Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
2.
Circ Res ; 133(1): 25-44, 2023 06 23.
Article in English | MEDLINE | ID: mdl-37264926

ABSTRACT

BACKGROUND: ERK5 (extracellular signal-regulated kinase 5) is a dual kinase transcription factor containing an N-terminal kinase domain and a C-terminal transcriptional activation domain. Many ERK5 kinase inhibitors have been developed and tested to treat cancer and inflammatory diseases. However, recent data have raised questions about the role of the catalytic activity of ERK5 in proliferation and inflammation. We aimed to investigate how ERK5 reprograms myeloid cells to the proinflammatory senescent phenotype, subsequently leading to atherosclerosis. METHODS: A ERK5 S496A (dephosphorylation mimic) knock in (KI) mouse model was generated using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/clustered regularly interspaced short palindromic repeat-associated 9), and atherosclerosis was characterized by hypercholesterolemia induction. The plaque phenotyping in homozygous ERK5 S496A KI and wild type (WT) mice was studied using imaging mass cytometry. Bone marrow-derived macrophages were isolated from hypercholesterolemic mice and characterized using RNA sequencing and functional in vitro approaches, including senescence, mitochondria reactive oxygen species, and inflammation assays, as well as by metabolic extracellular flux analysis. RESULTS: We show that atherosclerosis was inhibited in ERK5 S496A KI mice. Furthermore, ERK5 S496 phosphorylation mediates both senescence-associated secretory phenotype and senescence-associated stemness by upregulating AHR (aryl hydrocarbon receptor) in plaque and bone marrow-derived macrophages isolated from hypercholesterolemic mice. We also discovered that ERK5 S496 phosphorylation could induce NRF2 (NFE2-related factor 2) SUMOylation at a novel K518 site to inhibit NRF2 transcriptional activity without altering ERK5 catalytic activity and mediates oxidized LDL (low-density lipoprotein)-induced senescence-associated secretory phenotype. Specific ERK5 kinase inhibitors (AX15836 and XMD8-92) also inhibited ERK5 S496 phosphorylation, suggesting the involvement of ERK5 S496 phosphorylation in the anti-inflammatory effects of these ERK5 kinase inhibitors. CONCLUSIONS: We discovered a novel mechanism by which the macrophage ERK5-NRF2 axis develops a unique senescence-associated secretory phenotype/stemness phenotype by upregulating AHR to engender atherogenesis. The finding of senescence-associated stemness phenotype provides a molecular explanation to resolve the paradox of senescence in proliferative plaque by permitting myeloid cells to escape the senescence-induced cell cycle arrest during atherosclerosis formation.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Mice , Atherosclerosis/metabolism , Inflammation , Mitogen-Activated Protein Kinase 7/genetics , Mitogen-Activated Protein Kinase 7/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism
3.
Free Radic Biol Med ; 177: 404-418, 2021 12.
Article in English | MEDLINE | ID: mdl-34619327

ABSTRACT

Focal adhesion kinase (FAK) activation plays a crucial role in vascular diseases. In endothelial cells, FAK activation is involved in the activation of pro-inflammatory signaling and the progression of atherosclerosis. Disturbed flow (D-flow) induces endothelial activation and senescence, but the exact role of FAK in D-flow-induced endothelial activation and senescence remains unclear. The objective of this study is to investigate the role of FAK SUMOylation in D-flow-induced endothelial activation and senescence. The results showed that D-flow induced reactive oxygen species (ROS) production via NADPH oxidase activation and activated a redox-sensitive kinase p90RSK, leading to FAK activation by upregulating FAK K152 SUMOylation and the subsequent Vav2 phosphorylation, which in turn formed a positive feedback loop by upregulating ROS production. This feedback loop played a crucial role in regulating endothelial activation and senescence. D-flow-induced endothelial activation and senescence were significantly inhibited by mutating a FAK SUMOylation site lysine152 to arginine. Collectively, we concluded that FAK K152 SUMOylation plays a key role in D-flow-induced endothelial activation and senescence by forming a positive feedback loop through ROS production.


Subject(s)
Endothelial Cells , Sumoylation , Endothelial Cells/metabolism , Feedback , Focal Adhesion Protein-Tyrosine Kinases , Humans , Inflammation , Phosphorylation , Reactive Oxygen Species
4.
Redox Biol ; 47: 102132, 2021 11.
Article in English | MEDLINE | ID: mdl-34619528

ABSTRACT

The incidence of cardiovascular disease (CVD) is higher in cancer survivors than in the general population. Several cancer treatments are recognized as risk factors for CVD, but specific therapies are unavailable. Many cancer treatments activate shared signaling events, which reprogram myeloid cells (MCs) towards persistent senescence-associated secretory phenotype (SASP) and consequently CVD, but the exact mechanisms remain unclear. This study aimed to provide mechanistic insights and potential treatments by investigating how chemo-radiation can induce persistent SASP. We generated ERK5 S496A knock-in mice and determined SASP in myeloid cells (MCs) by evaluating their efferocytotic ability, antioxidation-related molecule expression, telomere length, and inflammatory gene expression. Candidate SASP inducers were identified by high-throughput screening, using the ERK5 transcriptional activity reporter cell system. Various chemotherapy agents and ionizing radiation (IR) up-regulated p90RSK-mediated ERK5 S496 phosphorylation. Doxorubicin and IR caused metabolic changes with nicotinamide adenine dinucleotide depletion and ensuing mitochondrial stunning (reversible mitochondria dysfunction without showing any cell death under ATP depletion) via p90RSK-ERK5 modulation and poly (ADP-ribose) polymerase (PARP) activation, which formed a nucleus-mitochondria positive feedback loop. This feedback loop reprogramed MCs to induce a sustained SASP state, and ultimately primed MCs to be more sensitive to reactive oxygen species. This priming was also detected in circulating monocytes from cancer patients after IR. When PARP activity was transiently inhibited at the time of IR, mitochondrial stunning, priming, macrophage infiltration, and coronary atherosclerosis were all eradicated. The p90RSK-ERK5 module plays a crucial role in SASP-mediated mitochondrial stunning via regulating PARP activation. Our data show for the first time that the nucleus-mitochondria positive feedback loop formed by p90RSK-ERK5 S496 phosphorylation-mediated PARP activation plays a crucial role of persistent SASP state, and also provide preclinical evidence supporting that transient inhibition of PARP activation only at the time of radiation therapy can prevent future CVD in cancer survivors.


Subject(s)
Coronary Artery Disease , Mitogen-Activated Protein Kinase 7 , Poly(ADP-ribose) Polymerases , Adenosine Diphosphate/metabolism , Animals , Coronary Artery Disease/metabolism , Feedback , Humans , Mice , Mitochondria/metabolism , Phenotype , Phosphorylation , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Ribose/metabolism
5.
Cancer Res ; 80(12): 2484-2497, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32156775

ABSTRACT

TAp63 is a p53 family member and potent tumor and metastasis suppressor. Here, we show that TAp63-/- mice exhibit an increased susceptibility to ultraviolet radiation-induced cutaneous squamous cell carcinoma (cuSCC). A human-to-mouse comparison of cuSCC tumors identified miR-30c-2* and miR-497 as underexpressed in TAp63-deficient cuSCC. Reintroduction of these miRNAs significantly inhibited the growth of cuSCC cell lines and tumors. Proteomic profiling of cells expressing either miRNA showed downregulation of cell-cycle progression and mitosis-associated proteins. A mouse to human and cross-platform comparison of RNA-sequencing and proteomics data identified a 7-gene signature, including AURKA, KIF18B, PKMYT1, and ORC1, which were overexpressed in cuSCC. Knockdown of these factors in cuSCC cell lines suppressed tumor cell proliferation and induced apoptosis. In addition, selective inhibition of AURKA suppressed cuSCC cell proliferation, induced apoptosis, and showed antitumor effects in vivo. Finally, treatment with miR-30c-2* or miR-497 miRNA mimics was highly effective in suppressing cuSCC growth in vivo. Our data establish TAp63 as an essential regulator of novel miRNAs that can be therapeutically targeted for potent suppression of cuSCC. SIGNIFICANCE: This study provides preclinical evidence for the use of miR-30c-2*/miR-497 delivery and AURKA inhibition in the treatment of cuSCC, which currently has no FDA-approved targeted therapies.See related commentary by Parrales and Iwakuma, p. 2439.


Subject(s)
Carcinoma, Squamous Cell , MicroRNAs , Skin Neoplasms , Animals , Aurora Kinase A/genetics , Carcinoma, Squamous Cell/genetics , Cell Proliferation/genetics , Humans , Kinesins , Membrane Proteins , Mice , MicroRNAs/genetics , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Proteomics , Skin Neoplasms/genetics , Ultraviolet Rays
6.
Metabolism ; 100: 153962, 2019 11.
Article in English | MEDLINE | ID: mdl-31476350

ABSTRACT

BACKGROUND: Disturbed flow (d-flow)-induced senescence and activation of endothelial cells (ECs) have been suggested to have critical roles in promoting atherosclerosis. Telomeric repeat-binding factor 2 (TERF2)-interacting protein (TERF2IP), a member of the shelterin complex at the telomere, regulates the senescence-associated secretory phenotype (SASP), in which EC activation and senescence are engendered simultaneously by p90RSK-induced phosphorylation of TERF2IP S205 and subsequent nuclear export of the TERF2IP-TERF2 complex. In this study, we investigated TERF2IP-dependent gene expression and its role in regulating d-flow-induced SASP. METHODS: A principal component analysis and hierarchical clustering were used to identify genes whose expression is regulated by TERF2IP in ECs under d-flow conditions. Senescence was determined by reduced telomere length, increased p53 and p21 expression, and increased apoptosis; EC activation was detected by NF-κB activation and the expression of adhesion molecules. The involvement of TERF2IP S205 phosphorylation in d-flow-induced SASP was assessed by depletion of TERF2IP and mutation of the phosphorylation site. RESULTS: Our unbiased transcriptome analysis showed that TERF2IP caused alteration in the expression of a distinct set of genes, including rapamycin-insensitive companion of mTOR (RICTOR) and makorin-1 (MKRN1) ubiquitin E3 ligase, under d-flow conditions. In particular, both depletion of TERF2IP and overexpression of the TERF2IP S205A phosphorylation site mutant in ECs increased the d-flow and p90RSK-induced MKRN1 expression and subsequently inhibited apoptosis, telomere shortening, and NF-κB activation in ECs via suppression of p53, p21, and telomerase (TERT) induction. CONCLUSIONS: MKRN1 and RICTOR belong to a distinct reciprocal gene set that is both negatively and positively regulated by p90RSK. TERF2IP S205 phosphorylation, a downstream event of p90RSK activation, uniquely inhibits MKRN1 expression and contributes to EC activation and senescence, which are key events for atherogenesis.


Subject(s)
Cellular Senescence , Endothelial Cells/metabolism , Ubiquitin-Protein Ligases/metabolism , Human Umbilical Vein Endothelial Cells , Humans , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Binding , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Ribonucleoproteins/genetics , Telomeric Repeat Binding Protein 2/metabolism
7.
JCI Insight ; 4(9)2019 05 02.
Article in English | MEDLINE | ID: mdl-31045573

ABSTRACT

The interplay among signaling events for endothelial cell (EC) senescence, apoptosis, and activation and how these pathological conditions promote atherosclerosis in the area exposed to disturbed flow (d-flow) in concert remain unclear. The aim of this study was to determine whether telomeric repeat-binding factor 2-interacting protein (TERF2IP), a member of the shelterin complex at the telomere, can regulate EC senescence, apoptosis, and activation simultaneously, and if so, by what molecular mechanisms. We found that d-flow induced p90RSK and TERF2IP interaction in a p90RSK kinase activity-dependent manner. An in vitro kinase assay revealed that p90RSK directly phosphorylated TERF2IP at the serine 205 (S205) residue, and d-flow increased TERF2IP S205 phosphorylation as well as EC senescence, apoptosis, and activation by activating p90RSK. TERF2IP phosphorylation was crucial for nuclear export of the TERF2IP-TRF2 complex, which led to EC activation by cytosolic TERF2IP-mediated NF-κB activation and also to senescence and apoptosis of ECs by depleting TRF2 from the nucleus. Lastly, using EC-specific TERF2IP-knockout (TERF2IP-KO) mice, we found that the depletion of TERF2IP inhibited d-flow-induced EC senescence, apoptosis, and activation, as well as atherosclerotic plaque formation. These findings demonstrate that TERF2IP is an important molecular switch that simultaneously accelerates EC senescence, apoptosis, and activation by S205 phosphorylation.


Subject(s)
Active Transport, Cell Nucleus/physiology , Atherosclerosis/metabolism , Cellular Senescence/physiology , Endothelial Cells/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Animals , Apoptosis , Cellular Senescence/drug effects , DNA Damage , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Plaque, Atherosclerotic/metabolism , Shelterin Complex , Signal Transduction , Telomere , Telomeric Repeat Binding Protein 2/genetics , Telomeric Repeat Binding Protein 2/metabolism , Transcriptome
8.
Front Cardiovasc Med ; 5: 26, 2018.
Article in English | MEDLINE | ID: mdl-29675417

ABSTRACT

BACKGROUND: The high incidence of cardiovascular events in cancer survivors has long been noted, but the mechanistic insights of cardiovascular toxicity of cancer treatments, especially for vessel diseases, remain unclear. It is well known that atherosclerotic plaque formation begins in the area exposed to disturbed blood flow, but the relationship between cancer therapy and disturbed flow in regulating plaque formation has not been well studied. Therefore, we had two goals for this study; (1) Generate an affordable, reliable, and reproducible mouse model to recapitulate the cancer therapy-induced cardiovascular events in cancer survivors, and (2) Establish a mouse model to investigate the interplay between disturbed flow and various cancer therapies in the process of atherosclerotic plaque formation. METHODS AND RESULTS: We examined the effects of two cancer drugs and ionizing radiation (IR) on disturbed blood flow-induced plaque formation using a mouse carotid artery partial ligation (PCL) model of atherosclerosis. We found that doxorubicin and cisplatin, which are commonly used anti-cancer drugs, had no effect on plaque formation in partially ligated carotid arteries. Similarly, PCL-induced plaque formation was not affected in mice that received IR (2 Gy) and PCL surgery performed one week later. In contrast, when PCL surgery was performed 26 days after IR treatment, not only the atherosclerotic plaque formation but also the necrotic core formation was significantly enhanced. Lastly, we found a significant increase in p90RSK phosphorylation in the plaques from the IR-treated group compared to those from the non-IR treated group. CONCLUSIONS: Our results demonstrate that IR not only increases atherosclerotic events but also vulnerable plaque formation. These increases were a somewhat delayed effect of IR as they were observed in mice with PCL surgery performed 26 days, but not 10 days, after IR exposure. A proper animal model must be developed to study how to minimize the cardiovascular toxicity due to cancer treatment.

9.
PLoS One ; 11(4): e0153933, 2016.
Article in English | MEDLINE | ID: mdl-27100181

ABSTRACT

Mutational processes and signatures that drive early tumorigenesis are centrally important for early cancer prevention. Yet, to date, biomarkers and risk factors for polyps (adenomas) that inordinately and rapidly develop into colon cancer remain poorly defined. Here, we describe surprisingly high mutational profiles through whole-genome sequence (WGS) analysis in 2 of 4 pairs of benign colorectal adenoma tissue samples. Unsupervised hierarchical clustered transcriptomic analysis of a further 7 pairs of adenomas reveals distinct mutational signatures regardless of adenoma size. Transitional single nucleotide substitutions of C:G>T:A predominate in the adenoma mutational spectrum. Strikingly, we observe mutations in the TGF-ß pathway and CEA-associated genes in 4 out of 11 adenomas, overlapping with the Wnt pathway. Immunohistochemical labeling reveals a nearly 5-fold increase in CEA levels in 23% of adenoma samples with a concomitant loss of TGF-ß signaling. We also define a functional role by which the CEA B3 domain interacts with TGFBR1, potentially inactivating the tumor suppressor function of TGF-ß signaling. Our study uncovers diverse mutational processes underlying the transition from early adenoma to cancer. This has broad implications for biomarker-driven targeting of CEA/TGF-ß in high-risk adenomas and may lead to early detection of aggressive adenoma to CRC progression.


Subject(s)
Adenoma/genetics , Carcinoembryonic Antigen/genetics , Colon/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutation/genetics , Transforming Growth Factor beta/genetics , Adenoma/metabolism , Adenoma/pathology , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Blotting, Western , Carcinoembryonic Antigen/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , High-Throughput Nucleotide Sequencing/methods , Humans , Immunoenzyme Techniques , Immunoprecipitation , Signal Transduction , Transforming Growth Factor beta/metabolism
10.
J Clin Invest ; 126(2): 527-42, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26784546

ABSTRACT

Beckwith-Wiedemann syndrome (BWS) is a human stem cell disorder, and individuals with this disease have a substantially increased risk (~800-fold) of developing tumors. Epigenetic silencing of ß2-spectrin (ß2SP, encoded by SPTBN1), a SMAD adaptor for TGF-ß signaling, is causally associated with BWS; however, a role of TGF-ß deficiency in BWS-associated neoplastic transformation is unexplored. Here, we have reported that double-heterozygous Sptbn1+/- Smad3+/- mice, which have defective TGF-ß signaling, develop multiple tumors that are phenotypically similar to those of BWS patients. Moreover, tumorigenesis-associated genes IGF2 and telomerase reverse transcriptase (TERT) were overexpressed in fibroblasts from BWS patients and TGF-ß-defective mice. We further determined that chromatin insulator CCCTC-binding factor (CTCF) is TGF-ß inducible and facilitates TGF-ß-mediated repression of TERT transcription via interactions with ß2SP and SMAD3. This regulation was abrogated in TGF-ß-defective mice and BWS, resulting in TERT overexpression. Imprinting of the IGF2/H19 locus and the CDKN1C/KCNQ1 locus on chromosome 11p15.5 is mediated by CTCF, and this regulation is lost in BWS, leading to aberrant overexpression of growth-promoting genes. Therefore, we propose that loss of CTCF-dependent imprinting of tumor-promoting genes, such as IGF2 and TERT, results from a defective TGF-ß pathway and is responsible at least in part for BWS-associated tumorigenesis as well as sporadic human cancers that are frequently associated with SPTBN1 and SMAD3 mutations.


Subject(s)
Beckwith-Wiedemann Syndrome/metabolism , Carrier Proteins/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Repressor Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Beckwith-Wiedemann Syndrome/genetics , CCCTC-Binding Factor , Carrier Proteins/genetics , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 11/metabolism , Cyclin-Dependent Kinase Inhibitor p57/genetics , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Hep G2 Cells , Humans , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/metabolism , KCNQ1 Potassium Channel/genetics , KCNQ1 Potassium Channel/metabolism , Mice , Mice, Knockout , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Smad3 Protein/genetics , Smad3 Protein/metabolism , Telomerase/biosynthesis , Telomerase/genetics , Telomerase/metabolism , Transforming Growth Factor beta/genetics
11.
Genes Cancer ; 5(9-10): 348-352, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25352951

ABSTRACT

ALCOHOLIC LIVER DISEASE HAS VARIOUS MANIFESTATIONS: asymptomatic steatosis, alcoholic hepatitis and alcoholic cirrhosis, which substantially increase the risk for developing hepatocellular carcinoma. Transforming growth factor (TGF-ß) signaling pathway is a major regulator in chronic liver diseases contributing to all liver disease progression from liver injury, inflammation and fibrosis to HCC. With the aim of generating a mouse model of alcoholic liver disease that would rapidly develop steatosis, inflammation as well as fibrosis, we formulated a regimen that combined chronic injections of low dose (2mg/kg) lipopolysaccharide (LPS) with Lieber DeCarli-based diet containing 6.7% ethanol feeding to mice with impaired TGF-ß signaling through constitutive disruption of ß2-spectrin and/or Smad3. Unexpectedly, the mice treated with chronic low dose LPS and fed the alcohol-containing diet developed very aggressive T-cell lymphomas to which the TGF-ß mutant mice succumbed more rapidly than the wild type mice. In contrast, their liver phenotype was mild as they only developed steatosis but not hepatitis or significant fibrosis. To our knowledge, this is the first report of a mouse model of aggressive T- cell lymphoma based on chronic challenge with low dose LPS and TGF-ß disruption.

12.
Cell Metab ; 16(4): 511-25, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23040072

ABSTRACT

TAp63 prevents premature aging, suggesting a link to genes that regulate longevity. Further characterization of TAp63-/- mice revealed that these mice develop obesity, insulin resistance, and glucose intolerance similar to those seen in mice lacking two key metabolic regulators, Silent information regulator T1 (Sirt1) and AMPK. While the roles of Sirt1 and AMPK in metabolism have been well studied, their upstream regulators are not well understood. We found that TAp63 is important in regulating energy metabolism by accumulating in response to metabolic stress and transcriptionally activating Sirt1, AMPKα2, and LKB1, resulting in increased fatty acid synthesis and decreased fatty acid oxidation. Moreover, we found that TAp63 lowers blood glucose levels in response to metformin. Restoration of Sirt1, AMPKα2, and LKB1 in TAp63-/- mice rescued some of the metabolic defects of the TAp63-/- mice. Our study defines a role for TAp63 in metabolism and weight control.


Subject(s)
Glucose/metabolism , Lipid Metabolism/genetics , Phosphoproteins/metabolism , Trans-Activators/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Energy Metabolism , Fatty Acids/biosynthesis , Fatty Liver/metabolism , Fatty Liver/pathology , Metformin/pharmacology , Mice , Mice, Knockout , Obesity/etiology , Obesity/metabolism , Phosphoproteins/deficiency , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Sirtuin 1/genetics , Sirtuin 1/metabolism , Trans-Activators/deficiency , Trans-Activators/genetics , Transcriptional Activation/drug effects
13.
Nature ; 467(7318): 986-90, 2010 Oct 21.
Article in English | MEDLINE | ID: mdl-20962848

ABSTRACT

Aberrant expression of microRNAs (miRNAs) and the enzymes that control their processing have been reported in multiple biological processes including primary and metastatic tumours, but the mechanisms governing this are not clearly understood. Here we show that TAp63, a p53 family member, suppresses tumorigenesis and metastasis, and coordinately regulates Dicer and miR-130b to suppress metastasis. Metastatic mouse and human tumours deficient in TAp63 express Dicer at very low levels, and we found that modulation of expression of Dicer and miR-130b markedly affected the metastatic potential of cells lacking TAp63. TAp63 binds to and transactivates the Dicer promoter, demonstrating direct transcriptional regulation of Dicer by TAp63. These data provide a novel understanding of the roles of TAp63 in tumour and metastasis suppression through the coordinate transcriptional regulation of Dicer and miR-130b and may have implications for the many processes regulated by miRNAs.


Subject(s)
DEAD-box RNA Helicases/metabolism , Endoribonucleases/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Neoplasm Metastasis/genetics , Phosphoproteins/metabolism , Ribonuclease III/metabolism , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cell Line , Cell Line, Tumor , Cellular Senescence , DEAD-box RNA Helicases/biosynthesis , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Endoribonucleases/genetics , Female , Genes, Tumor Suppressor/physiology , Genomic Instability , Humans , Male , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphoproteins/deficiency , Phosphoproteins/genetics , Promoter Regions, Genetic/genetics , Ribonuclease III/biosynthesis , Ribonuclease III/deficiency , Ribonuclease III/genetics , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors , Transcriptional Activation , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics
14.
Cell Stem Cell ; 5(1): 64-75, 2009 Jul 02.
Article in English | MEDLINE | ID: mdl-19570515

ABSTRACT

The cellular mechanisms that regulate the maintenance of adult tissue stem cells are still largely unknown. We show here that the p53 family member, TAp63, is essential for maintenance of epidermal and dermal precursors and that, in its absence, these precursors senesce and skin ages prematurely. Specifically, we have developed a TAp63 conditional knockout mouse and used it to ablate TAp63 in the germline (TAp63(-/-)) or in K14-expressing cells in the basal layer of the epidermis (TAp63(fl/fl);K14cre+). TAp63(-/-) mice age prematurely and develop blisters, skin ulcerations, senescence of hair follicle-associated dermal and epidermal cells, and decreased hair morphogenesis. These phenotypes are likely due to loss of TAp63 in dermal and epidermal precursors since both cell types show defective proliferation, early senescence, and genomic instability. These data indicate that TAp63 serves to maintain adult skin stem cells by regulating cellular senescence and genomic stability, thereby preventing premature tissue aging.


Subject(s)
Adult Stem Cells/physiology , Aging, Premature/etiology , Dermis/cytology , Epidermal Cells , Phosphoproteins/genetics , Phosphoproteins/physiology , Trans-Activators/genetics , Trans-Activators/physiology , Adult Stem Cells/cytology , Aging, Premature/pathology , Animals , Cellular Senescence , DNA Damage , Genes, p53 , Genomic Instability , Hair Follicle/cytology , Keratinocytes/cytology , Mice , Mice, Knockout , Skin Aging/genetics , Skin Aging/pathology , Wound Healing/genetics
15.
EMBO J ; 28(13): 1904-15, 2009 Jul 08.
Article in English | MEDLINE | ID: mdl-19494829

ABSTRACT

Mice lacking p63 cannot form skin, exhibit craniofacial and skeletal defects, and die soon after birth. The p63 gene regulates a complex network of target genes, and disruption of p63 has been shown to affect the maintenance of epithelial stem cells, the differentiation of keratinocytes, and the preservation of the adhesive properties of stratified epithelium. Here, we show that inactivation of p63 in mice is accompanied by aberrantly increased expression of the Ink4a and Arf tumour suppressor genes. In turn, anomalies of the p63-null mouse affecting the skin and skeleton are partially ameliorated in mice lacking either Ink4a or Arf. Rescue of epithelialization is accompanied by restoration of keratinocyte proliferative capacity both in vivo and in vitro and by expression of markers of squamous differentiation. Thus, in the absence of p63, abnormal upregulation of Ink4a and Arf is incompatible with skin development.


Subject(s)
ADP-Ribosylation Factors/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Phosphoproteins/genetics , Skin/embryology , Skin/metabolism , Trans-Activators/genetics , ADP-Ribosylation Factors/metabolism , Animals , Cell Differentiation , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Gene Expression Regulation, Developmental , Keratinocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphoproteins/metabolism , Pregnancy , Skin/cytology , Trans-Activators/metabolism
16.
Int J Radiat Oncol Biol Phys ; 67(5): 1519-25, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17394948

ABSTRACT

PURPOSE: Survivin, a member of the inhibitor of apoptosis gene family, has also been shown to regulate mitosis. It binds Aurora B kinase and the inner centromere protein to form the chromosome passenger complex. Both Aurora B and survivin are overexpressed in many tumors. In this study, we examined whether irradiation affected survivin and Aurora B expression in mesothelioma cells, and how inhibition of these molecules affected radiosensitivity. METHODS AND MATERIALS: ZM447439 and survivin antisense oligonucleotides were used to inhibit survivin and Aurora B kinase respectively. Western blot was performed to determine the expression of survivin, Aurora B, phosphorylated-histone H3 (Ser 10), and caspase cleavage. Multinucleated cells were counted using flow cytometry, and cell survival after treatment was determined using clonogenic assay. RESULTS: At 3-Gy irradiation an increase was observed in levels of survivin and Aurora B as well as the kinase activity of Aurora B, with an increase in G2/M phase. The radiation-induced upregulation of these molecules was effectively attenuated by antisense oligonucleotides against survivin and a small-molecule inhibitor of Aurora B, ZM447439. Dual inhibition of survivin and Aurora B synergistically radiosensitized mesothelioma cells with a dose enhancement ratio of 2.55. This treatment resulted in increased formation of multinucleated cells after irradiation but did not increase levels of cleaved caspase 3. CONCLUSION: Inhibition of survivin and Aurora B induces mitotic cell arrest in mesothelioma cells after irradiation. These two proteins may be potential therapeutic targets for the enhancement of radiotherapy in malignant pleural mesothelioma.


Subject(s)
Histones/metabolism , Mesothelioma/radiotherapy , Microtubule-Associated Proteins/antagonists & inhibitors , Mitosis/drug effects , Neoplasm Proteins/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Aurora Kinase B , Aurora Kinases , Benzamides , Caspase 3/metabolism , Caspase 3/radiation effects , Cell Survival , G2 Phase/radiation effects , Histones/radiation effects , Humans , Inhibitor of Apoptosis Proteins , Mesothelioma/metabolism , Mesothelioma/pathology , Microtubule-Associated Proteins/metabolism , Microtubule-Associated Proteins/radiation effects , Neoplasm Proteins/metabolism , Neoplasm Proteins/radiation effects , Oligonucleotides, Antisense/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/radiation effects , Quinazolines , Radiation Tolerance/drug effects , Survivin , Up-Regulation/radiation effects
17.
Cancer Res ; 66(20): 10040-7, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-17047067

ABSTRACT

The phosphatidylinositol 3-kinase/Akt pathway plays a critical role in oncogenesis, and dysregulation of this pathway through loss of PTEN suppression is a particularly common phenomenon in aggressive prostate cancers. The mammalian target of rapamycin (mTOR) is a downstream signaling kinase in this pathway, exerting prosurvival influence on cells through the activation of factors involved in protein synthesis. The mTOR inhibitor rapamycin and its derivatives are cytotoxic to a number of cell lines. Recently, mTOR inhibition has also been shown to radiosensitize endothelial and breast cancer cells in vitro. Because radiation is an important modality in the treatment of prostate cancer, we tested the ability of the mTOR inhibitor RAD001 (everolimus) to enhance the cytotoxic effects of radiation on two prostate cancer cell lines, PC-3 and DU145. We found that both cell lines became more vulnerable to irradiation after treatment with RAD001, with the PTEN-deficient PC-3 cell line showing the greater sensitivity. This increased susceptibility to radiation is associated with induction of autophagy. Furthermore, we show that blocking apoptosis with caspase inhibition and Bax/Bak small interfering RNA in these cell lines enhances radiation-induced mortality and induces autophagy. Together, these data highlight the emerging importance of mTOR as a molecular target for therapeutic intervention, and lend support to the idea that nonapoptotic modes of cell death may play a crucial role in improving tumor cell kill.


Subject(s)
PTEN Phosphohydrolase/deficiency , Prostatic Neoplasms/radiotherapy , Protein Kinases/metabolism , Radiation-Sensitizing Agents/pharmacology , Sirolimus/analogs & derivatives , Animals , Autophagy/drug effects , Autophagy/radiation effects , Cell Line, Tumor , Everolimus , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL