA Damage , DNA Repair , Fusion Regulatory Protein 1, Heavy Chain/physiology , Gene Expression Profiling , Gene Knockout Techniques , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Oxidative Stress
5.
Gut ; 66(7): 1306-1320, 2017 07.
Article in English | MEDLINE | ID: mdl-26984852

ABSTRACT

OBJECTIVE: Pathological neovascularisation is intimately involved in portal hypertension (PH). Here, we determined the contribution of vascular stem/progenitor cells (VSPCs) to neovessel growth in PH and whether the RNA-binding protein cytoplasmic polyadenylation element binding protein-4 (CPEB4) was behind the mechanism controlling VSPC function. DESIGN: To identify and monitor VSPCs in PH rats (portal vein-ligated), we used a combinatorial approach, including sphere-forming assay, assessment of self-renewal, 5-bromo-2'-desoxyuridine label retention technique, in vitro and in vivo stem/progenitor cell (SPC) differentiation and vasculogenic capability, cell sorting, as well as immunohistochemistry, immunofluorescence and confocal microscopy expression analysis. We also determined the role of CPEB4 on VSPC proliferation using genetically engineered mouse models. RESULTS: We demonstrated the existence in the mesenteric vascular bed of VSPCs displaying capability to form cellular spheres in suspension culture, self-renewal ability, expression of molecules commonly found in SPCs, slow-cycling features, in addition to other cardinal properties exhibited by SPCs, like capacity to differentiate into endothelial cells and pericytes with remarkable vasculogenic activity. Such VSPCs showed, after PH induction, an early switch in proliferation, and differentiated in vivo into endothelial cells and pericytes, contributing, structurally and functionally, to abnormal neovessel formation. Quantification of VSPC-dependent neovessel formation in PH further illustrated the key role played by VSPCs. We also demonstrated that CPEB4 regulates the proliferation of the activated VSPC progeny upon PH induction. CONCLUSIONS: These findings demonstrate that VSPC-derived neovessel growth (ie, vasculogenesis) and angiogenesis cooperatively stimulate mesenteric neovascularisation in PH and identify VSPC and CPEB4 as potential therapeutic targets.


Subject(s)
Hypertension, Portal/pathology , Neovascularization, Pathologic , RNA-Binding Proteins/metabolism , Stem Cells/cytology , Animals , Cell Differentiation , Cell Proliferation , Mice , Rats
6.
Bioessays ; 38 Suppl 1: S96-S106, 2016 07.
Article in English | MEDLINE | ID: mdl-27417127

ABSTRACT

Polo-like kinase 1 (PLK1) is a serine/threonine kinase that plays multiple and essential roles during the cell division cycle. Its inhibition in cultured cells leads to severe mitotic aberrancies and cell death. Whereas previous reports suggested that Plk1 depletion in mice leads to a non-mitotic arrest in early embryos, we show here that the bi-allelic Plk1 depletion in mice certainly results in embryonic lethality due to extensive mitotic aberrations at the morula stage, including multi- and mono-polar spindles, impaired chromosome segregation and cytokinesis failure. In addition, the conditional depletion of Plk1 during mid-gestation leads also to severe mitotic aberrancies. Our data also confirms that Plk1 is completely dispensable for mitotic entry in vivo. On the other hand, Plk1 haploinsufficient mice are viable, and Plk1-heterozygous fibroblasts do not harbor any cell cycle alterations. Plk1 is overexpressed in many human tumors, suggesting a therapeutic benefit of inhibiting Plk1, and specific small-molecule inhibitors for this kinase are now being evaluated in clinical trials. Therefore, the different Plk1 mouse models here presented are a valuable tool to reexamine the relevance of the mitotic kinase Plk1 during mammalian development and animal physiology.


Subject(s)
Cell Cycle Proteins/physiology , Chromosome Segregation , Cytokinesis , Mitosis , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins/physiology , Spindle Apparatus/metabolism , Animals , Female , Male , Mice , Spindle Apparatus/physiology , Polo-Like Kinase 1
7.
Gastroenterology ; 150(4): 982-97.e30, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26627607

ABSTRACT

BACKGROUND & AIMS: Vascular endothelial growth factor (VEGF) regulates angiogenesis, yet therapeutic strategies to disrupt VEGF signaling can interfere with physiologic angiogenesis. In a search for ways to inhibit pathologic production or activities of VEGF without affecting its normal production or functions, we investigated the post-transcriptional regulation of VEGF by the cytoplasmic polyadenylation element-binding proteins CPEB1 and CPEB4 during development of portal hypertension and liver disease. METHODS: We obtained transjugular liver biopsies from patients with hepatitis C virus-associated cirrhosis or liver tissues removed during transplantation; healthy human liver tissue was obtained from a commercial source (control). We also performed experiments with male Sprague-Dawley rats and CPEB-deficient mice (C57BL6 or mixed C57BL6/129 background) and their wild-type littermates. Secondary biliary cirrhosis was induced in rats by bile duct ligation, and portal hypertension was induced by partial portal vein ligation. Liver and mesenteric tissues were collected and analyzed in angiogenesis, reverse transcription polymerase chain reaction, polyA tail, 3' rapid amplification of complementary DNA ends, Southern blot, immunoblot, histologic, immunohistochemical, immunofluorescence, and confocal microscopy assays. CPEB was knocked down with small interfering RNAs in H5V endothelial cells, and translation of luciferase reporters constructs was assessed. RESULTS: Activation of CPEB1 promoted alternative nuclear processing within noncoding 3'-untranslated regions of VEGF and CPEB4 messenger RNAs in H5V cells, resulting in deletion of translation repressor elements. The subsequent overexpression of CPEB4 promoted cytoplasmic polyadenylation of VEGF messenger RNA, increasing its translation; the high levels of VEGF produced by these cells led to their formation of tubular structures in Matrigel assays. We observed increased levels of CPEB1 and CPEB4 in cirrhotic liver tissues from patients, compared with control tissue, as well as in livers and mesenteries of rats and mice with cirrhosis or/and portal hypertension. Mice with knockdown of CPEB1 or CPEB4 did not overexpress VEGF or have signs of mesenteric neovascularization, and developed less-severe forms of portal hypertension after portal vein ligation. CONCLUSIONS: We identified a mechanism of VEGF overexpression in liver and mesentery that promotes pathologic, but not physiologic, angiogenesis, via sequential and nonredundant functions of CPEB1 and CPEB4. Regulation of CPEB4 by CPEB1 and the CPEB4 autoamplification loop induces pathologic angiogenesis. Strategies to block the activities of CPEBs might be developed to treat chronic liver and other angiogenesis-dependent diseases.


Subject(s)
Hypertension, Portal/metabolism , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis/metabolism , Neovascularization, Pathologic , RNA-Binding Proteins/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , 3' Untranslated Regions , Adult , Animals , Case-Control Studies , Cell Line , Chronic Disease , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Hypertension, Portal/genetics , Hypertension, Portal/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/pathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Middle Aged , Polyadenylation , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Rats, Sprague-Dawley , Signal Transduction , Time Factors , Transcription Factors/deficiency , Transcription Factors/genetics , Transfection , mRNA Cleavage and Polyadenylation Factors/deficiency , mRNA Cleavage and Polyadenylation Factors/genetics
8.
Nat Cell Biol ; 17(10): 1304-16, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26322680

ABSTRACT

Blocking mitotic progression has been proposed as an attractive therapeutic strategy to impair proliferation of tumour cells. However, how cells survive during prolonged mitotic arrest is not well understood. We show here that survival during mitotic arrest is affected by the special energetic requirements of mitotic cells. Prolonged mitotic arrest results in mitophagy-dependent loss of mitochondria, accompanied by reduced ATP levels and the activation of AMPK. Oxidative respiration is replaced by glycolysis owing to AMPK-dependent phosphorylation of PFKFB3 and increased production of this protein as a consequence of mitotic-specific translational activation of its mRNA. Induction of autophagy or inhibition of AMPK or PFKFB3 results in enhanced cell death in mitosis and improves the anti-tumoral efficiency of microtubule poisons in breast cancer cells. Thus, survival of mitotic-arrested cells is limited by their metabolic requirements, a feature with potential implications in cancer therapies aimed to impair mitosis or metabolism in tumour cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy/physiology , Fibroblasts/metabolism , Glycolysis , M Phase Cell Cycle Checkpoints/physiology , Phosphofructokinase-2/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Autophagy/genetics , Blotting, Western , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/ultrastructure , Humans , M Phase Cell Cycle Checkpoints/genetics , MCF-7 Cells , Mice, Knockout , Mice, Nude , Microscopy, Confocal , Paclitaxel/pharmacology , Phosphofructokinase-2/genetics , RNA Interference , Xenograft Model Antitumor Assays
9.
Mol Cell Biol ; 35(20): 3566-78, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26240282

ABSTRACT

Aurora kinase B, one of the three members of the mammalian Aurora kinase family, is the catalytic component of the chromosomal passenger complex, an essential regulator of chromosome segregation in mitosis. Aurora B is overexpressed in human tumors although whether this kinase may function as an oncogene in vivo is not established. Here, we report a new mouse model in which expression of the endogenous Aurkb locus can be induced in vitro and in vivo. Overexpression of Aurora B in cultured cells induces defective chromosome segregation and aneuploidy. Long-term overexpression of Aurora B in vivo results in aneuploidy and the development of multiple spontaneous tumors in adult mice, including a high incidence of lymphomas. Overexpression of Aurora B also results in a reduced DNA damage response and decreased levels of the p53 target p21(Cip1) in vitro and in vivo, in line with an inverse correlation between Aurora B and p21(Cip1) expression in human leukemias. Thus, overexpression of Aurora B may contribute to tumor formation not only by inducing chromosomal instability but also by suppressing the function of the cell cycle inhibitor p21(Cip1).


Subject(s)
Aneuploidy , Aurora Kinase B/physiology , Cyclin-Dependent Kinase Inhibitor p21/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Fibroblasts/metabolism , Gene Expression , Gene Silencing , Mice, Inbred C57BL , Precursor Cell Lymphoblastic Leukemia-Lymphoma/enzymology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics
10.
Cancer Res ; 73(22): 6804-15, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24242071

ABSTRACT

Aurora-A is a kinase involved in the formation and maturation of the mitotic spindle and chromosome segregation. This kinase is frequently overexpressed in human cancer, and its activity may confer resistance to antitumoral drugs such as Taxol. Inhibition of Aurora-A results in mitotic defects, and this kinase is considered as an attractive therapeutic target for cancer. Nevertheless, the specific requirements for this kinase in adult mammalian tissues remain unclear. Conditional genetic ablation of Aurora-A in adult tissues results in polyploid cells that display a DNA-damage-like response characterized by the upregulation of p53 and the cell-cycle inhibitor p21(Cip1). This is accompanied by apoptotic, differentiation, or senescence markers in a tissue-specific manner. Therapeutic elimination of Aurora-A prevents the progression of skin and mammary gland tumors. However, this is not due to significant levels of apoptosis or senescence, but because Aurora-A-deficient tumors accumulate polyploid cells with limited proliferative potential. Thus, Aurora-A is required for tumor formation in vivo, and the differential response observed in various tissues might have relevant implications in current therapeutic strategies aimed at inhibiting this kinase in the treatment of human cancer.


Subject(s)
Aurora Kinase A/physiology , Cell Transformation, Neoplastic/genetics , Neoplasms/genetics , Regeneration/genetics , Animals , Aurora Kinase A/genetics , Cells, Cultured , Embryo, Mammalian , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic
11.
Cell Cycle ; 12(7): 1030-41, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23428904

ABSTRACT

Aurora kinase B is a critical component of the chromosomal passenger complex, which is involved in the regulation of microtubule-kinetochore attachments and cytokinesis. By using conditional knockout cells and chemical inhibition, we show here that inactivation of Aurora B results in delayed G(1)/S transition and premature mitotic exit. Aurora B deficiency results in delayed DNA replication in cultured fibroblasts as well as liver cells after hepatectomy. This is accompanied by increased transcription of the cell cycle inhibitor p21 (Cip1). Lack of Aurora B does not prevent mitotic entry but results in a premature exit from prometaphase in the presence of increased p21(Cip1)-Cdk1 inactive complexes. Aurora B-null cells display reduced degradation of cyclin B1, suggesting the presence of phenomenon known as adaptation to the mitotic checkpoint, previously described in yeast. Elimination of p21(Cip1) rescues Cdk1 activity and prevents premature mitotic exit in Aurora B-deficient cells. These results suggest that Aurora B represses p21(Cip1), preventing delayed DNA replication, Cdk inhibition and premature mitotic exit. The upregulation of p21(Cip1) observed after inhibition of Aurora B may have important implications in cell cycle progression, tetraploidy, senescence or cancer therapy.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/metabolism , Mitosis , Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinase B , Aurora Kinases , CDC2 Protein Kinase/metabolism , Cell Line , Cyclin B1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Replication , G1 Phase Cell Cycle Checkpoints , Interphase , Liver/metabolism , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , RNA Interference , RNA, Small Interfering/metabolism
12.
Ageing Res Rev ; 11(4): 460-72, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22542725

ABSTRACT

Cytoplasmic elongation of the poly(A) tail was originally identified as a mechanism to activate maternal mRNAs, stored as silent transcripts with short poly(A) tails, during meiotic progression. A family of RNA-binding proteins named CPEBs, which recruit the translational repression or cytoplasmic polyadenylation machineries to their target mRNAs, directly mediates cytoplasmic polyadenylation. Recent years have witnessed an explosion of studies showing that CPEBs are not only expressed in a variety of somatic tissues, but have essential functions controlling gene expression in time and space in the adult organism. These "new" functions of the CPEBs include regulating the balance between senescence and proliferation and its pathological manifestation, tumor development. In this review, we summarize current knowledge on the functions of the CPEB-family of proteins in the regulation of cell proliferation, their target mRNAs and the mechanism controlling their activities.


Subject(s)
Aging/genetics , Gene Expression Regulation, Neoplastic/physiology , Neoplasms/genetics , Protein Biosynthesis/genetics , Transcription Factors/genetics , mRNA Cleavage and Polyadenylation Factors/genetics , Animals , Humans , RNA Processing, Post-Transcriptional/genetics , Transcription Factors/physiology , mRNA Cleavage and Polyadenylation Factors/physiology
13.
Nat Med ; 18(1): 83-90, 2011 Dec 04.
Article in English | MEDLINE | ID: mdl-22138752

ABSTRACT

Malignant transformation, invasion and angiogenesis rely on the coordinated reprogramming of gene expression in the cells from which the tumor originated. Although deregulated gene expression has been extensively studied at genomic and epigenetic scales, the contribution of the regulation of mRNA-specific translation to this reprogramming is not well understood. Here we show that cytoplasmic polyadenylation element binding protein 4 (CPEB4), an RNA binding protein that mediates meiotic mRNA cytoplasmic polyadenylation and translation, is overexpressed in pancreatic ductal adenocarcinomas and glioblastomas, where it supports tumor growth, vascularization and invasion. We also show that, in pancreatic tumors, the pro-oncogenic functions of CPEB4 originate in the translational activation of mRNAs that are silenced in normal tissue, including the mRNA of tissue plasminogen activator, a key contributor to pancreatic ductal adenocarcinoma malignancy. Taken together, our results document a key role for post-transcriptional gene regulation in tumor development and describe a detailed mechanism for gene expression reprogramming underlying malignant tumor progression.


Subject(s)
Adenocarcinoma/pathology , Glioblastoma/pathology , Pancreatic Ducts/pathology , Pancreatic Neoplasms/pathology , Protein Biosynthesis/genetics , RNA-Binding Proteins/metabolism , Adenocarcinoma/blood supply , Adenocarcinoma/genetics , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/blood supply , Glioblastoma/genetics , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/genetics , Pancreatic Ducts/blood supply , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/genetics , Polyadenylation , RNA, Small Interfering/genetics , RNA-Binding Proteins/genetics
14.
Development ; 138(13): 2661-72, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21613325

ABSTRACT

Mitosis is controlled by multiple kinases that drive cell cycle progression and prevent chromosome mis-segregation. Aurora kinase B interacts with survivin, borealin and incenp to form the chromosomal passenger complex (CPC), which is involved in the regulation of microtubule-kinetochore attachments and cytokinesis. Whereas genetic ablation of survivin, borealin or incenp results in early lethality at the morula stage, we show here that aurora B is dispensable for CPC function during early cell divisions and aurora B-null embryos are normally implanted. This is due to a crucial function of aurora C during these early embryonic cycles. Expression of aurora C decreases during late blastocyst stages resulting in post-implantation defects in aurora B-null embryos. These defects correlate with abundant prometaphase figures and apoptotic cell death of the aurora B-deficient inner cell mass. Conditional deletion of aurora B in somatic cells that do not express aurora C results in chromosomal misalignment and lack of chromosome segregation. Re-expression of wild-type, but not kinase-dead, aurora C rescues this defect, suggesting functional overlap between these two kinases. Finally, aurora B-null cells partially arrest in the presence of nocodazole, suggesting that this kinase is not essential for the spindle assembly checkpoint.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Animals , Aurora Kinase B , Aurora Kinase C , Aurora Kinases , Blastocyst/metabolism , Cell Division/genetics , Cell Division/physiology , Cells, Cultured , Chromosome Segregation/genetics , Chromosome Segregation/physiology , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Mice , Mice, Transgenic , Mitosis/genetics , Mitosis/physiology , Pregnancy , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Zygote/metabolism
15.
Front Oncol ; 1: 50, 2011.
Article in English | MEDLINE | ID: mdl-22649767

ABSTRACT

Aurora-A is a serine/threonine kinase that plays critical roles in centrosome maturation, spindle dynamics, and chromosome orientation and it is frequently over-expressed in human cancers. In this work, we show that Aurora-A interacts with the SUMO-conjugating enzyme UBC9 and co-localizes with SUMO1 in mitotic cells. Aurora-A can be SUMOylated in vitro and in vivo. Mutation of the highly conserved SUMOylation residue lysine 249 significantly disrupts Aurora-A SUMOylation and mitotic defects characterized by defective and multipolar spindles ensue. The Aurora-A(K249R) mutant has normal kinase activity but displays altered dynamics at the mitotic spindle. In addition, ectopic expression of the Aurora-A(K249R) mutant results in a significant increase in susceptibility to malignant transformation induced by the Ras oncogene. These data suggest that modification by SUMO residues may control Aurora-A function at the spindle and that deficiency of SUMOylation of this kinase may have important implications for tumor development.

16.
Cancer Res ; 70(22): 9349-59, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20861187

ABSTRACT

Brick1 (Brk1) is the less-studied component of the Wave/Scar pathway involved in the branched nucleation of actin fibers. The clinical relevance of Brk1 is emphasized by correlative data showing that Von Hippel-Lindau (VHL) patients that also lose the BRK1 gene are protected against the development of tumors. This contrasts with recent evidence suggesting that the Wave complex may function as an invasion suppressor in epithelial cancers. Here, we show that the downregulation of Brk1 results in abnormal actin stress fiber formation and vinculin distribution and loss of Arp2/3 and Wave proteins at the cellular protrusions. Brk1 is required for cell proliferation and cell transformation by oncogenes. In addition, Brk1 downregulation results in defective directional migration and invasive growth in renal cell carcinoma cells as well as in other tumor cell types. Finally, genetic ablation of Brk1 results in dramatic defects in embryo compaction and development, suggesting an essential role for this protein in actin dynamics. Thus, genetic loss or inhibition of BRK1 is likely to be protective against tumor development due to proliferation and motility defects in affected cells.


Subject(s)
Actins/metabolism , Cell Transformation, Neoplastic , Cytoskeletal Proteins/physiology , Cytoskeleton/metabolism , Embryonic Development/physiology , Animals , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Down-Regulation , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Mice, SCID , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
17.
J Cell Sci ; 123(Pt 15): 2586-95, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20605920

ABSTRACT

RINGO/Speedy proteins are direct activators of Cdk1 and Cdk2 that have no sequence homology to cyclins. We have characterized the role in cell-cycle progression of a new human member of this protein family referred to as RINGO C. We show that siRNA-mediated knockdown of RINGO C results in premature mitotic exit with misaligned chromosomes, even in the presence of microtubule poisons. Time-lapse-microscopy experiments suggest that RINGO C is involved in the spindle-assembly checkpoint (SAC). Consistent with this idea, RINGO-C-depleted cells show impaired recruitment of the SAC components Mad2, Bub1 and BubR1. As the checkpoint is overridden, cells display defective chromosome segregation, which leads to an increased number of micronuclei and binucleated structures. Intriguingly, we found that RINGO C can associate with the mitotic kinase Aurora B, and downregulation of RINGO C produces mislocalization of the active form of Aurora B in prometaphase. Taken together, our results indicate a role for RINGO C in the mitotic checkpoint, which might be mediated by defective recruitment of SAC components and deregulation of the activity of Aurora kinase B.


Subject(s)
Cell Cycle Proteins/metabolism , Spindle Apparatus/metabolism , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/genetics , Cell Line , Cell Line, Tumor , Chromosomes, Human/drug effects , Chromosomes, Human/genetics , Flow Cytometry , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Kinetochores/metabolism , Metaphase/drug effects , Metaphase/genetics , Microscopy, Confocal , Microscopy, Fluorescence , Mitosis/drug effects , Mitosis/genetics , Nocodazole/pharmacology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Spindle Apparatus/drug effects
18.
J Cell Sci ; 123(Pt 16): 2823-33, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20663916

ABSTRACT

Aurora kinases are central regulators of mitotic-spindle assembly, chromosome segregation and cytokinesis. Aurora B is a member of the chromosomal passenger complex (CPC) with crucial functions in regulation of the attachment of kinetochores to microtubules and in cytokinesis. We report here that Aurora B contains a conserved SUMO modification motif within its kinase domain. Aurora B can bind SUMO peptides in vitro when bound to the IN-box domain of its CPC partner INCENP. Mutation of Lys207 to arginine (Aurora B(K207R)) impairs the formation of conjugates of Aurora B and SUMO in vivo. Expression of the SUMO-null form of Aurora B results in abnormal chromosome segregation and cytokinesis failure and it is not able to rescue mitotic defects in Aurora-B-knockout cells. These defects are accompanied by increased levels of the CPC on chromosome arms and defective centromeric function, as detected by decreased phosphorylation of the Aurora-B substrate CENP-A. The Aurora-B(K207R) mutant does not display reduced kinase activity, suggesting that functional defects are probably a consequence of the altered localization, rather than decreased intrinsic kinase activity. These data suggest that SUMOylation of Aurora B modulates its function, possibly by mediating the extraction of CPC complexes from chromosome arms during prometaphase.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , SUMO-1 Protein/metabolism , Animals , Aurora Kinase B , Aurora Kinases , Cell Cycle Proteins/metabolism , Cell Survival , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Chromosome Segregation , Cytokinesis/genetics , HeLa Cells , Humans , Mice , Mutation , Protein Serine-Threonine Kinases/genetics , Spindle Apparatus/genetics , Spindle Apparatus/metabolism , Sumoylation , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...