Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 119(49): e2213146119, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36449545

ABSTRACT

Activation of endogenous retrotransposons frequently occurs in cancer cells and contributes to tumor genomic instability. To test whether inhibition of retrotranspositions has an anticancer effect, we used treatment with the nucleoside reverse transcriptase inhibitor (NRTI) stavudine (STV) in mouse cancer models, MMTV-HER2/Neu and Th-MYCN, that spontaneously develop breast cancer and neuroblastoma, respectively. In both cases, STV in drinking water did not affect tumor incidence nor demonstrate direct antitumor effects. However, STV dramatically extended progression-free survival in both models following an initial complete response to chemotherapy. To approach the mechanism underlying this phenomenon, we analyzed the effect of NRTI on the selection of treatment-resistant variants in tumor cells in culture. Cultivation of mouse breast carcinoma 4T1 in the presence of STV dramatically reduced the frequency of cells capable of surviving treatment with anticancer drugs. Global transcriptome analysis demonstrated that the acquisition of drug resistance by 4T1 cells was accompanied by an increase in the constitutive activity of interferon type I and NF-κB pathways and an elevated expression of LINE-1 elements, which are known to induce inflammatory responses via their products of reverse transcription. Treatment with NRTI reduced NF-κB activity and reverted drug resistance. Furthermore, the inducible expression of LINE-1 stimulated inflammatory response and increased the frequency of drug-resistant variants in a tumor cell population. These results indicate a mechanism by which retrotransposon desilencing can stimulate tumor cell survival during treatment and suggest reverse transcriptase inhibition as a potential therapeutic approach for targeting the development of drug-resistant cancers.


Subject(s)
Retroelements , Reverse Transcriptase Inhibitors , Animals , Mice , Reverse Transcriptase Inhibitors/pharmacology , Retroelements/genetics , NF-kappa B , Drug Resistance, Neoplasm/genetics , Long Interspersed Nucleotide Elements
2.
Nucleic Acids Res ; 49(19): 11350-11366, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34554261

ABSTRACT

Long interspersed nuclear element-1 (L1) is an autonomous non-LTR retrotransposon comprising ∼20% of the human genome. L1 self-propagation causes genomic instability and is strongly associated with aging, cancer and other diseases. The endonuclease domain of L1's ORFp2 protein (L1-EN) initiates de novo L1 integration by nicking the consensus sequence 5'-TTTTT/AA-3'. In contrast, related nucleases including structurally conserved apurinic/apyrimidinic endonuclease 1 (APE1) are non-sequence specific. To investigate mechanisms underlying sequence recognition and catalysis by L1-EN, we solved crystal structures of L1-EN complexed with DNA substrates. This showed that conformational properties of the preferred sequence drive L1-EN's sequence-specificity and catalysis. Unlike APE1, L1-EN does not bend the DNA helix, but rather causes 'compression' near the cleavage site. This provides multiple advantages for L1-EN's role in retrotransposition including facilitating use of the nicked poly-T DNA strand as a primer for reverse transcription. We also observed two alternative conformations of the scissile bond phosphate, which allowed us to model distinct conformations for a nucleophilic attack and a transition state that are likely applicable to the entire family of nucleases. This work adds to our mechanistic understanding of L1-EN and related nucleases and should facilitate development of L1-EN inhibitors as potential anticancer and antiaging therapeutics.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA/chemistry , Deoxyribonuclease I/chemistry , Base Sequence , Binding Sites , Cloning, Molecular , Crystallography, X-Ray , DNA/genetics , DNA/metabolism , DNA Cleavage , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Deoxyribonuclease I/genetics , Deoxyribonuclease I/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Genome, Human , Genomic Instability , Humans , Models, Molecular , Nucleic Acid Conformation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Substrate Specificity , Thermodynamics
3.
Proc Natl Acad Sci U S A ; 116(7): 2603-2611, 2019 02 12.
Article in English | MEDLINE | ID: mdl-30683717

ABSTRACT

The activation of cellular senescence throughout the lifespan promotes tumor suppression, whereas the persistence of senescent cells contributes to aspects of aging. This theory has been limited, however, by an inability to identify and isolate individual senescent cells within an intact organism. Toward that end, we generated a murine reporter strain by "knocking-in" a fluorochrome, tandem-dimer Tomato (tdTom), into exon 1α of the p16INK4a locus. We used this allele (p16tdTom ) for the enumeration, isolation, and characterization of individual p16INK4a -expressing cells (tdTom+). The half-life of the knocked-in transcript was shorter than that of the endogenous p16INK4a mRNA, and therefore reporter expression better correlated with p16INK4a promoter activation than p16INK4a transcript abundance. The frequency of tdTom+ cells increased with serial passage in cultured murine embryo fibroblasts from p16tdTom/+ mice. In adult mice, tdTom+ cells could be readily detected at low frequency in many tissues, and the frequency of these cells increased with aging. Using an in vivo model of peritoneal inflammation, we compared the phenotype of cells with or without activation of p16INK4a and found that tdTom+ macrophages exhibited some features of senescence, including reduced proliferation, senescence-associated ß-galactosidase (SA-ß-gal) activation, and increased mRNA expression of a subset of transcripts encoding factors involved in SA-secretory phenotype (SASP). These results indicate that cells harboring activation of the p16INK4a promoter accumulate with aging and inflammation in vivo, and display characteristics of senescence.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Promoter Regions, Genetic , Animals , Cell Proliferation , Enzyme Activation , Fibroblasts/metabolism , Half-Life , Humans , Mice , Phenotype , RNA, Messenger/genetics , beta-Galactosidase/metabolism
4.
Cancer Immunol Immunother ; 70(7): 2073-2086, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33439292

ABSTRACT

Curaxins are small molecules that bind genomic DNA and interfere with DNA-histone interactions leading to the loss of histones and decondensation of chromatin. We named this phenomenon 'chromatin damage'. Curaxins demonstrated anti-cancer activity in multiple pre-clinical tumor models. Here, we present data which reveals, for the first time, a role for the immune system in the anti-cancer effects of curaxins. Using the lead curaxin, CBL0137, we observed elevated expression of several group of genes in CBL0137-treated tumor cells including interferon sensitive genes, MHC molecules, some embryo-specific antigens suggesting that CBL0137 increases tumor cell immunogenicity and improves recognition of tumor cells by the immune system. In support of this, we found that the anti-tumor activity of CBL0137 was reduced in immune deficient SCID mice when compared to immune competent mice. Anti-tumor activity of CBL0137 was abrogated in CD8+ T cell depleted mice but only partially lost when natural killer or CD4+ T cells were depleted. Further support for a key role for the immune system in the anti-tumor activity of CBL0137 is evidenced by an increased antigen-specific effector CD8+ T cell and NK cell response, and an increased ratio of effector T cells to Tregs in the tumor and spleen. CBL0137 also elevated the number of CXCR3-expressing CTLs in the tumor and the level of interferon-γ-inducible protein 10 (IP-10) in serum, suggesting IP-10/CXCR3 controls CBL0137-elicited recruitment of effector CTLs to tumors. Our collective data underscores a previously unrecognized role for both innate and adaptive immunity in the anti-tumor activity of curaxins.


Subject(s)
Carbazoles/pharmacology , Chromatin/drug effects , Colonic Neoplasms/drug therapy , Immunity/immunology , Animals , Apoptosis , Cell Proliferation , Chemokines/metabolism , Chromatin/genetics , Chromatin/metabolism , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Cytokines/metabolism , Female , Humans , Mice , Mice, Inbred BALB C , Mice, SCID , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Proc Natl Acad Sci U S A ; 114(9): E1668-E1677, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28193858

ABSTRACT

Studying the phenomenon of cellular senescence has been hindered by the lack of senescence-specific markers. As such, detection of proteins informally associated with senescence accompanies the use of senescence-associated ß-galactosidase as a collection of semiselective markers to monitor the presence of senescent cells. To identify novel biomarkers of senescence, we immunized BALB/c mice with senescent mouse lung fibroblasts and screened for antibodies that recognized senescence-associated cell-surface antigens by FACS analysis and a newly developed cell-based ELISA. The majority of antibodies that we isolated, cloned, and sequenced belonged to the IgM isotype of the innate immune system. In-depth characterization of one of these monoclonal, polyreactive natural antibodies, the IgM clone 9H4, revealed its ability to recognize the intermediate filament vimentin. By using 9H4, we observed that senescent primary human fibroblasts express vimentin on their cell surface, and MS analysis revealed a posttranslational modification on cysteine 328 (C328) by the oxidative adduct malondialdehyde (MDA). Moreover, elevated levels of secreted MDA-modified vimentin were detected in the plasma of aged senescence-accelerated mouse prone 8 mice, which are known to have deregulated reactive oxygen species metabolism and accelerated aging. Based on these findings, we hypothesize that humoral innate immunity may recognize senescent cells by the presence of membrane-bound MDA-vimentin, presumably as part of a senescence eradication mechanism that may become impaired with age and result in senescent cell accumulation.


Subject(s)
Antibodies/metabolism , Cell Membrane/metabolism , Cellular Senescence/physiology , Vimentin/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Female , Fibroblasts/metabolism , Immunity, Humoral/physiology , Immunity, Innate/physiology , Immunoglobulin M/metabolism , Intermediate Filaments/metabolism , Malondialdehyde/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , beta-Galactosidase/metabolism
6.
Proc Natl Acad Sci U S A ; 113(7): E874-83, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26831100

ABSTRACT

Activation of an anticancer innate immune response is highly desirable because of its inherent ability to generate an adaptive antitumor T-cell response. However, insufficient safety of innate immune modulators limits clinical use to topical applications. Toll-like receptor 5 (TLR5) agonists are favorably positioned as potential systemic immunotherapeutic agents because of unusual tissue specificity of expression, uniquely safe profile of induced cytokines, and antitumor efficacy demonstrated in a number of animal models. Here, we decipher the molecular and cellular events underlying the metastasis suppressive activity of entolimod, a clinical stage TLR5 agonist that activates NF-κB-, AP-1-, and STAT3-driven immunomodulatory signaling pathways specifically within the liver. Used as a single agent in murine colon and mammary metastatic cancer models, entolimod rapidly induces CXCL9 and -10 that support homing of blood-borne CXCR3-expressing NK cells to the liver predominantly through an IFN-γ signaling independent mechanism. NK cell-dependent activation of dendritic cells is followed by stimulation of a CD8(+) T-cell response, which exert both antimetastatic effect of entolimod and establishment of tumor-specific and durable immune memory. These results define systemically administered TLR5 agonists as organ-specific immunoadjuvants, enabling efficient antitumor vaccination that does not depend on identification of tumor-specific antigens.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Killer Cells, Natural/drug effects , Neoplasm Metastasis/prevention & control , Peptides/pharmacology , Toll-Like Receptor 5/agonists , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Killer Cells, Natural/immunology , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
7.
Proc Natl Acad Sci U S A ; 112(49): 15154-9, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26575629

ABSTRACT

Recent studies have demonstrated abundant transcription of a set of noncoding RNAs (ncRNAs) preferentially within tumors as opposed to normal tissue. Using an approach from statistical physics, we quantify global transcriptome-wide motif use for the first time, to our knowledge, in human and murine ncRNAs, determining that most have motif use consistent with the coding genome. However, an outlier subset of tumor-associated ncRNAs, typically of recent evolutionary origin, has motif use that is often indicative of pathogen-associated RNA. For instance, we show that the tumor-associated human repeat human satellite repeat II (HSATII) is enriched in motifs containing CpG dinucleotides in AU-rich contexts that most of the human genome and human adapted viruses have evolved to avoid. We demonstrate that a key subset of these ncRNAs functions as immunostimulatory "self-agonists" and directly activates cells of the mononuclear phagocytic system to produce proinflammatory cytokines. These ncRNAs arise from endogenous repetitive elements that are normally silenced, yet are often very highly expressed in cancers. We propose that the innate response in tumors may partially originate from direct interaction of immunogenic ncRNAs expressed in cancer cells with innate pattern recognition receptors, and thereby assign a previously unidentified danger-associated function to a set of dark matter repetitive elements. These findings potentially reconcile several observations concerning the role of ncRNA expression in cancers and their relationship to the tumor microenvironment.


Subject(s)
Neoplasms/genetics , RNA, Untranslated/immunology , Animals , Humans , Immunity, Innate , Mice , Neoplasms/immunology
8.
J Surg Res ; 219: 25-32, 2017 11.
Article in English | MEDLINE | ID: mdl-29078890

ABSTRACT

BACKGROUND: The liver has unique anatomy in that most blood flow to normal hepatocytes is derived from the portal venous system, whereas liver tumors obtain their nutrient blood supply exclusively from the hepatic artery. The focused arterial delivery of anticancer agents to liver tumors has been performed for decades; however, preclinical models to standardize drug regimens and examine novel agents have been lacking. The purpose of this study was to establish preclinical hepatic artery infusion (HAI) models in a mouse and to evaluate the safety and delivery capability of the models. MATERIAL AND METHODS: C57BL/6 and BALB/c mice were used to develop models of HAI via the hepatic artery (HA), superior pancreaticoduodenal artery (SPDA), or lienogastric artery (LGA). Success rates, distribution of perfusion, and associated morbidity and mortality were analyzed between groups. RESULTS: All three models were feasible and reproducible in mice, and there was no statistical difference on body weight change between models. The HA model had a 13.3% mortality from acute liver failure, and the SPDA model demonstrated duodenal and pancreatic toxicity. SPDA and LGA routes had the highest success rates (96.7% and 91.4%, respectively) with low mortality, better drug delivery, and preserved physiologic liver function compared with the HA model. CONCLUSIONS: The optimal route of HAI was mouse breed specific; SPDA access in BALB/c mice, and the LGA access in C57BL/6 mice. The described techniques serve as a reproducible platform for the identification and characterization of therapeutics for diverse metastatic liver tumors.


Subject(s)
Antineoplastic Agents/administration & dosage , Colonic Neoplasms/pathology , Fluorouracil/administration & dosage , Hepatic Artery , Liver Neoplasms, Experimental/secondary , Liver Neoplasms/secondary , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Female , Fluorouracil/therapeutic use , Infusions, Intra-Arterial , Liver Neoplasms/drug therapy , Liver Neoplasms, Experimental/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Reproducibility of Results , Treatment Outcome
9.
Proc Natl Acad Sci U S A ; 111(4): E474-83, 2014 Jan 28.
Article in English | MEDLINE | ID: mdl-24474797

ABSTRACT

Chronic inflammation is associated with increased cancer risk. Furthermore, the transcription factor NF-κB, a central regulator of inflammatory responses, is constitutively active in most tumors. To determine whether active NF-κB inherently contributes to malignant transformation, we isolated a set of NF-κB-activating genetic elements and tested their oncogenic potential in rodent cell transformation models. Genetic elements with desired properties were isolated using biologically active selectable peptide technology, which involves functional screening of lentiviral libraries encoding 20 or 50 amino acid-long polypeptides supplemented with endoplasmic reticulum-targeting and oligomerization domains. Twelve NF-κB-activating selectable peptides (NASPs) representing specific fragments of six proteins, none of which was previously associated with NF-κB activation, were isolated from libraries of 200,000 peptides derived from 500 human extracellular proteins. Using selective knockdown of distinct components of the NF-κB pathway, we showed that the isolated NASPs act either via or upstream of TNF receptor-associated factor 6. Transduction of NASPs into mouse and rat embryo fibroblasts did not, in itself, alter their growth. However, when coexpressed with oncogenic Ras (H-Ras(V12)), NASPs allowed rodent fibroblasts to overcome H-Ras(V12)-mediated p53-dependent senescence and acquire a transformed tumorigenic phenotype. Consistent with their ability to cooperate with oncogenic Ras in cell transformation, NASP expression reduced the transactivation activity of p53. This system provides an in vitro model of NF-κB-driven carcinogenesis and suggests that the known carcinogenic effects of inflammation may be at least partially due to NF-κB-mediated abrogation of oncogene-induced senescence.


Subject(s)
Carcinogenesis , Genes, ras , Inflammation/metabolism , NF-kappa B/metabolism , Peptides/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Humans , Inflammation/genetics , Mice , Molecular Sequence Data , Peptides/chemistry , Protein Binding , Rats
10.
Proc Natl Acad Sci U S A ; 110(1): E89-98, 2013 Jan 02.
Article in English | MEDLINE | ID: mdl-23236145

ABSTRACT

Large parts of mammalian genomes are transcriptionally inactive and enriched with various classes of interspersed and tandem repeats. Here we show that the tumor suppressor protein p53 cooperates with DNA methylation to maintain silencing of a large portion of the mouse genome. Massive transcription of major classes of short, interspersed nuclear elements (SINEs) B1 and B2, both strands of near-centromeric satellite DNAs consisting of tandem repeats, and multiple species of noncoding RNAs was observed in p53-deficient but not in p53 wild-type mouse fibroblasts treated with the DNA demethylating agent 5-aza-2'-deoxycytidine. The abundance of these transcripts exceeded the level of ß-actin mRNA by more than 150-fold. Accumulation of these transcripts, which are capable of forming double-stranded RNA (dsRNA), was accompanied by a strong, endogenous, apoptosis-inducing type I IFN response. This phenomenon, which we named "TRAIN" (for "transcription of repeats activates interferon"), was observed in spontaneous tumors in two models of cancer-prone mice, presumably reflecting naturally occurring DNA hypomethylation and p53 inactivation in cancer. These observations suggest that p53 and IFN cooperate to prevent accumulation of cells with activated repeats and provide a plausible explanation for the deregulation of IFN function frequently seen in tumors. Overall, this work reveals roles for p53 and IFN that are key for genetic stability and therefore relevant to both tumorigenesis and the evolution of species.


Subject(s)
DNA Methylation , Epigenetic Repression/physiology , Interferon Type I/metabolism , RNA, Untranslated/genetics , Repetitive Sequences, Nucleic Acid/genetics , Tumor Suppressor Protein p53/genetics , Actins/genetics , Animals , Azacitidine/analogs & derivatives , Computational Biology , Decitabine , Epigenetic Repression/genetics , Mice , Microarray Analysis , Sequence Analysis, RNA , Tumor Suppressor Protein p53/deficiency
11.
Proc Natl Acad Sci U S A ; 110(20): E1857-66, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23630282

ABSTRACT

Vertebrate Toll-like receptor 5 (TLR5) recognizes bacterial flagellin proteins and activates innate immune responses to motile bacteria. In addition, activation of TLR5 signaling can inhibit growth of TLR5-expressing tumors and protect normal tissues from radiation and ischemia-reperfusion injuries. To understand the mechanisms behind these phenomena at the organismal level, we assessed nuclear factor kappa B (NF-κB) activation (indicative of TLR5 signaling) in tissues and cells of mice treated with CBLB502, a pharmacologically optimized flagellin derivative. This identified the liver and gastrointestinal tract as primary CBLB502 target organs. In particular, liver hepatocytes were the main cell type directly and specifically responding to systemic administration of CBLB502 but not to that of the TLR4 agonist LPS. To assess CBLB502 impact on other pathways, we created multireporter mice with hepatocytes transduced in vivo with reporters for 46 inducible transcription factor families and found that along with NF-κB, CBLB502 strongly activated STAT3-, phenobarbital-responsive enhancer module (PREM), and activator protein 1 (AP-1-) -driven pathways. Livers of CBLB502-treated mice displayed induction of numerous immunomodulatory factors and massive recruitment of various types of immune cells. This led to inhibition of growth of liver metastases of multiple tumors regardless of their TLR5 status. The changed liver microenvironment was not, however, hepatotoxic, because CBLB502 induced resistance to Fas-mediated apoptosis in normal liver cells. Temporary occlusion of liver blood circulation prevented CBLB502 from protecting hematopoietic progenitors in lethally irradiated mice, indicating involvement of a factor secreted by responding liver cells. These results define the liver as the key mediator of TLR5-dependent effects in vivo and suggest clinical applications for TLR5 agonists as hepatoprotective and antimetastatic agents.


Subject(s)
Liver/metabolism , Peptides/pharmacology , STAT3 Transcription Factor/metabolism , Toll-Like Receptor 5/agonists , Animals , Anticarcinogenic Agents/pharmacology , Breast Neoplasms/metabolism , Cell Line, Tumor , Colonic Neoplasms/metabolism , Female , Flow Cytometry , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Killer Cells, Natural/metabolism , Liver/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NF-kappa B/metabolism , Neoplasm Transplantation , Neutrophils/metabolism , Radiation-Protective Agents/pharmacology , Signal Transduction , fas Receptor/metabolism
12.
Proc Natl Acad Sci U S A ; 109(47): 19392-6, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23129611

ABSTRACT

Blind mole rats Spalax (BMR) are small subterranean rodents common in the Middle East. BMR is distinguished by its adaptations to life underground, remarkable longevity (with a maximum documented lifespan of 21 y), and resistance to cancer. Spontaneous tumors have never been observed in spalacids. To understand the mechanisms responsible for this resistance, we examined the growth of BMR fibroblasts in vitro of the species Spalax judaei and Spalax golani. BMR cells proliferated actively for 7-20 population doublings, after which the cells began secreting IFN-ß, and the cultures underwent massive necrotic cell death within 3 d. The necrotic cell death phenomenon was independent of culture conditions or telomere shortening. Interestingly, this cell behavior was distinct from that observed in another long-lived and cancer-resistant African mole rat, Heterocephalus glaber, the naked mole rat in which cells display hypersensitivity to contact inhibition. Sequestration of p53 and Rb proteins using SV40 large T antigen completely rescued necrotic cell death. Our results suggest that cancer resistance of BMR is conferred by massive necrotic response to overproliferation mediated by p53 and Rb pathways, and triggered by the release of IFN-ß. Thus, we have identified a unique mechanism that contributes to cancer resistance of this subterranean mammal extremely adapted to life underground.


Subject(s)
Disease Resistance/immunology , Fibroblasts/pathology , Necrosis/pathology , Neoplasms/pathology , Spalax/immunology , Animals , Antigens, Polyomavirus Transforming/metabolism , Contact Inhibition , Fibroblasts/metabolism , Humans , Interferon-beta/metabolism , Male , Mice , Phenotype , Retinoblastoma Protein/metabolism , Signal Transduction , Telomere Shortening , Tumor Suppressor Protein p53/metabolism
13.
Proc Natl Acad Sci U S A ; 109(33): 13314-8, 2012 Aug 14.
Article in English | MEDLINE | ID: mdl-22847439

ABSTRACT

Unlike reversible quiescence, cellular senescence is characterized by a large flat cell morphology, ß-gal staining and irreversible loss of regenerative (i.e., replicative) potential. Conversion from proliferative arrest to irreversible senescence, a process named geroconversion, is driven in part by growth-promoting pathways such as mammalian target of rapamycin (mTOR). During cell cycle arrest, mTOR converts reversible arrest into senescence. Inhibitors of mTOR can suppress geroconversion, maintaining quiescence instead. It was shown that hypoxia inhibits mTOR. Therefore, we suggest that hypoxia may suppress geroconversion. Here we tested this hypothesis. In HT-p21-9 cells, expression of inducible p21 caused cell cycle arrest without inhibiting mTOR, leading to senescence. Hypoxia did not prevent p21 induction and proliferative arrest, but instead inhibited the mTOR pathway and geroconversion. Exposure to hypoxia during p21 induction prevented senescent morphology and loss of regenerative potential, thus maintaining reversible quiescence so cells could restart proliferation after switching p21 off. Suppression of geroconversion was p53- and HIF-1-independent, as hypoxia also suppressed geroconversion in cells lacking functional p53 and HIF-1α. Also, in normal fibroblasts and retinal cells, hypoxia inhibited the mTOR pathway and suppressed senescence caused by etoposide without affecting DNA damage response, p53/p21 induction and cell cycle arrest. Also hypoxia suppressed geroconversion in cells treated with nutlin-3a, a nongenotoxic inducer of p53, in cell lines susceptible to nutlin-3a-induced senescence (MEL-10, A172, and NKE). Thus, in normal and cancer cell lines, hypoxia suppresses geroconversion caused by diverse stimuli. Physiological and clinical implications of the present findings are discussed.


Subject(s)
Cellular Senescence , Epithelial Cells/cytology , Fibroblasts/cytology , Cell Hypoxia/drug effects , Cell Line , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Etoposide/pharmacology , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Imidazoles/pharmacology , Piperazines/pharmacology , Tumor Suppressor Protein p53/metabolism
14.
Proc Natl Acad Sci U S A ; 109(37): E2457-65, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22895791

ABSTRACT

The circadian clock controls many physiological parameters including immune response to infectious agents, which is mediated by activation of the transcription factor NF-κB. It is widely accepted that circadian regulation is based on periodic changes in gene expression that are triggered by transcriptional activity of the CLOCK/BMAL1 complex. Through the use of a mouse model system we show that daily variations in the intensity of the NF-κB response to a variety of immunomodulators are mediated by core circadian protein CLOCK, which can up-regulate NF-κB-mediated transcription in the absence of BMAL1; moreover, BMAL1 counteracts the CLOCK-dependent increase in the activation of NF-κB-responsive genes. Consistent with its regulatory function, CLOCK is found in protein complexes with the p65 subunit of NF-κB, and its overexpression correlates with an increase in specific phosphorylated and acetylated transcriptionally active forms of p65. In addition, activation of NF-κB in response to immunostimuli in mouse embryonic fibroblasts and primary hepatocytes isolated from Clock-deficient mice is significantly reduced compared with WT cells, whereas Clock-Δ19 mutation, which reduces the transactivation capacity of CLOCK on E-box-containing circadian promoters, has no effect on the ability of CLOCK to up-regulate NF-κB-responsive promoters. These findings establish a molecular link between two essential determinants of the circadian and immune mechanisms, the transcription factors CLOCK and NF-κB, respectively.


Subject(s)
CLOCK Proteins/metabolism , Circadian Rhythm/physiology , Gene Expression Regulation/immunology , Gene Expression Regulation/physiology , Transcription Factor RelA/metabolism , Transcription, Genetic/physiology , Analysis of Variance , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation/genetics , Humans , Immunoprecipitation , Luciferases , Mice , Mice, Inbred BALB C , Microscopy, Fluorescence , Peptides , Transcription, Genetic/genetics
15.
J Immunol ; 189(10): 4719-27, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23045613

ABSTRACT

Allogeneic hematopoietic cell transplantation is an established treatment for hematologic and nonhematologic malignancies. Donor-derived immune cells can identify and attack host tumor cells, producing a graft-versus-tumor (GVT) effect that is crucial to the effectiveness of the transplantation therapy. CBLB502 is a novel agonist for TLR5 derived from Salmonella flagellin. On the basis of TLR5-mediated immunomodulatory function, we examined the effect of CBLB502 on GVT activity. Using two tumor models that do not express TLR5, and thereby do not directly respond to CBLB502, we found that CBLB502 treatment significantly enhanced allogeneic CD8(+) T cell-mediated GVT activity, which was evidenced by decreased tumor burden and improved host survival. Importantly, histopathologic analyses showed that CBLB502 treatment did not exacerbate the moderate graft-versus-host disease condition caused by the allogeneic CD8(+) T cells. Moreover, mechanistic analyses showed that CBLB502 stimulates CD8(+) T cell proliferation and enhances their tumor killing activity mainly indirectly through a mechanism that involves the IL-12 signaling pathway and the CD11c(+) and CD11b(+) populations in the bone marrow cells. This study demonstrates a new beneficial effect of CBLB502, and suggests that TLR5-mediated immune modulation may be a promising approach to improve GVT immunity without exacerbating graft-versus-host disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Flagellin/pharmacology , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation , Immunity, Cellular/drug effects , Neoplasms, Experimental/therapy , Salmonella/chemistry , Toll-Like Receptor 5/agonists , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Flagellin/chemistry , Flagellin/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Tumor Effect/immunology , Immunity, Cellular/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Toll-Like Receptor 5/immunology , Transplantation, Homologous
16.
Infect Immun ; 81(10): 3855-64, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897616

ABSTRACT

Pathogen recognition receptors (PRRs) are essential components of host innate immune systems that detect specific conserved pathogen-associated molecular patterns (PAMPs) presented by microorganisms. Members of two families of PRRs, transmembrane Toll-like receptors (TLRs 1, 2, 4, 5, and 6) and cytosolic NOD receptors (NOD1 and NOD2), are stimulated upon recognition of various bacterial PAMPs. Such stimulation leads to induction of a number of immune defense reactions, mainly triggered via activation of the transcription factor NF-κB. While coordination of responses initiated via different PRRs sensing multiple PAMPS present during an infection makes clear biological sense for the host, such interactions have not been fully characterized. Here, we demonstrate that combined stimulation of NOD1 and TLR5 (as well as other NOD and TLR family members) strongly potentiates activity of NF-κB and induces enhanced levels of innate immune reactions (e.g., cytokine production) both in vitro and in vivo. Moreover, we show that an increased level of NF-κB activity plays a critical role in formation of downstream responses. In live mice, synergy between these receptors resulting in potentiation of NF-κB activity was organ specific, being most prominent in the gastrointestinal tract. Coordinated activity of NOD1 and TLR5 significantly increased protection of mice against enteroinvasive Salmonella infection. Obtained results suggest that cooperation of NOD and TLR receptors is important for effective responses to microbial infection in vivo.


Subject(s)
Immunity, Innate/physiology , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Salmonella typhimurium , Toll-Like Receptor 5/metabolism , Animals , Female , Gene Expression Regulation/immunology , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , Nod1 Signaling Adaptor Protein/genetics , Salmonella Infections, Animal/immunology , Salmonella Infections, Animal/microbiology , Toll-Like Receptor 5/genetics
17.
J Immunol ; 187(7): 3831-9, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21890657

ABSTRACT

Reperfusion of ischemic organs induces a potent inflammatory response initiated by the generation of reactive oxygen species that directly damage tissue and promote leukocyte infiltration and activation that also mediate tissue injury. We recently found that radiation-induced tissue injury, which is caused by radiation-induced reactive oxygen species, is attenuated by administration of CBLB502, a pharmacologically optimized derivative of the TLR5 agonist flagellin. Therefore, we tested the ability of CBLB502 to attenuate injury in a murine model of acute ischemic renal failure. CBLB502 given 30 min before imposition of bilateral renal pedicle occlusion provided marked protection against the renal dysfunction and inflammation that follows reperfusion of ischemic kidneys, including marked decreases in leukocyte infiltration, proinflammatory cytokine production, and tubular injury. Importantly, CBLB502 given within 30 min after ischemic kidney reperfusion reproduced the protective effects of pretreatment with the TLR5 agonist, indicating a window following reperfusion in which CBLB502 administration abrogates acute renal ischemic failure. Bone marrow-reconstituted chimeras were used to show that the protective effects of CBLB502 could be delivered by intact MyD88 signaling on renal parenchymal cells. Consistent with this, Ab staining of kidney sections indicated that cells lining the renal vasculature expressed TLR5. Overall, these results indicate the use of TLR5 agonists as mitigators and protectants of acute renal ischemic failure.


Subject(s)
Acute Kidney Injury/prevention & control , Anti-Inflammatory Agents/pharmacology , Peptides/pharmacology , Reperfusion Injury/prevention & control , Toll-Like Receptor 5/agonists , Acute Kidney Injury/immunology , Acute Kidney Injury/pathology , Animals , Immunohistochemistry , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/drug effects , Reperfusion Injury/immunology , Reperfusion Injury/pathology
18.
Nat Rev Cancer ; 3(2): 117-29, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12563311

ABSTRACT

Ionizing radiation (IR) has proven to be a powerful medical treatment in the fight against cancer. Rational and effective use of its killing power depends on understanding IR-mediated responses at the molecular, cellular and tissue levels. Tumour cells frequently acquire defects in the molecular regulatory mechanisms of the response to IR, which sensitizes them to radiation therapy. One of the key molecules involved in a cell's response to IR is p53. Understanding these mechanisms indicates new rational approaches to improving cancer treatment by IR.


Subject(s)
Neoplasms/radiotherapy , Radiation Tolerance , Tumor Suppressor Protein p53/physiology , Humans
19.
Proc Natl Acad Sci U S A ; 107(21): 9660-4, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20457898

ABSTRACT

The tumor suppressor p53 is a canonical inducer of cellular senescence (irreversible loss of proliferative potential and senescent morphology). p53 can also cause reversible arrest without senescent morphology, which has usually been interpreted as failure of p53 to induce senescence. Here we demonstrate that p53-induced quiescence actually results from suppression of senescence by p53. In previous studies, suppression of senescence by p53 was masked by p53-induced cell cycle arrest. Here, we separated these two activities by inducing senescence through overexpression of p21 and then testing the effect of p53 on senescence. We found that in p21-arrested cells, p53 converted senescence into quiescence. Suppression of senescence by p53 required its transactivation function. Like rapamycin, which is known to suppress senescence, p53 inhibited the mTOR pathway. We suggest that, while inducing cell cycle arrest, p53 may simultaneously suppress the senescence program, thus causing quiescence and that suppression of senescence and induction of cell cycle arrest are distinct functions of p53. Thus, in spite of its ability to induce cell cycle arrest, p53 can act as a suppressor of cellular senescence.


Subject(s)
Cellular Senescence , Tumor Suppressor Protein p53/metabolism , Cell Line, Tumor , Cellular Senescence/drug effects , Humans , Imidazoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Piperazines/pharmacology , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Transcriptional Activation , Tumor Suppressor Protein p53/genetics
20.
Proc Natl Acad Sci U S A ; 107(1): 46-51, 2010 Jan 05.
Article in English | MEDLINE | ID: mdl-20080798

ABSTRACT

NF-kappaB, a central coordinator of immune and inflammatory responses, must be tightly regulated. We describe a NF-kappaB regulatory pathway that is driven by reversible lysine methylation of the p65 subunit, carried out by a lysine methylase, the nuclear receptor-binding SET domain-containing protein 1 (NSD1), and a lysine demethylase, F-box and leucine-rich repeat protein 11 (FBXL11). Overexpression of FBXL11 inhibits NF-kappaB activity, and a high level of NSD1 activates NF-kappaB and reverses the inhibitory effect of FBXL11, whereas reduced expression of NSD1 decreases NF-kappaB activation. The targets are K218 and K221 of p65, which are methylated in cells with activated NF-kappaB. Overexpression of FBXL11 slowed the growth of HT29 cancer cells, whereas shRNA-mediated knockdown had the opposite effect, and these phenotypes were dependent on K218/K221 methylation. In mouse embryo fibroblasts, the activation of most p65-dependent genes relied on K218/K221 methylation. Importantly, expression of the FBXL11 gene is driven by NF-kappaB, revealing a negative regulatory feedback loop. We conclude that reversible lysine methylation of NF-kappaB is an important element in the complex regulation of this key transcription factor.


Subject(s)
F-Box Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lysine/metabolism , Nuclear Proteins/metabolism , Transcription Factor RelA/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Embryo, Mammalian/cytology , F-Box Proteins/genetics , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Humans , Intracellular Signaling Peptides and Proteins/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Methylation , Mice , Mice, Knockout , Molecular Sequence Data , Nuclear Proteins/genetics , Oxidoreductases, N-Demethylating , Protein Methyltransferases/genetics , Protein Methyltransferases/metabolism , Protein Subunits/genetics , Protein Subunits/metabolism , Transcription Factor RelA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL