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










Publication year range
1.
Semin Cancer Biol ; 99: 45-55, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38346544

ABSTRACT

Accurate and complete DNA duplication is critical for maintaining genome integrity. Multiple mechanisms regulate when and where DNA replication takes place, to ensure that the entire genome is duplicated once and only once per cell cycle. Although the bulk of the genome is copied during the S phase of the cell cycle, increasing evidence suggests that parts of the genome are replicated in G2 or mitosis, in a last attempt to secure that daughter cells inherit an accurate copy of parental DNA. Remaining unreplicated gaps may be passed down to progeny and replicated in the next G1 or S phase. These findings challenge the long-established view that genome duplication occurs strictly during the S phase, bridging DNA replication to DNA repair and providing novel therapeutic strategies for cancer treatment.


Subject(s)
DNA Replication , Mitosis , Humans , S Phase/genetics , Cell Cycle/genetics , DNA Replication/genetics , Mitosis/genetics , DNA
2.
Clin Exp Med ; 23(3): 871-885, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35729367

ABSTRACT

Ras suppressor-1 (RSU1), originally described as a suppressor of Ras oncogenic transformation, localizes to focal adhesions interacting with the ILK-PINCH-PARVIN (IPP) complex that exerts a well-established oncogenic role in cancer. However, RSU1 implication in lung cancer is currently unknown. Our study aims to address the role of RSU1 in lung adenocarcinoma (LUADC). We here show that RSU1 protein expression by immunohistochemistry is downregulated in LUADC human tissue samples and represents a significant prognostic indicator. In silico analysis of gene chip and RNA seq data validated our findings. Depletion of RSU1 by siRNA in lung cancer cells promotes anchorage-independent cell growth, cell motility and epithelial to mesenchymal transition (EMT). Silencing of RSU1 also alters IPP complex expression in lung cancer cells. The p29 RSU1 truncated isoform is detected in lung cancer cells, and its expression is downregulated upon RSU1 silencing, whereas it is overexpressed upon ILK overexpression. These findings suggest that RSU1 exerts a tumor suppressive role with prognostic significance in LUADC.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Epithelial-Mesenchymal Transition , Prognosis , Adenocarcinoma of Lung/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Cell Movement , Cell Line, Tumor , Transcription Factors/metabolism
3.
Int J Mol Sci ; 23(23)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36499229

ABSTRACT

Data on animals emphasize the importance of the neuronal glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) for feeding suppression, although it is unclear whether astrocytes participate in the transduction of anorectic GLP-1R-dependent signals. In humans, the brain circuitry underlying these effects remains insufficiently investigated. The present study aimed to explore GLP-1R protein expression in the human hypothalamus and its correlation with body mass index (BMI). Sections of hypothalamus from 28 autopsy cases, 11 with normal weight (BMI < 25 kg/m2) and 17 with non-normal weight (BMI ≥ 25 kg/m2), were examined using immunohistochemistry and double immunofluorescence labeling. Prominent GLP-1R immunoexpression was detected in neurons of several hypothalamic nuclei, including paraventricular, supraoptic, and infundibular nuclei; the lateral hypothalamic area (LH); and basal forebrain nuclei. Interestingly, in the LH, GLP-1R was significantly decreased in individuals with BMI ≥ 25 kg/m2 compared with their normal weight counterparts (p = 0.03). Furthermore, GLP-1R was negatively correlated (τb = −0.347, p = 0.024) with BMI levels only in the LH. GLP-1R extensively colocalized with the anorexigenic and antiobesogenic neuropeptide nucleobindin-2/nesfatin-1 but not with the astrocytic marker glial fibrillary acidic protein. These data suggest a potential role for GLP-1R in the regulation of energy balance in the human hypothalamus. In the LH, an appetite- and reward-related brain region, reduced GLP-1R immunoexpression may contribute to the dysregulation of homeostatic and/or hedonic feeding behavior. Possible effects of NUCB2/nesfatin-1 on central GLP-1R signaling require further investigation.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Neuropeptides , Animals , Humans , Glucagon-Like Peptide-1 Receptor/metabolism , Body Mass Index , Hypothalamus/metabolism , Neuropeptides/metabolism , Arcuate Nucleus of Hypothalamus/metabolism
4.
Pathol Res Pract ; 235: 153950, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35642986

ABSTRACT

AIM: Hepatocellular carcinoma (HCC) is a common cause a cancer-related death. Focal adhesions (FAs) represent multiprotein complexes at integrin-mediated cell-extracellular matrix adhesion sites that orchestrate vital cellular functions. The heterotrimeric ILK-PINCH-PARVB (IPP) complex, RSU1, a PINCH binding protein and CTEN, a member of the tensin family of proteins exert a critical role in FAs, where they regulate important cancer related functions such as cell adhesion, migration, proliferation and survival. Previous studies implicate these FA proteins in liver pathophysiology but their detailed role in human HCC is not fully understood. Here in we investigated expression and function of IPP, RSU1 and CTEN in human HCC. METHODS: The expression of focal adhesion proteins was studied in human HCC by immunohistochemistry in relation to clinicopathological parameters, previous studied genomic instability markers and patient's survival. Effects on cell proliferation and FA proteins expression upon ILK inhibition and RSU1 silencing were also investigated in HCC in vitro. RESULTS: IPP complex and CTEN proteins are overexpressed while RSU1 expression is decreased in human HCC. CTEN expression correlates with reduced patients' survival while RSU1 represents an independent favorable prognostic indicator in human HCC. Nuclear ILK expression correlates with markers of genomic instability. Pharmacological targeting of ILK suppresses, while RSU1 silencing promotes cell growth of HCC cells in vitro, while in both experimental conditions expression and/or localization of focal adhesion proteins is deregulated. CONCLUSION: Our results suggest that FA signaling is implicated in hepatocellular carcinogenesis with prognostic significance. RSU1 seems to exert tumor suppressive functions in HCC and represents a novel favorable prognostic indicator.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Transcription Factors , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Focal Adhesions/genetics , Focal Adhesions/metabolism , Genomic Instability , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Prognosis , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Stem Cell Reports ; 17(6): 1395-1410, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35623353

ABSTRACT

Impaired replication has been previously linked to growth retardation and microcephaly; however, why the brain is critically affected compared with other organs remains elusive. Here, we report the differential response between early neural progenitors (neuroepithelial cells [NECs]) and fate-committed neural progenitors (NPs) to replication licensing defects. Our results show that, while NPs can tolerate altered expression of licensing factors, NECs undergo excessive replication stress, identified by impaired replication, increased DNA damage, and defective cell-cycle progression, leading eventually to NEC attrition and microcephaly. NECs that possess a short G1 phase license and activate more origins than NPs, by acquiring higher levels of DNA-bound MCMs. In vivo G1 shortening in NPs induces DNA damage upon impaired licensing, suggesting that G1 length correlates with replication stress hypersensitivity. Our findings propose that NECs possess distinct cell-cycle characteristics to ensure fast proliferation, although these inherent features render them susceptible to genotoxic stress.


Subject(s)
Microcephaly , Neural Stem Cells , Brain/metabolism , Cell Cycle Proteins/metabolism , DNA Damage , DNA Replication , Humans , Microcephaly/genetics , Neural Stem Cells/metabolism , Replication Origin
6.
Biol Rev Camb Philos Soc ; 96(5): 2321-2332, 2021 10.
Article in English | MEDLINE | ID: mdl-34132477

ABSTRACT

Cell differentiation is a process that must be precisely regulated for the maintenance of tissue homeostasis. Differentiation towards a multiciliated cell fate is characterized by well-defined stages, where a transcriptional cascade is activated leading to the formation of multiple centrioles and cilia. Centrioles migrate and dock to the apical cell surface and, acting as basal bodies, give rise to multiple motile cilia. The concerted movement of cilia ensures directional fluid flow across epithelia and defects either in their number or structure can lead to disease phenotypes. Micro-RNAs (miRNAs; miRs) are small, non-coding RNA molecules that play an important role in post-transcriptional regulation of gene expression. miR-34b/c and miR-449a/b/c specifically function throughout the differentiation of multiciliated cells, fine-tuning the expression of many different centriole- and cilia-related genes. They strictly regulate the expression levels of genes that are required both for commitment towards the multiciliated cell fate (e.g. Notch) and for the establishment and maintenance of this fate by regulating the expression of transcription factors and structural components of the pathway. Herein we review miR-34 and miR-449 spatiotemporal regulation along with their roles during the different stages of multiciliogenesis.


Subject(s)
Centrioles , MicroRNAs , Cell Differentiation/genetics , Cilia/genetics , MicroRNAs/genetics
7.
Dig Dis Sci ; 66(5): 1510-1523, 2021 05.
Article in English | MEDLINE | ID: mdl-32495257

ABSTRACT

BACKGROUND: Genomic instability is a hallmark of cancer cells contributing to tumor development and progression. Integrin-linked kinase (ILK) is a focal adhesion protein with well-established role in carcinogenesis. We have previously shown that ILK overexpression is critically implicated in human colorectal cancer (CRC) progression. In light of the recent findings that ILK regulates centrosomes and mitotic spindle formation, we aimed to determine its implication in mechanisms of genomic instability in human CRC. METHODS: Association of ILK expression with markers of genomic instability (micronuclei formation, nucleus size, and intensity) was investigated in diploid human colon cancer cells HCT116 upon ectopic ILK overexpression, by immunofluorescence and in human CRC samples by Feulgen staining. We also evaluated the role of ILK in mitotic spindle formation, by immunofluorescence, in HCT116 cells upon inhibition and overexpression of ILK. Finally, we evaluated association of ILK overexpression with markers of DNA damage (p-H2AX, p-ATM/ATR) in human CRC tissue samples by immunohistochemistry and in ILK-overexpressing cells by immunofluorescence. RESULTS: We showed that ILK overexpression is associated with genomic instability markers in human colon cancer cells and tissues samples. Aberrant mitotic spindles were observed in cells treated with specific ILK inhibitor (QLT0267), while ILK-overexpressing cells failed to undergo nocodazole-induced mitotic arrest. ILK overexpression was also associated with markers of DNA damage in HCT116 cells and human CRC tissue samples. CONCLUSIONS: The above findings indicate that overexpression of ILK is implicated in mechanisms of genomic instability in CRC suggesting a novel role of this protein in cancer.


Subject(s)
Colorectal Neoplasms/enzymology , DNA Damage , Genomic Instability , Micronuclei, Chromosome-Defective , Protein Serine-Threonine Kinases/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , HCT116 Cells , Histones/metabolism , Humans , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Spindle Apparatus/enzymology , Spindle Apparatus/genetics , Spindle Apparatus/pathology
8.
J Mol Histol ; 51(4): 385-400, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32592097

ABSTRACT

Integrin-linked kinase (ILK) forms a heterotrimeric protein complex with PINCH and PARVIN (IPP) in Focal Adhesions (FAs) that acts as a signaling platform between the cell and its microenvironment regulating important cancer-related functions. We aimed to elucidate the role of ILK in lung adenocarcinoma (LUADC) focusing on a possible link with KRAS oncogene. We used immunohistochemistry on human tissue samples and KRAS-driven LUADC in mice, analysis of large scale publicly available RNA sequencing data, ILK overexpression and pharmacological inhibition as well as knockdown of KRAS in lung cancer cells. ILK, PINCH1 and PARVB (IPP) proteins are overexpressed in human LUADC and KRAS-driven LUADC in mice representing poor prognostic indicators. Genes implicated in ILK signaling are significantly enriched in KRAS-driven LUADC. Silencing of KRAS, as well as, overexpression and pharmacological inhibition of ILK in lung cancer cells provide evidence of a two-way association between ILK and KRAS. Upregulation of PINCH, PARVB and Ras suppressor-1 (RSU1) expression was demonstrated in ILK overexpressing lung cancer cells in addition to a significant positive correlation between these factors in tissue samples, while KRAS silencing downregulates IPP and RSU1. Pharmacological inhibition of ILK in KRAS mutant lung cancer cells suppresses cell growth, migration, EMT and increases sensitivity to platinum-based chemotherapy. ILK promotes an aggressive lung cancer phenotype with prognostic and therapeutic value through functions that involve KRAS, IPP complex and RSU1, rendering ILK a promising biomarker and therapeutic target in lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/metabolism , Cytoskeletal Proteins/metabolism , Lung Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Transcription Factors/metabolism , A549 Cells , Adenocarcinoma of Lung/pathology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Humans , Lung Neoplasms/pathology , Mice , Signal Transduction/physiology , Tumor Microenvironment/physiology , Up-Regulation/physiology
9.
J Cell Sci ; 132(11)2019 06 03.
Article in English | MEDLINE | ID: mdl-31028178

ABSTRACT

A distinct combination of transcription factors elicits the acquisition of a specific fate and the initiation of a differentiation program. Multiciliated cells (MCCs) are a specialized type of epithelial cells that possess dozens of motile cilia on their apical surface. Defects in cilia function have been associated with ciliopathies that affect many organs, including brain and airway epithelium. Here we show that the geminin coiled-coil domain-containing protein 1 GemC1 (also known as Lynkeas) regulates the transcriptional activation of p73, a transcription factor central to multiciliogenesis. Moreover, we show that GemC1 acts in a trimeric complex with transcription factor E2F5 and tumor protein p73 (officially known as TP73), and that this complex is important for the activation of the p73 promoter. We also provide in vivo evidence that GemC1 is necessary for p73 expression in different multiciliated epithelia. We further show that GemC1 regulates multiciliogenesis through the control of chromatin organization, and the epigenetic marks/tags of p73 and Foxj1. Our results highlight novel signaling cues involved in the commitment program of MCCs across species and tissues.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Cell Cycle Proteins/metabolism , Cilia/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation/genetics , Tumor Protein p73/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Line , Chromatin/metabolism , Epithelial Cells/cytology , Forkhead Transcription Factors/metabolism , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Signal Transduction , Transcriptional Activation/genetics , Tumor Protein p73/genetics
10.
Chromosoma ; 127(2): 151-174, 2018 06.
Article in English | MEDLINE | ID: mdl-29243212

ABSTRACT

To ensure that the genetic material is accurately passed down to daughter cells during mitosis, dividing cells must duplicate their chromosomes and centrosomes once and only once per cell cycle. The same key steps-licensing, duplication, and segregation-control both the chromosome and the centrosome cycle, which must occur in concert to safeguard genome integrity. Aberrations in genome content or centrosome numbers lead to genomic instability and are linked to tumorigenesis. Such aberrations, however, can also be part of the normal life cycle of specific cell types. Multiciliated cells best exemplify the deviation from a normal centrosome cycle. They are post-mitotic cells which massively amplify their centrioles, bypassing the rule for once-per-cell-cycle centriole duplication. Hundreds of centrioles dock to the apical cell surface and generate motile cilia, whose concerted movement ensures fluid flow across epithelia. The early steps that control the generation of multiciliated cells have lately started to be elucidated. Geminin and the vertebrate-specific GemC1 and McIdas are distantly related coiled-coil proteins, initially identified as cell cycle regulators associated with the chromosome cycle. Geminin is required to ensure once-per-cell-cycle genome replication, while McIdas and GemC1 bind to Geminin and are implicated in DNA replication control. Recent findings highlight Geminin family members as early regulators of multiciliogenesis. GemC1 and McIdas specify the multiciliate cell fate by forming complexes with the E2F4/5 transcription factors to switch on a gene expression program leading to centriole amplification and cilia formation. Positive and negative interactions among Geminin family members may link cell cycle control to centriole amplification and multiciliogenesis, acting close to the point of transition from proliferation to differentiation. We review key steps of centrosome duplication and amplification, present the role of Geminin family members in the centrosome and chromosome cycle, and discuss links with disease.


Subject(s)
Centrioles/metabolism , Cilia/metabolism , Geminin/genetics , Genome , Mitosis , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrioles/ultrastructure , Cilia/ultrastructure , DNA Replication , Dwarfism/genetics , Dwarfism/metabolism , Dwarfism/pathology , E2F4 Transcription Factor/genetics , E2F4 Transcription Factor/metabolism , E2F5 Transcription Factor/genetics , E2F5 Transcription Factor/metabolism , Geminin/metabolism , Gene Expression Regulation , Genomic Instability , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Signal Transduction , Transcription Factors
11.
EMBO Rep ; 17(3): 400-13, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26882546

ABSTRACT

Multiciliated cells are terminally differentiated, post-mitotic cells that form hundreds of motile cilia on their apical surface. Defects in multiciliated cells lead to disease, including mucociliary clearance disorders that result from ciliated cell disfunction in airways. The pathway controlling multiciliogenesis, however, remains poorly characterized. We showed that GemC1, previously implicated in cell cycle control, is a central regulator of ciliogenesis. GemC1 is specifically expressed in ciliated epithelia. Ectopic expression of GemC1 is sufficient to induce early steps of multiciliogenesis in airway epithelial cells ex vivo, upregulating McIdas and FoxJ1, key transcriptional regulators of multiciliogenesis. GemC1 directly transactivates the McIdas and FoxJ1 upstream regulatory sequences, and its activity is enhanced by E2F5 and inhibited by Geminin. GemC1-knockout mice are born with airway epithelia devoid of multiciliated cells. Our results identify GemC1 as an essential regulator of ciliogenesis in the airway epithelium and a candidate gene for mucociliary disorders.


Subject(s)
Carrier Proteins/metabolism , Respiratory Mucosa/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Cilia/metabolism , E2F5 Transcription Factor/genetics , E2F5 Transcription Factor/metabolism , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Geminin/genetics , Geminin/metabolism , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Respiratory Mucosa/cytology , Up-Regulation
12.
Development ; 142(21): 3661-74, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26395491

ABSTRACT

Multiciliated cells are abundant in the epithelial surface of different tissues, including cells lining the walls of the lateral ventricles in the brain and the airway epithelium. Their main role is to control fluid flow and defects in their differentiation are implicated in many human disorders, such as hydrocephalus, accompanied by defects in adult neurogenesis and mucociliary disorder in the airway system. Here we show that Mcidas, which is mutated in human mucociliary clearance disorder, and GemC1 (Gmnc or Lynkeas), previously implicated in cell cycle progression, are key regulators of multiciliated ependymal cell generation in the mouse brain. Overexpression and knockdown experiments show that Mcidas and GemC1 are sufficient and necessary for cell fate commitment and differentiation of radial glial cells to multiciliated ependymal cells. Furthermore, we show that GemC1 and Mcidas operate in hierarchical order, upstream of Foxj1 and c-Myb transcription factors, which are known regulators of ependymal cell generation, and that Notch signaling inhibits GemC1 and Mcidas function. Our results suggest that Mcidas and GemC1 are key players in the generation of multiciliated ependymal cells of the adult neurogenic niche.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Ependyma/cytology , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Neurogenesis , Nuclear Proteins/metabolism , Animals , Carrier Proteins/genetics , Cell Cycle Proteins/genetics , Ependyma/metabolism , Forkhead Transcription Factors/metabolism , Mice , Nuclear Proteins/genetics , Proto-Oncogene Proteins c-myb/metabolism , Receptors, Notch/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
13.
Dev Comp Immunol ; 35(8): 865-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21527279

ABSTRACT

Hydrogen peroxide (H(2)O(2)) participates as a second messenger in cell signaling. In this paper, the role of H(2)O(2) was investigated, in Escherichia coli phagocytosis by the haemocytes of the medfly Ceratitis capitata. Block of H(2)O(2) synthesis by specific enzymic inhibitors, namely N-ethylmaleimide (NEM) for NADPH oxidase and diethyldithiocarbamate (DDC) for SOD, resulted in the increase of E. coli phagocytosis. Immunoblot analysis, flow cytometry and confocal microscopy, revealed the constitutive expression of SOD, in the medfly haemocytes. Phagocytosis increased by small interfering RNA (siRNA) for SOD, revealing the active involvement of SOD and H(2)O(2). Immunoblot analysis showed an increase of the ERK1/2 phosphorylation, in the presence of the above H(2)O(2) synthesis enzymic inhibitors. In addition, confocal microscopy showed no co-localization of SOD with ß integrin subunit. It appears that SOD participates in the regulation of bacterial phagocytosis, due to involvement of the produced H(2)O(2) in the differential phosphorylation of MAP kinases.


Subject(s)
Ceratitis capitata/immunology , Hemocytes/enzymology , Hydrogen Peroxide/metabolism , Phagocytosis , Superoxide Dismutase/metabolism , Animals , Cells, Cultured , Ceratitis capitata/enzymology , Ceratitis capitata/microbiology , Ditiocarb/pharmacology , Escherichia coli , Ethylmaleimide/pharmacology , Flow Cytometry , Hemocytes/cytology , Hemocytes/microbiology , Integrin beta Chains/metabolism , Mitogen-Activated Protein Kinases/metabolism , NADPH Oxidases/antagonists & inhibitors , Phosphorylation , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...