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










Publication year range
1.
Res Sq ; 2023 Jul 28.
Article in English | MEDLINE | ID: mdl-37546804

ABSTRACT

While RNA secondary structures are critical to regulate alternative splicing of long-range pre-mRNA, the factors that modulate RNA structure and interfere with the recognition of the splice sites are largely unknown. Previously, we identified a small, non-coding microRNA that sufficiently affects stable stem structure formation of Nmnat pre-mRNA to regulate the outcomes of alternative splicing. However, the fundamental question remains whether such microRNA-mediated interference with RNA secondary structures is a global molecular mechanism for regulating mRNA splicing. We designed and refined a bioinformatic pipeline to predict candidate microRNAs that potentially interfere with pre-mRNA stem-loop structures, and experimentally verified splicing predictions of three different long-range pre-mRNAs in the Drosophila model system. Specifically, we observed that microRNAs can either disrupt or stabilize stem-loop structures to influence splicing outcomes. Our study suggests that MicroRNA-Mediated Obstruction of Stem-loop Alternative Splicing (MIMOSAS) is a novel regulatory mechanism for the transcriptome-wide regulation of alternative splicing, increases the repertoire of microRNA function and further indicates cellular complexity of post-transcriptional regulation.

2.
bioRxiv ; 2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37425843

ABSTRACT

While RNA secondary structures are critical to regulate alternative splicing of long-range pre-mRNA, the factors that modulate RNA structure and interfere with the recognition of the splice sites are largely unknown. Previously, we identified a small, non-coding microRNA that sufficiently affects stable stem structure formation of Nmnat pre-mRNA to regulate the outcomes of alternative splicing. However, the fundamental question remains whether such microRNA-mediated interference with RNA secondary structures is a global molecular mechanism for regulating mRNA splicing. We designed and refined a bioinformatic pipeline to predict candidate microRNAs that potentially interfere with pre-mRNA stem-loop structures, and experimentally verified splicing predictions of three different long-range pre-mRNAs in the Drosophila model system. Specifically, we observed that microRNAs can either disrupt or stabilize stem-loop structures to influence splicing outcomes. Our study suggests that MicroRNA-Mediated Obstruction of Stem-loop Alternative Splicing (MIMOSAS) is a novel regulatory mechanism for the transcriptome-wide regulation of alternative splicing, increases the repertoire of microRNA function and further indicates cellular complexity of post-transcriptional regulation. One-Sentence Summary: MicroRNA-Mediated Obstruction of Stem-loop Alternative Splicing (MIMOSAS) is a novel regulatory mechanism for the transcriptome-wide regulation of alternative splicing.

4.
Elife ; 102021 12 17.
Article in English | MEDLINE | ID: mdl-34919052

ABSTRACT

Gliomas are highly malignant brain tumors with poor prognosis and short survival. NAD+ has been shown to impact multiple processes that are dysregulated in cancer; however, anti-cancer therapies targeting NAD+ synthesis have had limited success due to insufficient mechanistic understanding. Here, we adapted a Drosophila glial neoplasia model and discovered the genetic requirement for NAD+ synthase nicotinamide mononucleotide adenylyltransferase (NMNAT) in glioma progression in vivo and in human glioma cells. Overexpressing enzymatically active NMNAT significantly promotes glial neoplasia growth and reduces animal viability. Mechanistic analysis suggests that NMNAT interferes with DNA damage-p53-caspase-3 apoptosis signaling pathway by enhancing NAD+-dependent posttranslational modifications (PTMs) poly(ADP-ribosyl)ation (PARylation) and deacetylation of p53. Since PARylation and deacetylation reduce p53 pro-apoptotic activity, modulating p53 PTMs could be a key mechanism by which NMNAT promotes glioma growth. Our findings reveal a novel tumorigenic mechanism involving protein complex formation of p53 with NAD+ synthetic enzyme NMNAT and NAD+-dependent PTM enzymes that regulates glioma growth.


One of the most common types of brain cancer, glioma, emerges when harmful mutations take place in the 'glial' cells tasked with supporting neurons. When these genetically damaged cells are not fixed or eliminated, they can go on to multiply uncontrollability. A protein known as p53 can help to repress emerging tumors by stopping mutated cells in their tracks. Glioma is a highly deadly cancer, and treatments are often ineffective. Some of these approaches have focused on a protein involved in the creation of the coenzyme NAD+, which is essential to the life processes of all cells. However, these drugs have had poor outcomes. Instead, Liu et al. focused on NMNAT, the enzyme that participates in the final stage of the creation of NAD+. NMNAT is known to protect neurons, but it is unclear how it involved in cancer. Experiments in fruit flies which were then validated in human glioma cells showed that increased NMNAT activity allowed glial cells with harmful mutations to survive and multiply. Detailed molecular analysis showed that NMNAT orchestrates chemical modifications that inactivate p53. It does so by working with other molecular actors to direct NAD+ to add and remove chemical groups that control the activity of p53. Taken together, these results show how NMNAT can participate in the emergence of brain cancers. They also highlight the need for further research on whether drugs that inhibit this enzyme could help to suppress tumors before they become deadly.


Subject(s)
Cell Proliferation , Drosophila Proteins/genetics , Glioma/metabolism , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Protein Processing, Post-Translational , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Disease Models, Animal , Drosophila Proteins/metabolism , Glioma/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism
5.
Nat Neurosci ; 24(12): 1673-1685, 2021 12.
Article in English | MEDLINE | ID: mdl-34782793

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a devastating disorder in which motor neurons degenerate, the causes of which remain unclear. In particular, the basis for selective vulnerability of spinal motor neurons (sMNs) and resistance of ocular motor neurons to degeneration in ALS has yet to be elucidated. Here, we applied comparative multi-omics analysis of human induced pluripotent stem cell-derived sMNs and ocular motor neurons to identify shared metabolic perturbations in inherited and sporadic ALS sMNs, revealing dysregulation in lipid metabolism and its related genes. Targeted metabolomics studies confirmed such findings in sMNs of 17 ALS (SOD1, C9ORF72, TDP43 (TARDBP) and sporadic) human induced pluripotent stem cell lines, identifying elevated levels of arachidonic acid. Pharmacological reduction of arachidonic acid levels was sufficient to reverse ALS-related phenotypes in both human sMNs and in vivo in Drosophila and SOD1G93A mouse models. Collectively, these findings pinpoint a catalytic step of lipid metabolism as a potential therapeutic target for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/metabolism , Lipid Metabolism/genetics , Mice , Mice, Transgenic , Motor Neurons/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics
6.
Cell ; 184(3): 689-708.e20, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33482083

ABSTRACT

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.


Subject(s)
C9orf72 Protein/metabolism , DNA Repeat Expansion/genetics , Nerve Degeneration/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Axons/metabolism , C9orf72 Protein/genetics , Cell Death , Cells, Cultured , Cerebral Cortex/pathology , Chromatin/metabolism , DNA Damage , Disease Models, Animal , Drosophila , Mice, Inbred C57BL , Nerve Degeneration/pathology , Protein Stability , Transcription, Genetic , Tumor Suppressor Proteins/metabolism
7.
Elife ; 92020 12 10.
Article in English | MEDLINE | ID: mdl-33300868

ABSTRACT

Disrupted nucleocytoplasmic transport (NCT) has been implicated in neurodegenerative disease pathogenesis; however, the mechanisms by which disrupted NCT causes neurodegeneration remain unclear. In a Drosophila screen, we identified ref(2)P/p62, a key regulator of autophagy, as a potent suppressor of neurodegeneration caused by the GGGGCC hexanucleotide repeat expansion (G4C2 HRE) in C9orf72 that causes amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). We found that p62 is increased and forms ubiquitinated aggregates due to decreased autophagic cargo degradation. Immunofluorescence and electron microscopy of Drosophila tissues demonstrate an accumulation of lysosome-like organelles that precedes neurodegeneration. These phenotypes are partially caused by cytoplasmic mislocalization of Mitf/TFEB, a key transcriptional regulator of autophagolysosomal function. Additionally, TFEB is mislocalized and downregulated in human cells expressing GGGGCC repeats and in C9-ALS patient motor cortex. Our data suggest that the C9orf72-HRE impairs Mitf/TFEB nuclear import, thereby disrupting autophagy and exacerbating proteostasis defects in C9-ALS/FTD.


Subject(s)
Active Transport, Cell Nucleus/genetics , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/physiology , Microphthalmia-Associated Transcription Factor/physiology , Amyotrophic Lateral Sclerosis/genetics , Animals , Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Blotting, Western , C9orf72 Protein/genetics , Disease Models, Animal , Drosophila melanogaster , Female , Fluorescent Antibody Technique , Frontotemporal Dementia/genetics , HeLa Cells , Humans , Lysosomes/genetics , Male , Microphthalmia-Associated Transcription Factor/metabolism , Microscopy, Electron, Transmission , Motor Cortex/metabolism
8.
J Nutr Biochem ; 64: 170-181, 2019 02.
Article in English | MEDLINE | ID: mdl-30530257

ABSTRACT

The intestinal microbiota actively converts dietary flavanols into phenolic acids, some of which are bioavailable in vivo and may promote resilience to select neurological disorders by interfering with key pathologic mechanisms. Since every person harbors a unique set of gut bacteria, we investigated the influence of the gut microbiota's interpersonal heterogeneity on the production and bioavailability of flavonoid metabolites that may interfere with the misfolding of alpha (α)-synuclein, a process that plays a central role in Parkinson's disease and other α-synucleinopathies. We generated two experimental groups of humanized gnotobiotic mice with compositionally diverse gut bacteria and orally treated the mice with a flavanol-rich preparation (FRP). The two gnotobiotic mouse groups exhibited distinct differences in the generation and bioavailability of FRP-derived microbial phenolic acid metabolites that have bioactivity towards interfering with α-synuclein misfolding or inflammation. We also demonstrated that these bioactive phenolic acids are effective in modulating the development and progression of motor dysfunction in a Drosophila model of α-synucleinopathy. Lastly, through in vitro bacterial fermentation studies, we identified select bacteria that are capable of supporting the generation of these bioavailable and bioactive phenolic acids. Outcomes from our studies provide a better understanding of how interpersonal heterogeneity in the gut microbiota differentially modulates the efficacy of dietary flavanols to protect against select pathologic mechanisms. Collectively, our findings provide the basis for future developments of probiotic, prebiotic, or synbiotic approaches for modulating the onset and/or progression of α-synucleinopathies and other neurological disorders involving protein misfolding and/or inflammation.


Subject(s)
Gastrointestinal Microbiome/physiology , Polyphenols/pharmacokinetics , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , alpha-Synuclein/toxicity , Animals , Animals, Genetically Modified , Biological Availability , Brain/drug effects , Brain/metabolism , Brain/pathology , Disease Models, Animal , Drosophila , Female , Humans , Male , Mice, Inbred C57BL , Parkinson Disease/metabolism , Parkinson Disease/pathology , Polyphenols/metabolism , Protein Aggregation, Pathological/metabolism , Protein Folding , Specific Pathogen-Free Organisms , Synucleinopathies/pathology , alpha-Synuclein/chemistry , alpha-Synuclein/genetics
9.
Cell ; 173(4): 958-971.e17, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29628143

ABSTRACT

Defects in nucleocytoplasmic transport have been identified as a key pathogenic event in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) mediated by a GGGGCC hexanucleotide repeat expansion in C9ORF72, the most common genetic cause of ALS/FTD. Furthermore, nucleocytoplasmic transport disruption has also been implicated in other neurodegenerative diseases with protein aggregation, suggesting a shared mechanism by which protein stress disrupts nucleocytoplasmic transport. Here, we show that cellular stress disrupts nucleocytoplasmic transport by localizing critical nucleocytoplasmic transport factors into stress granules, RNA/protein complexes that play a crucial role in ALS pathogenesis. Importantly, inhibiting stress granule assembly, such as by knocking down Ataxin-2, suppresses nucleocytoplasmic transport defects as well as neurodegeneration in C9ORF72-mediated ALS/FTD. Our findings identify a link between stress granule assembly and nucleocytoplasmic transport, two fundamental cellular processes implicated in the pathogenesis of C9ORF72-mediated ALS/FTD and other neurodegenerative diseases.


Subject(s)
Active Transport, Cell Nucleus/physiology , Amyotrophic Lateral Sclerosis/pathology , Ataxin-2/metabolism , C9orf72 Protein/genetics , Frontotemporal Dementia/pathology , Active Transport, Cell Nucleus/drug effects , Aged , Amyotrophic Lateral Sclerosis/metabolism , Arsenites/toxicity , Ataxin-2/antagonists & inhibitors , Ataxin-2/genetics , C9orf72 Protein/metabolism , DNA Repeat Expansion/genetics , Female , Frontotemporal Dementia/metabolism , HEK293 Cells , Humans , Male , Membrane Glycoproteins/metabolism , Middle Aged , Nuclear Pore Complex Proteins/metabolism , Oxidative Stress/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Sodium Compounds/toxicity , alpha Karyopherins/antagonists & inhibitors , alpha Karyopherins/genetics , alpha Karyopherins/metabolism , beta Karyopherins/antagonists & inhibitors , beta Karyopherins/genetics , beta Karyopherins/metabolism , ran GTP-Binding Protein/antagonists & inhibitors , ran GTP-Binding Protein/genetics , ran GTP-Binding Protein/metabolism
10.
Microb Cell ; 3(8): 338-351, 2016 Jul 26.
Article in English | MEDLINE | ID: mdl-28357370

ABSTRACT

Defects in mitochondrial biogenesis and function are common in many neurodegenerative disorders, including Huntington's disease (HD). We have previously shown that in yeast models of HD, enhancement of mitochondrial biogenesis through overexpression of Hap4, the catalytic subunit of the transcriptional complex that regulates mitochondrial gene expression, alleviates the growth arrest induced by expanded polyglutamine (polyQ) tract peptides in rapidly dividing cells. However, the mechanism through which HAP4 overexpression exerts this protection remains unclear. Furthermore, it remains unexplored whether HAP4 overexpression and increased respiratory function during growth can also protect against polyQ-induced toxicity during yeast chronological lifespan. Here, we show that in yeast, mitochondrial respiration and oxidative phosphorylation (OXPHOS) are essential for protection against the polyQ-induced growth defect by HAP4 overexpression. In addition, we show that not only increased HAP4 levels, but also alternative interventions, including calorie restriction, that result in enhanced mitochondrial biogenesis confer protection against polyQ toxicity during stationary phase. The data obtained in yeast models guided experiments in a fly model of HD, where we show that enhancement of mitochondrial biogenesis can also protect against neurodegeneration and behavioral deficits. Our results suggest that therapeutic interventions aiming at the enhancement of mitochondrial respiration and OXPHOS could reduce polyQ toxicity and delay disease onset.

11.
Nat Commun ; 6: 10057, 2015 Nov 30.
Article in English | MEDLINE | ID: mdl-26616331

ABSTRACT

Nicotinamide mononucleotide adenylyltransferase (NMNAT) is a conserved enzyme in the NAD synthetic pathway. It has also been identified as an effective and versatile neuroprotective factor. However, it remains unclear how healthy neurons regulate the dual functions of NMNAT and achieve self-protection under stress. Here we show that Drosophila Nmnat (DmNmnat) is alternatively spliced into two mRNA variants, RA and RB, which translate to protein isoforms with divergent neuroprotective capacities against spinocerebellar ataxia 1-induced neurodegeneration. Isoform PA/PC translated from RA is nuclear-localized with minimal neuroprotective ability, and isoform PB/PD translated from RB is cytoplasmic and has robust neuroprotective capacity. Under stress, RB is preferably spliced in neurons to produce the neuroprotective PB/PD isoforms. Our results indicate that alternative splicing functions as a switch that regulates the expression of functionally distinct DmNmnat variants. Neurons respond to stress by driving the splicing switch to produce the neuroprotective variant and therefore achieve self-protection.


Subject(s)
Alternative Splicing , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/enzymology , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Animals , Drosophila/genetics , Drosophila/physiology , Female , Isoenzymes/genetics , Isoenzymes/metabolism , Male , Neurons/metabolism , Neuroprotection , Stress, Physiological
12.
EMBO Rep ; 14(1): 87-94, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23154466

ABSTRACT

Active zones are specialized presynaptic structures critical for neurotransmission. We show that a neuronal maintenance factor, nicotinamide mononucleotide adenylyltransferase (NMNAT), is required for maintaining active zone structural integrity in Drosophila by interacting with the active zone protein, Bruchpilot (BRP), and shielding it from activity-induced ubiquitin-proteasome-mediated degradation. NMNAT localizes to the peri-active zone and interacts biochemically with BRP in an activity-dependent manner. Loss of NMNAT results in ubiquitination, mislocalization and aggregation of BRP, and subsequent active zone degeneration. We propose that, as a neuronal maintenance factor, NMNAT specifically maintains active zone structure by direct protein-protein interaction.


Subject(s)
Drosophila Proteins/genetics , Drosophila/genetics , Nicotinamide-Nucleotide Adenylyltransferase/genetics , Presynaptic Terminals/enzymology , Animals , Drosophila/enzymology , Drosophila Proteins/metabolism , Gene Expression Regulation , Nicotinamide-Nucleotide Adenylyltransferase/antagonists & inhibitors , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Presynaptic Terminals/ultrastructure , Proteasome Endopeptidase Complex , Protein Binding , Proteolysis , RNA, Small Interfering/genetics , Signal Transduction , Synaptic Transmission/physiology , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitination
13.
Hum Mol Genet ; 21(2): 237-50, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-21965302

ABSTRACT

Tauopathies, including Alzheimer's disease, are a group of neurodegenerative diseases characterized by abnormal tau hyperphosphorylation that leads to formation of neurofibrillary tangles. Drosophila models of tauopathy display prominent features of the human disease including compromised lifespan, impairments of learning, memory and locomotor functions and age-dependent neurodegeneration visible as vacuolization. Here, we use a Drosophila model of frontotemporal dementia with parkinsonism linked to chromosome 17 (FTDP-17), in order to study the neuroprotective capacity of a recently identified neuronal maintenance factor, nicotinamide mononucleotide (NAD) adenylyl transferase (NMNAT), a protein that has both NAD synthase and chaperone function. NMNAT is essential for maintaining neuronal integrity under normal conditions and has been shown to protect against several neurodegenerative conditions. However, its protective role in tauopathy has not been examined. Here, we show that overexpression of NMNAT significantly suppresses both behavioral and morphological deficits associated with tauopathy by means of reducing the levels of hyperphosphorylated tau oligomers. Importantly, the protective activity of NMNAT protein is independent of its NAD synthesis activity, indicating a role for direct protein-protein interaction. Next, we show that NMNAT interacts with phosphorylated tau in vivo and promotes the ubiquitination and clearance of toxic tau species. Consequently, apoptosis activation was significantly reduced in brains overexpressing NMNAT, and neurodegeneration was suppressed. Our report on the molecular basis of NMNAT-mediated neuroprotection in tauopathies opens future investigation of this factor in other protein foldopathies.


Subject(s)
Disease Models, Animal , Nicotinamide-Nucleotide Adenylyltransferase/metabolism , Tauopathies/metabolism , tau Proteins/antagonists & inhibitors , Animals , Drosophila , Phosphorylation , Tauopathies/enzymology , Tauopathies/pathology , tau Proteins/metabolism
14.
J Vis Exp ; (49)2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21445036

ABSTRACT

Advances in genetic methods have enabled the study of genes involved in human neurodegenerative diseases using Drosophila as a model system. Most of these diseases, including Alzheimer's, Parkinson's and Huntington's disease are characterized by age-dependent deterioration in learning and memory functions and movement coordination. Here we use behavioral assays, including the negative geotaxis assay and the aversive phototaxic suppression assay (APS assay), to show that some of the behavior characteristics associated with human neurodegeneration can be recapitulated in flies. In the negative geotaxis assay, the natural tendency of flies to move against gravity when agitated is utilized to study genes or conditions that may hinder locomotor capacities. In the APS assay, the learning and memory functions are tested in positively-phototactic flies trained to associate light with aversive bitter taste and hence avoid this otherwise natural tendency to move toward light. Testing these trained flies 6 hours post-training is used to assess memory functions. Using these assays, the contribution of any genetic or environmental factors toward developing neurodegeneration can be easily studied in flies.


Subject(s)
Disease Models, Animal , Drosophila/physiology , Locomotion/physiology , Memory/physiology , Neurodegenerative Diseases/physiopathology , Animals , Humans
15.
J Biomed Opt ; 15(4): 048002, 2010.
Article in English | MEDLINE | ID: mdl-20799845

ABSTRACT

To overcome the inconsecutive drawback of shadow and schlieren photography, the complete dynamics of cavitation bubble oscillation or ablation products induced by a single holmium laser pulse [2.12 microm, 300 micros (FWHM)] transmitted in different core diameter (200, 400, and 600 microm) fibers is recorded by means of high-speed photography. Consecutive images from high-speed cameras can stand for the true and complete process of laser-water or laser-tissue interaction. Both laser pulse energy and fiber diameter determine cavitation bubble size, which further determines acoustic transient amplitudes. Based on the pictures taken by high-speed camera and scanned by an optical coherent microscopy (OCM) system, it is easily seen that the liquid layer at the distal end of the fiber plays an important role during the process of laser-tissue interaction, which can increase ablation efficiency, decrease heat side effects, and reduce cost.


Subject(s)
Bone and Bones/pathology , Bone and Bones/surgery , Immersion , Laser Therapy/instrumentation , Laser Therapy/methods , Lasers, Solid-State , Animals , Humans
16.
Cell Biol Int ; 33(12): 1237-44, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19732843

ABSTRACT

Genistein is a major isoflavonoid in dietary soybean, commonly consumed in Asia. Genistein exerts inhibitory effects on the proliferation of various cancer cells and plays an important role in cancer prevention. However, the molecular and cellular mechanisms of genistein on human ovarian cancer cells are still little known. We show that exposure of human ovarian cancer HO-8910 cells to genistein induces DNA damage, and triggers G2/M phase arrest and apoptosis. Furthermore, we also found that checkpoint proteins ATM and ATR are phosphorylated and activated in the cells treated with genistein. It is also shown that genistein increases the phosphorylation and activation of Chk1 and Chk2, which results in the phosphorylation and inactivation of phosphatases Cdc25C and Cdc25A, and thereby the phosphorylation and inactivation of Cdc2 which arrests cells in G2/M phase. Moreover, genistein enhances the phosphorylation and activation of p53, while decreases the ratio of Bcl-2/Bax and Bcl-xL/Bax and the level of phosphorylated Akt, which result in cells undergoing apoptosis. These results demonstrate that genistein-activated ATM-Chk2-Cdc25 and ATR-Chk1-Cdc25 DNA damage checkpoint pathways can arrest ovarian cancer cells in G2/M phase, and induce apoptosis while the cellular DNA damage is too serious to be repaired. Thus, the antiproliferative, DNA damage-inducing and pro-apoptotic activities of genistein are probably responsible for its genotoxic effects on human ovarian cancer HO-8910 cells.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , DNA Damage/drug effects , Genistein/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Signal Transduction/drug effects , Apoptosis/genetics , Ataxia Telangiectasia Mutated Proteins , BRCA1 Protein/metabolism , CDC2 Protein Kinase , Cell Cycle/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Checkpoint Kinase 1 , Checkpoint Kinase 2 , Cyclin B/metabolism , Cyclin-Dependent Kinases , DNA-Binding Proteins/metabolism , Female , Humans , Ovarian Neoplasms/metabolism , Phosphorylation , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Tumor Suppressor Proteins/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , cdc25 Phosphatases/metabolism
17.
J Cell Biochem ; 107(6): 1053-62, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19479945

ABSTRACT

Hypoxia has been recognized as one of the fundamentally important features of solid tumors and plays a critical role in various cellular and physiologic events, including cell proliferation, survival, angiogenesis, immunosurveillance, metabolism, as well as tumor invasion and metastasis. These responses to hypoxia are at least partially orchestrated by activation of the hypoxia-inducible factors (HIFs). HIF-1 is a key regulator of the response of mammalian cells to oxygen deprivation and plays critical roles in the adaptation of tumor cells to a hypoxic microenvironment. Hypoxia and overexpression of HIF-1 have been associated with radiation therapy and chemotherapy resistance, an increased risk of invasion and metastasis, and a poor clinical prognosis of solid tumors. The discovery of HIF-1 signaling has led to a rapidly increasing understanding of the complex mechanisms involved in tumor hypoxia and has helped greatly in screening novel anticancer agents. In this review, we will first introduce the cellular responses to hypoxia and HIF-1 signaling pathway in hypoxia, and then summarize the multifaceted role of hypoxia in the hallmarks of human cancers.


Subject(s)
Hypoxia/pathology , Neoplasms/pathology , Humans , Hypoxia/metabolism , Hypoxia-Inducible Factor 1/metabolism , Neoplasms/metabolism , Signal Transduction
18.
Cancer Lett ; 285(2): 116-26, 2009 Nov 28.
Article in English | MEDLINE | ID: mdl-19464788

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs of 18-25 nucleotides in length that function as negative regulators. miRNAs post-transcriptionally regulate gene expression by either inhibiting mRNA translation or inducing mRNA degradation, and participate in a wide variety of physiological and pathological cellular processes. Recent reports have revealed that the deregulation of miRNAs correlates with various human cancers and is involved in the initiation and progression of human cancers. miRNAs can act as oncogenes or tumor suppressors to inhibit the expression of cancer-related target genes and to promote or suppress tumorigenesis in various tissues. Therefore, abnormal miRNA expression can be regarded as a common feature of human cancers, and the identification of miRNAs and their respective targets may provide potential diagnostic and prognostic tumor biomarkers and new therapeutic strategies to treat cancers. In the present review, we discuss the emerging roles of miRNAs in the hallmarks of human cancers.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Neoplasms/genetics , Humans
19.
Cancer Lett ; 281(2): 213-9, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19328625

ABSTRACT

Periostin is a secreted protein and has been shown to be frequently overexpressed in various types of human cancers. We have previously reported that periostin potently promotes metastatic growth of colon cancer by augmenting cell survival. However, little is known about the functions of periostin in non-small-cell lung cancer. Here, we revealed that increased expression of periostin in non-small-cell lung cancer A549 cells was one kind of cellular responses to the stress of chemical-mimic hypoxia, and this effect could be regulated by hypoxia inducible growth factors, such as TGF-alpha and bFGF. We further demonstrated that RTK/PI3-K pathway activated by TGF-alpha and bFGF was evoked in upregulating the expression of periostin, and then periostin promoted the survival of A549 cells under hypoxic microenvironment via activation of Akt/PKB pathway. Therefore, periostin and the pathway that it involved might provide a target for lung cancer treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Adhesion Molecules/metabolism , Cell Hypoxia/physiology , Lung Neoplasms/metabolism , Apoptosis/physiology , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , Fibroblast Growth Factor 2/metabolism , Humans , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Transfection , Transforming Growth Factor alpha/metabolism , Up-Regulation
20.
Cell Mol Life Sci ; 66(14): 2219-30, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19308325

ABSTRACT

Periostin, also called osteoblast-specific factor 2 (OSF-2), is a member of the fasciclin family and a disulfide-linked cell adhesion protein that has been shown to be expressed preferentially in the periosteum and periodontal ligaments, where it acts as a critical regulator of bone and tooth formation and maintenance. Furthermore, periostin plays an important role in cardiac development. Recent clinical evidence has also revealed that periostin is involved in the development of various tumors, such as breast, lung, colon, pancreatic, and ovarian cancers. Periostin interacts with multiple cell-surface receptors, most notably integrins, and signals mainly via the PI3-K/Akt and other pathways to promote cancer cell survival, epithelial-mesenchymal transition (EMT), invasion, and metastasis. In this review, aspects related to the function of periostin in tumorigenesis are summarized.


Subject(s)
Cell Adhesion Molecules/physiology , Neoplasms/classification , Neoplasms/metabolism , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cell Survival , Humans , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...