Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Exp Cell Res ; 349(1): 15-22, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27693451

ABSTRACT

The vitamin D receptor (VDR), and its ligand 1α,25-dihydroxyvitamin D3 (1,25D3) prevent breast cancer development and progression, yet the molecular mechanisms governing this are unclear. MicroRNAs (miRNAs) on the other hand, promote or inhibit breast cancer growth. To understand how VDR regulates miRNAs, we compared miRNA expression of wild-type (WT) and VDR knockout (VDRKO) breast cancer cells by a Mouse Breast Cancer miRNA PCR array. Compared to VDR WT cells, expressions of miR-214, miR-199a-3p and miR-199a-5p of the miR-199a/miR-214 cluster were 42, 15, and 10 fold higher in VDRKO cells respectively. Overexpression of VDR in breast cancer cells reduced the miR-199a/miR-214 cluster expression by 30%. VDR status also negatively correlated with Dnm3os expression, a non-coding RNA transcript of the dynamin-3 gene encoding the miR-199a/miR-214 cluster, suggesting that VDR represses this cluster through Dnm3os. Conversely, overexpression of miR-214 in MCF-7 and T47D cells antagonized VDR mediated signaling. Furthermore, there was a positive correlation between VDR status and the expression of Suppressor of fused gene (SuFu), a hedgehog pathway inhibitor. miR-214 on the other hand suppressed SuFu protein expression. These findings suggest a crosstalk between VDR and miR-214 in regulating hedgehog signaling in breast cancer cells, providing new therapies for breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Hedgehog Proteins/metabolism , MicroRNAs/metabolism , Receptors, Calcitriol/metabolism , Repressor Proteins/metabolism , Signal Transduction , Animals , Breast Neoplasms/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Signal Transduction/drug effects , Vitamin D/analogs & derivatives , Vitamin D/pharmacology
2.
Br J Cancer ; 114(11): 1180-4, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27140310

ABSTRACT

Cellular senescence is an established tumour-suppressive mechanism that prevents the proliferation of premalignant cells. However, several lines of evidence show that senescent cells, which often persist in vivo, can also promote tumour progression in addition to other age-related pathologies via the senescence-associated secretory phenotype (SASP). Moreover, new insights suggest the SASP can facilitate tissue repair. Here, we review the beneficial and detrimental roles of senescent cells, highlighting conditions under which the senescence response does and does not promote pathology, particularly cancer. By better understanding the context-dependent effects of cellular senescence, it may be feasible to limit its detrimental properties while preserving its beneficial effects, and develop novel therapeutic strategies to prevent or treat cancer and possibly other age-associated diseases.


Subject(s)
Cellular Senescence/physiology , Neoplasms/pathology , Animals , Cell Division , Cellular Microenvironment , Embryonic Development , Extracellular Matrix/metabolism , Humans , Inflammation , Mice , Neoplasm Invasiveness , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic , Phenotype , Regeneration
3.
Mol Cell Biochem ; 372(1-2): 249-56, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23001870

ABSTRACT

The nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ) plays a central role in regulating metabolism, including interaction with the estrogen receptor-α (ERα). Significantly, PPARγ activity can be modulated by small molecules to control cancer both in vitro and in vivo (Yin et al., Cancer Res 69:687-694, 2009). Here, we evaluated the effects of the PPARγ agonist GW7845 and the PPARγ antagonist GW9662 on DMBA-induced mammary alveolar lesions (MAL) in a mouse mammary organ culture. The results were as follows: (a) the incidence of MAL development was significantly inhibited by GW 7845 and GW 9662; (b) GW9662 but not GW7845, in the presence of estradiol, induced ER and PR expression in mammary glands and functional ERα in MAL; (c) while GW9662 inhibited expression of adipsin and ap2, GW 7845 enhanced expression of these PPARγ-response genes; and (d) Tamoxifen caused significant inhibition of GW9662 treated MAL, suggesting that GW9662 sensitizes MAL to antiestrogen treatment, presumably through rendering functional ERα and induction of PR. The induction of ERα by GW9662, including newer analogs, may permit use of anti-ER strategies to inhibit breast cancer in ER- patients.


Subject(s)
Anilides/pharmacology , Anticarcinogenic Agents/pharmacology , Estrogen Receptor alpha/metabolism , Mammary Glands, Animal/metabolism , PPAR gamma/antagonists & inhibitors , 9,10-Dimethyl-1,2-benzanthracene , Animals , Drug Synergism , Estradiol/physiology , Estrogen Receptor alpha/genetics , Female , Mammary Glands, Animal/drug effects , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Oxazoles/pharmacology , PPAR gamma/agonists , PPAR gamma/metabolism , Precancerous Conditions/chemically induced , Precancerous Conditions/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Tamoxifen/pharmacology , Tissue Culture Techniques , Transcriptional Activation/drug effects , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
4.
Exp Cell Res ; 318(19): 2490-7, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-22884583

ABSTRACT

Heterodimerization and cross-talk between nuclear hormone receptors often occurs. For example, estrogen receptor alpha (ERα) physically binds to peroxisome proliferator-activated receptor gamma (PPARγ) and inhibits its transcriptional activity. The interaction between PPARγ and the vitamin D receptor (VDR) however, is unknown. Here, we elucidate the molecular mechanisms linking PPARγ and VDR signaling, and for the first time we show that PPARγ physically associates with VDR in human breast cancer cells. We found that overexpression of PPARγ decreased 1α,25-dihydroxyvitamin D(3) (1,25D(3)) mediated transcriptional activity of the vitamin D target gene, CYP24A1, by 49% and the activity of VDRE-luc, a vitamin D responsive reporter, by 75% in T47D human breast cancer cells. Deletion mutation experiments illustrated that helices 1 and 4 of PPARγ's hinge and ligand binding domains, respectively, governed this suppressive function. Additionally, abrogation of PPARγ's AF2 domain attenuated its repressive action on 1,25D(3) transactivation, indicating that this domain is integral in inhibiting VDR signaling. PPARγ was also found to compete with VDR for their binding partner retinoid X receptor alpha (RXRα). Overexpression of RXRα blocked PPARγ's suppressive effect on 1,25D(3) action, enhancing VDR signaling. In conclusion, these observations uncover molecular mechanisms connecting the PPARγ and VDR pathways.


Subject(s)
Breast Neoplasms/genetics , Cholestanetriol 26-Monooxygenase/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Receptor Cross-Talk/physiology , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Transcriptional Activation , Breast Neoplasms/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , MCF-7 Cells , Male , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Protein Binding , Protein Structure, Tertiary , Retinoid X Receptor alpha/genetics , Retinoid X Receptor alpha/metabolism , Sequence Deletion , Signal Transduction , Steroid Hydroxylases/genetics , Steroid Hydroxylases/metabolism , Tumor Cells, Cultured , Vitamin D3 24-Hydroxylase
5.
Nat Metab ; 4(8): 995-1006, 2022 08.
Article in English | MEDLINE | ID: mdl-35902645

ABSTRACT

ABSTACT: Ageing is the largest risk factor for many chronic diseases. Studies of heterochronic parabiosis, substantiated by blood exchange and old plasma dilution, show that old-age-related factors are systemically propagated and have pro-geronic effects in young mice. However, the underlying mechanisms how bloodborne factors promote ageing remain largely unknown. Here, using heterochronic blood exchange in male mice, we show that aged mouse blood induces cell and tissue senescence in young animals after one single exchange. This induction of senescence is abrogated if old animals are treated with senolytic drugs before blood exchange, therefore attenuating the pro-geronic influence of old blood on young mice. Hence, cellular senescence is neither simply a response to stress and damage that increases with age, nor a chronological cell-intrinsic phenomenon. Instead, senescence quickly and robustly spreads to young mice from old blood. Clearing senescence cells that accumulate with age rejuvenates old circulating blood and improves the health of multiple tissues.


Subject(s)
Cellular Senescence , Parabiosis , Aging/physiology , Animals , Cellular Senescence/physiology , Male , Mice
6.
Mech Ageing Dev ; 199: 111561, 2021 10.
Article in English | MEDLINE | ID: mdl-34411604

ABSTRACT

Wound healing is impaired with advanced age and certain chronic conditions, such as diabetes and obesity. Moreover, common cancer treatments, including chemotherapy and radiation, can cause unintended tissue damage and impair wound healing. Available wound care treatments are not always effective, as some wounds fail to heal or recur after treatment. Hence, a more thorough understanding of the pathophysiology of chronic, nonhealing wounds may offer new ideas for the development of effective wound care treatments. Cancers are sometimes referred to as wounds that never heal, sharing mechanisms similar to wound healing. We describe in this review how cellular senescence and the senescence-associated secretory phenotype (SASP) contribute to chronic wounds versus cancer.


Subject(s)
Immunosenescence , Neoplasms , Senescence-Associated Secretory Phenotype , Wounds and Injuries , Cellular Senescence/physiology , Humans , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Paracrine Communication , Wound Healing , Wounds and Injuries/immunology , Wounds and Injuries/pathology , Wounds and Injuries/therapy
7.
J Invest Dermatol ; 141(4S): 1119-1126, 2021 04.
Article in English | MEDLINE | ID: mdl-33349436

ABSTRACT

Chronic exposure to UVR is known to disrupt tissue homeostasis, accelerate the onset of age-related phenotypes, and increase the risk for skin cancer-a phenomenon defined as photoaging. In this paper, we review the current knowledge on how UV exposure causes cells to prematurely enter cellular senescence. We describe the mechanisms contributing to the accumulation of senescent cells in the skin and how the persistence of cellular senescence can promote impaired regenerative capacity, chronic inflammation, and tumorigenesis associated with photoaging. We conclude by highlighting the potential of senolytic drugs in delaying the onset and progression of age-associated phenotypes in the skin.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Cellular Senescence/radiation effects , Skin Aging/pathology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Cell Transformation, Neoplastic/pathology , Humans , Skin/cytology , Skin/pathology , Skin/radiation effects , Skin Aging/radiation effects , Skin Neoplasms/etiology
8.
Cell Metab ; 33(6): 1124-1136.e5, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33811820

ABSTRACT

Cellular senescence is a stress or damage response that causes a permanent proliferative arrest and secretion of numerous factors with potent biological activities. This senescence-associated secretory phenotype (SASP) has been characterized largely for secreted proteins that participate in embryogenesis, wound healing, inflammation, and many age-related pathologies. By contrast, lipid components of the SASP are understudied. We show that senescent cells activate the biosynthesis of several oxylipins that promote segments of the SASP and reinforce the proliferative arrest. Notably, senescent cells synthesize and accumulate an unstudied intracellular prostaglandin, 1a,1b-dihomo-15-deoxy-delta-12,14-prostaglandin J2. Released 15-deoxy-delta-12,14-prostaglandin J2 is a biomarker of senolysis in culture and in vivo. This and other prostaglandin D2-related lipids promote the senescence arrest and SASP by activating RAS signaling. These data identify an important aspect of cellular senescence and a method to detect senolysis.


Subject(s)
Oxylipins/metabolism , Senescence-Associated Secretory Phenotype , Senotherapeutics/metabolism , Animals , Biomarkers/metabolism , Cell Line , Humans , Mice , Mice, Inbred C57BL
9.
J Cell Biochem ; 110(6): 1324-33, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20564226

ABSTRACT

25-Hydroxyvitamin D(3) (25(OH)D(3)) is a prohormone and a major vitamin D metabolite. The discovery of (25(OH)D(3)) 1 alpha-hydroxylase in many vitamin D target organs has yielded an increased interest in defining the role(s) of 25(OH)D(3) in these tissues. The etiology of cancer appears to be complex and multi-factorial. Cellular stress (e.g., DNA damage, hypoxia, oncogene activation) has been identified as one of the key factors responsible for initiating the carcinogenesis process. In this study, we investigated whether 25(OH)D(3) protects breast epithelial cells from cellular stress using an established breast epithelial cell line MCF12F. To better elucidate the role of 25(OH)D(3) in the stress response, we used multiple in vitro stress models including serum starvation, hypoxia, oxidative stress, and apoptosis induction. Under all these stress conditions, 25(OH)D(3) (250 nmol/L) treatment significantly protected cells against cell death. Low-serum stress induced p53 expression accompanied with downregulation of PCNA, the presence of 25(OH)D(3) consistently inhibited the alteration of p53 and PCNA, suggesting that these molecules were involved in the stress process and may be potential target genes of 25(OH)D(3). miRNA microarray analysis demonstrated that stress induced by serum starvation caused significant alteration in the expression of multiple miRNAs including miR182, but the presence of 25(OH)D(3) effectively reversed this alteration. These data suggest that there is a significant protective role for 25(OH)D(3) against cellular stress in the breast epithelial cells and these effects may be mediated by altered miRNA expression.


Subject(s)
Apoptosis/drug effects , Calcifediol/pharmacology , Epithelial Cells/drug effects , Oxidative Stress/drug effects , Blotting, Western , Breast/cytology , Breast/metabolism , Cell Hypoxia , Cell Line , Cell Line, Tumor , Culture Media/chemistry , Culture Media/pharmacology , Culture Media, Serum-Free/pharmacology , Cysteine Proteinase Inhibitors/pharmacology , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Humans , Hydrogen Peroxide/pharmacology , Leupeptins/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Oxidants/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/metabolism , Vitamins/pharmacology
10.
Mol Cell Biochem ; 342(1-2): 143-50, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20440542

ABSTRACT

It is well-established that CYP24, an immediate target gene of VDR is upregulated by VDR ligands. This study is focused on the functional role of unliganded VDR by investigating the correlation between the expression of VDR protein and basal mRNA levels of CYP24 in breast cancer cell lines. Analyses of multiple breast cancer cell lines demonstrated an inverse correlation between VDR protein expression and CYP24 mRNA expression levels; while in the presence of ligand, VDR protein level was positively correlated with CYP24 expression. In MCF-7 cells, VDR was mainly distributed in the nuclei in the absence of ligand. VDR overexpression in MCF-7 cells and MDA-MB231 cells decreased CYP24 mRNA expression levels and CYP24 promoter activity. Conversely, knock-down of VDR using siRNA techniques in MCF-7 and T47D cells significantly increased CYP24 mRNA expression. We also found that overexpression of VDR with a polymorphic site (FokI-FF) at its AF-1 domain, which makes VDR shorter by three amino acids, failed to repress CYP24 promoter activity. This report provides conclusive evidence for the repressive action of unliganded VDR on the expression of its target gene CYP24 and the importance of an intact VDR AF-1 domain for its repressive action.


Subject(s)
Receptors, Calcitriol/metabolism , Steroid Hydroxylases/genetics , Transcription, Genetic , Blotting, Western , Breast Neoplasms , Cell Proliferation , Female , Humans , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Receptors, Calcitriol/genetics , Reverse Transcriptase Polymerase Chain Reaction , Steroid Hydroxylases/antagonists & inhibitors , Steroid Hydroxylases/metabolism , Tumor Cells, Cultured , Vitamin D3 24-Hydroxylase
11.
PLoS One ; 15(1): e0227887, 2020.
Article in English | MEDLINE | ID: mdl-31945125

ABSTRACT

Neurodegeneration is a major age-related pathology. Cognitive decline is characteristic of patients with Alzheimer's and related dementias and cancer patients after chemo- or radio-therapies. A recently emerged driver of these and other age-related pathologies is cellular senescence, a cell fate that entails a permanent cell cycle arrest and pro-inflammatory senescence-associated secretory phenotype (SASP). Although there is a link between inflammation and neurodegenerative diseases, there are many open questions regarding how cellular senescence affects neurodegenerative pathologies. Among the various cell types in the brain, astrocytes are the most abundant. Astrocytes have proliferative capacity and are essential for neuron survival. Here, we investigated the phenotype of primary human astrocytes made senescent by X-irradiation, and identified genes encoding glutamate and potassium transporters as specifically downregulated upon senescence. This down regulation led to neuronal cell death in co-culture assays. Unbiased RNA sequencing of transcripts expressed by non-senescent and senescent astrocytes confirmed that glutamate homeostasis pathway declines upon senescence. Our results suggest a key role for cellular senescence, particularly in astrocytes, in excitotoxicity, which may lead to neurodegeneration including Alzheimer's disease and related dementias.


Subject(s)
Alzheimer Disease/genetics , Astrocytes/metabolism , Cellular Senescence/genetics , Neurons/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Transport System X-AG/genetics , Astrocytes/pathology , Brain/metabolism , Brain/pathology , Cell Cycle Checkpoints/radiation effects , Cellular Senescence/radiation effects , Gene Expression Regulation/radiation effects , Glutamic Acid/genetics , Glutamic Acid/metabolism , Humans , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Primary Cell Culture , X-Rays
12.
Cancer Res ; 80(17): 3606-3619, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32641409

ABSTRACT

Cellular senescence entails an irreversible growth arrest that evolved in part to prevent cancer. Paradoxically, senescent cells secrete proinflammatory and growth-stimulatory molecules, termed the senescence-associated secretory phenotype (SASP), which is correlated with cancer cell proliferation in culture and xenograft models. However, at what tumor stage and how senescence and the SASP act on endogenous tumor growth in vivo is unknown. To understand the role of senescence in cancer etiology, we subjected p16-3MR transgenic mice, which permit the identification and selective elimination of senescent cells in vivo, to the well-established two-step protocol of squamous cell skin carcinoma, in which tumorigenesis is initiated by a carcinogen 7,12-dimethylbenz[α]anthracene, and then promoted by 12-O-tetradecanoyl-phorbol-13-acetate (TPA). We show that TPA promotes skin carcinogenesis by inducing senescence and a SASP. Systemic induction of senescence in nontumor-bearing p16-3MR mice using a chemotherapy followed by the two-step carcinogenesis protocol potentiated the conversion of benign papillomas to carcinomas by elevating p38MAPK and MAPK/ERK signaling. Ablation of senescent cells reduced p38MAPK and MAPK/ERK signaling, thereby preventing the progression of benign papillomas to carcinomas. Thus, we show for the first time that senescent cells are tumor promoters, not tumor initiators, and that they stimulate skin carcinogenesis by elevating p38MAPK and MAPK/ERK signaling. These findings pave the way for developing novel therapeutics against senescence-fueled cancers. SIGNIFICANCE: These findings identify chemotherapy-induced senescence as a culprit behind tumor promotion, suggesting that elimination of senescent cells after chemotherapy may reduce occurrence of second cancers decades later. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/17/3606/F1.large.jpg.


Subject(s)
Carcinogenesis/metabolism , Carcinoma, Squamous Cell/pathology , Cellular Senescence/physiology , MAP Kinase Signaling System/physiology , Skin Neoplasms/pathology , Animals , Carcinogenesis/pathology , Carcinoma, Squamous Cell/metabolism , Humans , Mice , Mice, Transgenic , Skin Neoplasms/metabolism
13.
Mol Cancer Res ; 6(11): 1732-41, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18974396

ABSTRACT

IFN-inducible IFI16 protein (encoded by IFI16 gene at 1q23.1) is the human member of the IFN-inducible structurally related p200 family proteins. Increased expression of the IFI16 protein, a positive modulator of p53-mediated transcription, in normal old human diploid fibroblasts (HDF) is associated with cellular senescence-mediated cell growth arrest. However, the underlying mechanisms that contribute to transcriptional activation of the IFI16 gene in old HDFs remain to be elucidated. Here, we reported that functional activation of p53 in normal young HDFs and p53-null Saos2 cell line resulted in transcriptional activation of the IFI16 gene. We identified a potential p53 DNA-binding site (indicated as IFI16-p53-BS) in the 5'-regulatory region of the IFI16 gene. Importantly, p53 bound to IFI16-p53-BS in a sequence-specific manner in gel-mobility shift assays. Furthermore, p53 associated with the 5'-regulatory region of the IFI16 gene in chromatin immunoprecipitation assays. Interestingly, p53 associated with the regulatory region of the IFI16 gene only on treatment of cells with DNA-damaging agents or in the old, but not in the young, HDFs. Importantly, our promoter-reporter assays, which were coupled with site-directed mutagenesis of IFI16-p53-BS, showed that p53 activates transcription of the IFI16 gene in HDFs through the p53 DNA-binding site. Together, our observations provide support for the idea that up-regulation of IFI16 expression by p53 and functional interactions between IFI16 protein and p53 contribute to cellular senescence.


Subject(s)
Cellular Senescence , Fibroblasts/physiology , Gene Expression Regulation , Genes, p53 , Nuclear Proteins/genetics , Phosphoproteins/genetics , Tumor Suppressor Protein p53/metabolism , Binding Sites , Cell Line , Chromatin Immunoprecipitation/methods , Electrophoretic Mobility Shift Assay/methods , Humans , Interferons/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Regulatory Sequences, Nucleic Acid , Transcription, Genetic , Up-Regulation
14.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31687975

ABSTRACT

Accumulation of senescent cells is associated with the progression of pulmonary fibrosis, but mechanisms accounting for this linkage are not well understood. To explore this issue, we investigated whether a class of biologically active profibrotic lipids, the leukotrienes (LT), is part of the senescence-associated secretory phenotype. The analysis of conditioned medium (CM), lipid extracts, and gene expression of LT biosynthesis enzymes revealed that senescent cells secreted LT, regardless of the origin of the cells or the modality of senescence induction. The synthesis of LT was biphasic and followed by antifibrotic prostaglandin (PG) secretion. The LT-rich CM of senescent lung fibroblasts (IMR-90) induced profibrotic signaling in naive fibroblasts, which were abrogated by inhibitors of ALOX5, the principal enzyme in LT biosynthesis. The bleomycin-induced expression of genes encoding LT and PG synthases, level of cysteinyl LT in the bronchoalveolar lavage, and overall fibrosis were reduced upon senescent cell removal either in a genetic mouse model or after senolytic treatment. Quantification of ALOX5+ cells in lung explants obtained from idiopathic pulmonary fibrosis (IPF) patients indicated that half of these cells were also senescent (p16Ink4a+). Unlike human fibroblasts from unused donor lungs made senescent by irradiation, senescent IPF fibroblasts secreted LTs but failed to synthesize PGs. This study demonstrates for the first time to our knowledge that senescent cells secrete functional LTs, significantly contributing to the LT pool known to cause or exacerbate IPF.


Subject(s)
Cellular Senescence , Fibroblasts/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Leukotrienes/metabolism , Lung/pathology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Bleomycin/toxicity , Bronchoalveolar Lavage Fluid/chemistry , Cell Line , Culture Media, Conditioned/metabolism , Disease Models, Animal , Disease Progression , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Idiopathic Pulmonary Fibrosis/diagnosis , Leukotrienes/analysis , Lipoxygenase Inhibitors/pharmacology , Lung/cytology , Male , Mice , Primary Cell Culture , Prostaglandins/metabolism , Signal Transduction/drug effects
15.
Cell Rep ; 28(13): 3329-3337.e5, 2019 09 24.
Article in English | MEDLINE | ID: mdl-31553904

ABSTRACT

Cellular senescence irreversibly arrests cell proliferation, accompanied by a multi-component senescence-associated secretory phenotype (SASP) that participates in several age-related diseases. Using stable isotope labeling with amino acids (SILACs) and cultured cells, we identify 343 SASP proteins that senescent human fibroblasts secrete at 2-fold or higher levels compared with quiescent cell counterparts. Bioinformatic analysis reveals that 44 of these proteins participate in hemostasis, a process not previously linked with cellular senescence. We validated the expression of some of these SASP factors in cultured cells and in vivo. Mice treated with the chemotherapeutic agent doxorubicin, which induces widespread cellular senescence in vivo, show increased blood clotting. Conversely, selective removal of senescent cells using transgenic p16-3MR mice showed that clearing senescent cells attenuates the increased clotting caused by doxorubicin. Our study provides an in-depth, unbiased analysis of the SASP and unveils a function for cellular senescence in hemostasis.


Subject(s)
Cellular Senescence/genetics , Hemostasis , Humans
16.
Mol Cancer Res ; 5(3): 251-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17339605

ABSTRACT

Increased expression of IFI16 protein (encoded by the IFI16 gene) in normal human prostate epithelial cells is associated with cellular senescence-associated cell growth arrest. Consistent with a role for IFI16 protein in cellular senescence, the expression of IFI16 protein is either very low or not detectable in human prostate cancer cell lines. We now report that treatment of DU-145 and LNCaP prostate cancer cell lines with histone deacetylase inhibitor trichostatin A (TSA) or CGK1026 resulted in transcriptional activation of the IFI16 gene. The induction of IFI16 protein in LNCaP cells was dependent on the duration of TSA treatment. Furthermore, TSA treatment of LNCaP cells up-regulated the expression of Janus-activated kinase 1 protein kinase and modulated the transcription of certain IFN-activatable genes. However, overexpression of exogenous Janus-activated kinase 1 protein in LNCaP cells and treatment of cells with IFNs (alpha and gamma) did not increase the expression of IFI16. Instead, the transcriptional activation of IFI16 gene by TSA treatment of LNCaP cells was dependent on transcriptional activation by c-Jun/activator protein-1 transcription factor. Importantly, increased expression of IFI16 in LNCaP cells was associated with decreases in the expression of androgen receptor and apoptosis of cells. Conversely, knockdown of IFI16 expression in TSA-treated LNCaP cells increased androgen receptor protein levels with concomitant decreases in apoptosis. Together, our observations provide support for the idea that histone deacetylase-dependent transcriptional silencing of the IFI16 gene in prostate epithelial cells contributes to the development of prostate cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Gene Silencing , Histone Deacetylases/metabolism , Nuclear Proteins/genetics , Phosphoproteins/genetics , Prostatic Neoplasms/genetics , Cell Line, Tumor , Down-Regulation , Enzyme Inhibitors/pharmacology , Gene Silencing/drug effects , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Janus Kinase 1/metabolism , Male , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Prostatic Neoplasms/enzymology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Transcriptional Activation , Up-Regulation
17.
Sci Rep ; 8(1): 2410, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402901

ABSTRACT

Processes that have been linked to aging and cancer include an inflammatory milieu driven by senescent cells. Senescent cells lose the ability to divide, essentially irreversibly, and secrete numerous proteases, cytokines and growth factors, termed the senescence-associated secretory phenotype (SASP). Senescent cells that lack p53 tumor suppressor function show an exaggerated SASP, suggesting the SASP is negatively controlled by p53. Here, we show that increased p53 activity caused by small molecule inhibitors of MDM2, which promotes p53 degradation, reduces inflammatory cytokine production by senescent cells. Upon treatment with the MDM2 inhibitors nutlin-3a or MI-63, human cells acquired a senescence-like growth arrest, but the arrest was reversible. Importantly, the inhibitors reduced expression of the signature SASP factors IL-6 and IL-1α by cells made senescent by genotoxic stimuli, and suppressed the ability of senescent fibroblasts to stimulate breast cancer cell aggressiveness. Our findings suggest that MDM2 inhibitors could reduce cancer progression in part by reducing the pro-inflammatory environment created by senescent cells.


Subject(s)
Cellular Senescence/drug effects , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Indoles/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/genetics , Spiro Compounds/pharmacology , Tumor Suppressor Protein p53/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line , Cellular Senescence/genetics , Cellular Senescence/radiation effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/radiation effects , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Foreskin/cytology , Gamma Rays , Humans , Interleukin-1alpha/antagonists & inhibitors , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Interleukin-6/metabolism , Lung/cytology , Male , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/agonists , Tumor Suppressor Protein p53/metabolism
18.
Oncotarget ; 8(16): 27661-27672, 2017 Apr 18.
Article in English | MEDLINE | ID: mdl-28416761

ABSTRACT

Cellular senescence is defined as an irreversible growth arrest with the acquisition of a distinctive secretome. The growth arrest is a potent anticancer mechanism whereas the secretome facilitates wound healing, tissue repair, and development. The senescence response has also become increasingly recognized as an important contributor to aging and age-related diseases, including cancer. Although oncogenic mutations are capable of inducing a beneficial senescence response that prevents the growth of premalignant cells and promotes cancer immune-surveillance, the secretome of senescent cells also includes factors with pro-tumorigenic properties. On June 23rd and 24th, 2016, the Division of Cancer Biology of the National Cancer Institute sponsored a workshop to discuss the complex role of cellular senescence in tumorigenesis with the goal to define the major challenges and opportunities within this important field of cancer research. Additionally, it was noted how the development of novel tools and technologies are required to accelerate research into a mechanistic understanding of senescent cells in carcinogenesis in order to overcome the current limitations in this exciting, yet ill-defined area.


Subject(s)
Cell Transformation, Neoplastic , Cellular Senescence , Neoplasms/etiology , Neoplasms/metabolism , Aging , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Cellular Senescence/drug effects , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Humans , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/genetics
19.
Cancer Discov ; 7(2): 165-176, 2017 02.
Article in English | MEDLINE | ID: mdl-27979832

ABSTRACT

Cellular senescence suppresses cancer by irreversibly arresting cell proliferation. Senescent cells acquire a proinflammatory senescence-associated secretory phenotype. Many genotoxic chemotherapies target proliferating cells nonspecifically, often with adverse reactions. In accord with prior work, we show that several chemotherapeutic drugs induce senescence of primary murine and human cells. Using a transgenic mouse that permits tracking and eliminating senescent cells, we show that therapy-induced senescent (TIS) cells persist and contribute to local and systemic inflammation. Eliminating TIS cells reduced several short- and long-term effects of the drugs, including bone marrow suppression, cardiac dysfunction, cancer recurrence, and physical activity and strength. Consistent with our findings in mice, the risk of chemotherapy-induced fatigue was significantly greater in humans with increased expression of a senescence marker in T cells prior to chemotherapy. These findings suggest that senescent cells can cause certain chemotherapy side effects, providing a new target to reduce the toxicity of anticancer treatments. SIGNIFICANCE: Many genotoxic chemotherapies have debilitating side effects and also induce cellular senescence in normal tissues. The senescent cells remain chronically present where they can promote local and systemic inflammation that causes or exacerbates many side effects of the chemotherapy. Cancer Discov; 7(2); 165-76. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 115.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p16/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Female , Humans , Mice , Mice, Transgenic , Neoplasm Recurrence, Local
20.
FEBS Lett ; 580(6): 1659-64, 2006 Mar 06.
Article in English | MEDLINE | ID: mdl-16494870

ABSTRACT

Expression of androgen receptor (AR) in prostate epithelial cells is thought to regulate cell proliferation, differentiation, and survival. However, the molecular mechanisms remain unclear. We report that re-expression of AR in PC-3 human prostate cancer cell line resulted in upregulation of IFI16 protein, a negative regulator of cell growth. We found that the IFI16 protein bound to AR in a ligand-dependent manner and the DNA-binding domain (DBD) of the AR was sufficient to bind IFI16. Furthermore, re-expression of IFI16 protein in LNCaP prostate cancer cells, which do not express IFI16 protein, resulted in downregulation of AR expression and an inhibition of the expression of AR target genes. Our observations identify a role for IFI16 protein in AR-mediated functions.


Subject(s)
Feedback, Physiological , Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Humans , Male , Nuclear Proteins/genetics , Phosphoproteins/genetics , Prostatic Neoplasms/genetics , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL