151
|
Comprehensive network analysis of genes expressed in human oropharyngeal cancer. Am J Otolaryngol 2015; 36:235-41. [PMID: 25484365 DOI: 10.1016/j.amjoto.2014.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/13/2014] [Indexed: 01/02/2023]
Abstract
PURPOSE Oropharyngeal cancer (OPC) is the eighth most common cancer worldwide, however the genes involved in the development of OPC have been reported few. We constructed a co-expression network to extend knowledge of the molecular biomarkers in OPC development. MATERIALS AND METHODS Microarray data of HPV-active, -inactive, -negative OPC and normal benign tissue (uvula, tonsil) (Series GSE55550) were retrieved from NCBI GEO DataSets. We performed co-expression analysis of OPC transcriptome data by the Pearson correlation coefficient (PCC) method with the mutual rank (MR)-based cut-off using 13 guide genes. RESULTS The OPC subnetwork contained three clusters: cell cycle (62 node genes and 125 edge genes), immune system (44 node genes and 70 edge genes) and organ morphogenesis (128 node gene and 215 edge genes) process separately. CONCLUSION Our co-expression analysis includes separated transcriptomes of OPC, which is a useful resource for OPC researchers to elucidate important and complex biological events, to prevent and to predict cancer.
Collapse
|
152
|
Human C6orf211 encodes Armt1, a protein carboxyl methyltransferase that targets PCNA and is linked to the DNA damage response. Cell Rep 2015; 10:1288-96. [PMID: 25732820 DOI: 10.1016/j.celrep.2015.01.054] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 12/08/2014] [Accepted: 01/24/2015] [Indexed: 11/20/2022] Open
Abstract
Recent evidence supports the presence of an L-glutamyl methyltransferase(s) in eukaryotic cells, but this enzyme class has been defined only in certain prokaryotic species. Here, we characterize the human C6orf211 gene product as "acidic residue methyltransferase-1" (Armt1), an enzyme that specifically targets proliferating cell nuclear antigen (PCNA) in breast cancer cells, predominately methylating glutamate side chains. Armt1 homologs share structural similarities with the SAM-dependent methyltransferases, and negative regulation of activity by automethylation indicates a means for cellular control. Notably, shRNA-based knockdown of Armt1 expression in two breast cancer cell lines altered survival in response to genotoxic stress. Increased sensitivity to UV, adriamycin, and MMS was observed in SK-Br-3 cells, while in contrast, increased resistance to these agents was observed in MCF7 cells. Together, these results lay the foundation for defining the mechanism by which this post-translational modification operates in the DNA damage response (DDR).
Collapse
|
153
|
Atilgan R, Kuloğlu T, Boztosun A, Orak U, Baspinar M, Can B, Sapmaz E. Investigation of the effects of unilateral total salpingectomy on ovarian proliferating cell nuclear antigen and follicular reserve: experimental study. Eur J Obstet Gynecol Reprod Biol 2015; 188:56-60. [PMID: 25790915 DOI: 10.1016/j.ejogrb.2015.02.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Revised: 02/12/2015] [Accepted: 02/19/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVE We aimed to investigate the effects of unilateral total salpingectomy procedure on ovarian follicular reserve, apoptosis, and proliferating cell nuclear antigen (PCNA) staining in this study. STUDY DESIGN Fourteen female Wistar Albino rats of 12 weeks were randomly divided into two groups. Abdomen midline incision was conducted under general anesthesia. Group 1(G1) (n=7): Group in which only the abdomen was opened and closed, Group 2(G2) (n=7): Group that underwent right total salpingectomy. After 1 month, abdomens of all rats were opened. Ovaries were macroscopically evaluated. Right ovarian tissue was quickly removed, fixed in 10% formaldehyde, and paraffin blocks were prepared.' The existence of fibrosis was identified with the usage of light microscope. Follicles were microscopically classified and counted. The prevalence of cytoplasmic immune staining and TUNEL staining was scored semi-quantitatively. STATISTICAL ANALYSIS SPSS 17.0 software was used for the statistical analysis of data. First, Kruskal-Wallis variance analysis was conducted, and then Mann-Whitney U test was utilized for inter-group dual comparisons for parameters found as p<0.05. RESULTS While the number of CL was found out dramatically high, secondary follicle count was found out to be significantly low in G2. Also in G2, although the number of atretic follicle and fibrosis were found out significantly increased, and the score of the angiogenesis was found to be significantly decreased in CL. When compared PCNA immunoreactivity in granulosa cells with the control group, there was a significant decrease in G2. When compared the malondialdehyde (MDA) immunoreactivity with G1 a significant increase was established in G2. Apoptosis score of ovarian follicles in granulosa cells was significantly higher in G2. CONCLUSIONS In this experimental study, the decrease in the ovarian reserve and PCNA staning of granulosa cells, an increase in apoptosis, fibrosis and the number of atretic follicles in unilateral total salpingectomy operation were analyzed in rats. We found out significantly higher MDA staining rates in G2 in comparison to in G1. According to the study, the unilateral total salpingectomy procedure can damage to the same side ovarian tissue by means of the ischemia and reperfusion injury at the ovarian tissue.
Collapse
Affiliation(s)
- Remzi Atilgan
- Firat University School of Medicine, Department of Obstetrics and Gynecology, Elazig, Turkey.
| | - Tuncay Kuloğlu
- Firat University School of Medicine, Department of Histology, Elazig, Turkey
| | - Abdullah Boztosun
- Akdeniz University School of Medicine, Department of Obstetrics and Gynecology, Antalya, Turkey
| | - Uğur Orak
- Firat University School of Medicine, Department of Obstetrics and Gynecology, Elazig, Turkey
| | - Melike Baspinar
- Firat University School of Medicine, Department of Obstetrics and Gynecology, Elazig, Turkey
| | - Behzat Can
- Firat University School of Medicine, Department of Obstetrics and Gynecology, Elazig, Turkey
| | - Ekrem Sapmaz
- Firat University School of Medicine, Department of Obstetrics and Gynecology, Elazig, Turkey
| |
Collapse
|
154
|
Gerits N, Johannessen M, Tümmler C, Walquist M, Kostenko S, Snapkov I, van Loon B, Ferrari E, Hübscher U, Moens U. Agnoprotein of polyomavirus BK interacts with proliferating cell nuclear antigen and inhibits DNA replication. Virol J 2015; 12:7. [PMID: 25638270 PMCID: PMC4318453 DOI: 10.1186/s12985-014-0220-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 12/01/2014] [Indexed: 12/25/2022] Open
Abstract
Background The human polyomavirus BK expresses a 66 amino-acid peptide referred to as agnoprotein. Though mutants lacking agnoprotein are severely reduced in producing infectious virions, the exact function of this peptide remains incompletely understood. To elucidate the function of agnoprotein, we searched for novel cellular interaction partners. Methods Yeast-two hybrid assay was performed with agnoprotein as bait against human kidney and thymus libraries. The interaction between agnoprotein and putative partners was further examined by GST pull down, co-immunoprecipitation, and fluorescence resonance energy transfer studies. Biochemical and biological studies were performed to examine the functional implication of the interaction of agnoprotein with cellular target proteins. Results Proliferating cell nuclear antigen (PCNA), which acts as a processivity factor for DNA polymerase δ, was identified as an interaction partner. The interaction between agnoprotein and PCNA is direct and occurs also in human cells. Agnoprotein exerts an inhibitory effect on PCNA-dependent DNA synthesis in vitro and reduces cell proliferation when ectopically expressed. Overexpression of PCNA restores agnoprotein-mediated inhibition of cell proliferation. Conclusion Our data suggest that PCNA is a genuine interaction partner of agnoprotein and the inhibitory effect on PCNA-dependent DNA synthesis by the agnoprotein may play a role in switching off (viral) DNA replication late in the viral replication cycle when assembly of replicated genomes and synthesized viral capsid proteins occurs. Electronic supplementary material The online version of this article (doi:10.1186/s12985-014-0220-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ugo Moens
- UiT - The Arctic University of Norway, Faculty of Health Sciences, Department of Medical Biology, Molecular Inflammation Research Group, Tromsø NO-9037, Norway.
| |
Collapse
|
155
|
Pawar S, Mahantshetty U, Deodhar K, Teni T. A simple cost-effective modification improves the quality of immunocytochemical staining in cervical scrape samples characterized by presence of excess mucus. J Histotechnol 2015; 37:3-8. [PMID: 25620823 PMCID: PMC4304270 DOI: 10.1179/2046023613y.0000000034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immunocytochemistry (ICC) is a very important tool in a diverse range of biomedical research as well as in diagnostic cytopathology. Smears prepared from cervical scrapes contain a large amount of overlying mucus that interferes with the standard immunocytochemical staining protocol. A modified ICC protocol is described, which involves pretreatment of these smears with 1 mg/ml solution of Ambroxol hydrochloride in methanol for 1 hour. Source of Ambroxol hydrochloride was a 30 mg Mucolite™ tablet, at a cost of 1.70 rupees (∼3·5 US cents) per tablet. This mucolytic solution effectively clears the mucus, facilitating the accessibility of the antibody to the antigenic determinants. This pretreatment resulted in the increased percentage of positively stained cells as well as staining intensity, leading to improved overall ICC staining and score. This is a novel modification that can be cost-effectively applied in ICC staining protocols for cytology samples characterized by the presence of excess mucus.
Collapse
Affiliation(s)
- Sagar Pawar
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Umesh Mahantshetty
- Department of Radiation Oncology and Medical Physics, Tata Memorial Hospital, Parel, Mumbai, India
| | - Kedar Deodhar
- Department of Pathology, Tata Memorial Hospital, Parel, Mumbai, India
| | - Tanuja Teni
- Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
| |
Collapse
|
156
|
Pannexin‑1 silencing inhibits the proliferation of U87‑MG cells. Mol Med Rep 2015; 11:3487-92. [PMID: 25572468 DOI: 10.3892/mmr.2015.3169] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Accepted: 07/23/2014] [Indexed: 11/05/2022] Open
Abstract
Pannexin‑1 (Panx‑1) is abundantly expressed in vertebrates and has been shown to assemble into high‑conductance single‑membrane channels, which are permeable to large molecules and regulate cellular function. However, the association between Panx‑1 and astrocyte proliferation is poorly understood. This study provides evidence for a difference in cell proliferation between wild‑type and Panx‑1‑knockdown cells. Proliferation of the U87‑MG malignant glioma cell line was reduced following transfection with Panx‑1‑short interfering RNA. In addition, treatment with the Panx‑1 activator, adenosine triphosphate, significantly reduced cell proliferation at 48 h in Panx‑1‑knockdown cells compared with wild type cells. In conclusion, on the basis of the present findings, Panx‑1 is likely to be important in the regulation of U87‑MG cell proliferation. This provides further support for the hypothesis that there is a correlation between Panx‑1 expression and U87‑MG cell proliferation.
Collapse
|
157
|
Liu W, Yue Y, Li Y, Zheng X, Zhang K, Du Z. Inspired by magnolol: design of NSAID-based compounds with excellent anti-inflammatory effects. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00308c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A10was selected to elucidate the anti-inflammatory mechanism at the transcriptional level, suggesting its potential to serve as a novel anti-inflammatory agent.
Collapse
Affiliation(s)
- Wenfeng Liu
- Laboratory of Natural Medicinal Chemistry & Green Chemistry
- Faculty of Light Industry and Chemical Engineering
- Guangdong University of Technology
- Guangzhou
- China
| | - Yuan Yue
- Laboratory of Natural Medicinal Chemistry & Green Chemistry
- Faculty of Light Industry and Chemical Engineering
- Guangdong University of Technology
- Guangzhou
- China
| | - Yonglian Li
- Guangdong Industry Technical College
- Guangzhou
- China
| | - Xi Zheng
- Laboratory of Natural Medicinal Chemistry & Green Chemistry
- Faculty of Light Industry and Chemical Engineering
- Guangdong University of Technology
- Guangzhou
- China
| | - Kun Zhang
- Laboratory of Natural Medicinal Chemistry & Green Chemistry
- Faculty of Light Industry and Chemical Engineering
- Guangdong University of Technology
- Guangzhou
- China
| | - Zhiyun Du
- Laboratory of Natural Medicinal Chemistry & Green Chemistry
- Faculty of Light Industry and Chemical Engineering
- Guangdong University of Technology
- Guangzhou
- China
| |
Collapse
|
158
|
Smith KP, Gifford KM, Waitzman JS, Rice SE. Survey of phosphorylation near drug binding sites in the Protein Data Bank (PDB) and their effects. Proteins 2015; 83:25-36. [PMID: 24833420 PMCID: PMC4233198 DOI: 10.1002/prot.24605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/28/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
While it is currently estimated that 40 to 50% of eukaryotic proteins are phosphorylated, little is known about the frequency and local effects of phosphorylation near pharmaceutical inhibitor binding sites. In this study, we investigated how frequently phosphorylation may affect the binding of drug inhibitors to target proteins. We examined the 453 non-redundant structures of soluble mammalian drug target proteins bound to inhibitors currently available in the Protein Data Bank (PDB). We cross-referenced these structures with phosphorylation data available from the PhosphoSitePlus database. Three hundred twenty-two of 453 (71%) of drug targets have evidence of phosphorylation that has been validated by multiple methods or labs. For 132 of 453 (29%) of those, the phosphorylation site is within 12 Å of the small molecule-binding site, where it would likely alter small molecule binding affinity. We propose a framework for distinguishing between drug-phosphorylation site interactions that are likely to alter the efficacy of drugs versus those that are not. In addition we highlight examples of well-established drug targets, such as estrogen receptor alpha, for which phosphorylation may affect drug affinity and clinical efficacy. Our data suggest that phosphorylation may affect drug binding and efficacy for a significant fraction of drug target proteins.
Collapse
Affiliation(s)
- Kyle P Smith
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611
| | | | | | | |
Collapse
|
159
|
Activation of oxidative stress and inflammatory factors could account for histopathological progression of aflatoxin-B1 induced hepatocarcinogenesis in rat. Mol Cell Biochem 2014; 401:185-96. [DOI: 10.1007/s11010-014-2306-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 12/20/2014] [Indexed: 12/27/2022]
|
160
|
Gederaas OA, Søgaard CD, Viset T, Bachke S, Bruheim P, Arum CJ, Otterlei M. Increased Anticancer Efficacy of Intravesical Mitomycin C Therapy when Combined with a PCNA Targeting Peptide. Transl Oncol 2014; 7:812-23. [PMID: 25500092 PMCID: PMC4311026 DOI: 10.1016/j.tranon.2014.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/13/2014] [Accepted: 10/13/2014] [Indexed: 12/21/2022] Open
Abstract
Non-muscle-invasive bladder cancers (NMIBCs) are tumors confined to the mucosa or the mucosa/submucosa. An important challenge in treatment of NMIBC is both high recurrence and high progression rates. Consequently, more efficacious intravesical treatment regimes are in demand. Inhibition of the cell's DNA repair systems is a new promising strategy to improve cancer therapy, and proliferating cell nuclear antigen (PCNA) is a new promising target. PCNA is an essential scaffold protein in multiple cellular processes including DNA replication and repair. More than 200 proteins, many involved in stress responses, interact with PCNA through the AlkB homologue 2 PCNA-interacting motif (APIM), including several proteins directly or indirectly involved in repair of DNA interstrand crosslinks (ICLs). In this study, we targeted PCNA with a novel peptide drug containing the APIM sequence, ATX-101, to inhibit repair of the DNA damage introduced by the chemotherapeutics. A bladder cancer cell panel and two different orthotopic models of bladder cancer in rats, the AY-27 implantation model and the dietary BBN induction model, were applied. ATX-101 increased the anticancer efficacy of the ICL-inducing drug mitomycin C (MMC), as well as bleomycin and gemcitabine in all bladder cancer cell lines tested. Furthermore, we found that ATX-101 given intravesically in combination with MMC penetrated the bladder wall and further reduced the tumor growth in both the slow growing endogenously induced and the rapidly growing transplanted tumors. These results suggest that ATX-101 has the potential to improve the efficacy of current MMC treatment in NMIBC.
Collapse
Affiliation(s)
- Odrun A Gederaas
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Caroline D Søgaard
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Trond Viset
- Department of Pathology and Medical Genetics, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Siri Bachke
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Per Bruheim
- Department of Biotechnology, Norwegian University of Science and Technology, Trondheim Norway
| | - Carl-Jørgen Arum
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Urology and Surgery, St Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Marit Otterlei
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; APIM Therapeutics A/S, Trondheim, Norway.
| |
Collapse
|
161
|
Dennis CV, Sheahan PJ, Graeber MB, Sheedy DL, Kril JJ, Sutherland GT. Microglial proliferation in the brain of chronic alcoholics with hepatic encephalopathy. Metab Brain Dis 2014; 29:1027-39. [PMID: 24346482 PMCID: PMC4063896 DOI: 10.1007/s11011-013-9469-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 12/04/2013] [Indexed: 12/11/2022]
Abstract
Hepatic encephalopathy (HE) is a common complication of chronic alcoholism and patients show neurological symptoms ranging from mild cognitive dysfunction to coma and death. The HE brain is characterized by glial changes, including microglial activation, but the exact pathogenesis of HE is poorly understood. During a study investigating cell proliferation in the subventricular zone of chronic alcoholics, a single case with widespread proliferation throughout their adjacent grey and white matter was noted. This case also had concomitant HE raising the possibility that glial proliferation might be a pathological feature of the disease. In order to explore this possibility fixed postmortem human brain tissue from chronic alcoholics with cirrhosis and HE (n = 9), alcoholics without HE (n = 4) and controls (n = 4) were examined using immunohistochemistry and cytokine assays. In total, 4/9 HE cases had PCNA- and a second proliferative marker, Ki-67-positive cells throughout their brain and these cells co-stained with the microglial marker, Iba1. These cases were termed 'proliferative HE' (pHE). The microglia in pHEs displayed an activated morphology with hypertrophied cell bodies and short, thickened processes. In contrast, the microglia in white matter regions of the non-proliferative HE cases were less activated and appeared dystrophic. pHEs were also characterized by higher interleukin-6 levels and a slightly higher neuronal density . These findings suggest that microglial proliferation may form part of an early neuroprotective response in HE that ultimately fails to halt the course of the disease because underlying etiological factors such as high cerebral ammonia and systemic inflammation remain.
Collapse
Affiliation(s)
- Claude V Dennis
- Discipline of Pathology, Sydney Medical School, Camperdown, NSW, 2050, Australia
| | | | | | | | | | | |
Collapse
|
162
|
Hendry WJ, Hariri HY, Alwis ID, Gunewardena SS, Hendry IR. Altered gene expression patterns during the initiation and promotion stages of neonatally diethylstilbestrol-induced hyperplasia/dysplasia/neoplasia in the hamster uterus. Reprod Toxicol 2014; 50:68-86. [PMID: 25242112 DOI: 10.1016/j.reprotox.2014.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Revised: 08/18/2014] [Accepted: 09/08/2014] [Indexed: 10/24/2022]
Abstract
Neonatal treatment of hamsters with diethylstilbestrol (DES) induces uterine hyperplasia/dysplasia/neoplasia (endometrial adenocarcinoma) in adult animals. We subsequently determined that the neonatal DES exposure event directly and permanently disrupts the developing hamster uterus (initiation stage) so that it responds abnormally when it is stimulated with estrogen in adulthood (promotion stage). To identify candidate molecular elements involved in progression of the disruption/neoplastic process, we performed: (1) immunoblot analyses and (2) microarray profiling (Affymetrix Gene Chip System) on sets of uterine protein and RNA extracts, respectively, and (3) immunohistochemical analysis on uterine sections; all from both initiation stage and promotion stage groups of animals. Here we report that: (1) progression of the neonatal DES-induced hyperplasia/dysplasia/neoplasia phenomenon in the hamster uterus involves a wide spectrum of specific gene expression alterations and (2) the gene products involved and their manner of altered expression differ dramatically during the initiation vs. promotion stages of the phenomenon.
Collapse
Affiliation(s)
- William J Hendry
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260-0026, United States.
| | - Hussam Y Hariri
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260-0026, United States
| | - Imala D Alwis
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260-0026, United States
| | - Sumedha S Gunewardena
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, United States; Bioinformatics Core, University of Kansas Medical Center, Kansas City, KS 66160, United States
| | - Isabel R Hendry
- Department of Biological Sciences, Wichita State University, Wichita, KS 67260-0026, United States
| |
Collapse
|
163
|
Genome-wide profiling reveals a role for T-cell intracellular antigens TIA1 and TIAR in the control of translational specificity in HeLa cells. Biochem J 2014; 461:43-50. [PMID: 24927121 DOI: 10.1042/bj20140227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
TIA (T-cell intracellular antigens)-knockdown HeLa cells show an increase in ribosomes and translational machinery components. This increase correlates with specific changes in translationally up-regulated mRNAs involved in cell-cycle progression and DNA repair, as shown in polysomal profiling analysis. Our data support the hypothesis that a concerted activation of both global and selective translational rates leads to the transition to a more proliferative status in TIA-knockdown HeLa cells.
Collapse
|
164
|
Pessina P, Castillo V, Sartore I, Borrego J, Meikle A. Semiquantitative immunohistochemical marker staining and localization in canine thyroid carcinoma and normal thyroid gland. Vet Comp Oncol 2014; 14:e102-12. [DOI: 10.1111/vco.12111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Accepted: 06/22/2014] [Indexed: 12/27/2022]
Affiliation(s)
- P. Pessina
- Laboratorio de Técnicas Nucleares; Facultad de Veterinaria; Montevideo Uruguay
| | - V. Castillo
- Cátedra Clínica Médica Pequeños Animales, U. Endocrinología, Htal. Escuela de Medicina Veterinaria, Facultad de Ciencias Veterinarias; Universidad de Buenos Aires; Buenos Aires Argentina
| | - I. Sartore
- Laboratorio de Técnicas Nucleares; Facultad de Veterinaria; Montevideo Uruguay
| | - J. Borrego
- Instituto Veterinario de Oncología Comparada (IVOC); Valencia Spain
- Hospital Veterinario Facultad de Veterinaria y Ciencias Experimentales; Universidad Católica de Valencia “San Vicente Martir”; Valencia Spain
| | - A. Meikle
- Laboratorio de Técnicas Nucleares; Facultad de Veterinaria; Montevideo Uruguay
| |
Collapse
|
165
|
Pedley AM, Lill MA, Davisson VJ. Flexibility of PCNA-protein interface accommodates differential binding partners. PLoS One 2014; 9:e102481. [PMID: 25036435 PMCID: PMC4103810 DOI: 10.1371/journal.pone.0102481] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 06/19/2014] [Indexed: 11/18/2022] Open
Abstract
The expanding roles of PCNA in functional assembly of DNA replication and repair complexes motivated investigation of the structural and dynamic properties guiding specificity of PCNA-protein interactions. A series of biochemical and computational analyses were combined to evaluate the PIP Box recognition features impacting complex formation. The results indicate subtle differences in topological and molecular descriptors distinguishing both affinity and stoichiometry of binding among PCNA-peptide complexes through cooperative effects. These features were validated using peptide mimics of p85α and Akt, two previously unreported PCNA binding partners. This study characterizes for the first time a reverse PIP Box interaction with PCNA. Small molecule ligand binding at the PIP Box interaction site confirmed the adaptive nature of the protein in dictating overall shape and implicates allosterism in transmitting biological effects.
Collapse
Affiliation(s)
- Anthony M. Pedley
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| | - Markus A. Lill
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
| | - V. Jo Davisson
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana, United States of America
- * E-mail:
| |
Collapse
|
166
|
Rajcevic U, Knol JC, Piersma S, Bougnaud S, Fack F, Sundlisaeter E, Søndenaa K, Myklebust R, Pham TV, Niclou SP, Jiménez CR. Colorectal cancer derived organotypic spheroids maintain essential tissue characteristics but adapt their metabolism in culture. Proteome Sci 2014; 12:39. [PMID: 25075203 PMCID: PMC4114130 DOI: 10.1186/1477-5956-12-39] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 06/09/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Organotypic tumor spheroids, a 3D in vitro model derived from patient tumor material, preserve tissue heterogeneity and retain structural tissue elements, thus replicating the in vivo tumor more closely than commonly used 2D and 3D cell line models. Such structures harbour tumorigenic cells, as revealed by xenograft implantation studies in animal models and maintain the genetic makeup of the original tumor material. The aim of our work was a morphological and proteomic characterization of organotypic spheroids derived from colorectal cancer tissue in order to get insight into their composition and associated biology. RESULTS Morphological analysis showed that spheroids were of about 250 μm in size and varied in structure, while the spheroid cells differed in shape and size and were tightly packed together by desmosomes and tight junctions. Our proteomic data revealed significant alterations in protein expression in organotypic tumor spheroids cultured as primary explants compared to primary colorectal cancer tissue. Components underlying cellular and tissue architecture were changed; nuclear DNA/ chromatin maintenance systems were up-regulated, whereas various mitochondrial components were down-regulated in spheroids. Most interestingly, the mesenchymal cells appear to be substantial component in such cellular assemblies. Thus the observed changes may partly occur in this cellular compartment. Finally, in the proteomics analysis stem cell-like characteristics were observed within the spheroid cellular assembly, reflected by accumulation of Alcam, Ctnnb1, Aldh1, Gpx2, and CD166. These findings were underlined by IHC analysis of Ctnnb1, CD24 and CD44, therefore warranting closer investigation of the tumorigenic compartment in this 3D culture model for tumor tissue. CONCLUSIONS Our analysis of organotypic CRC tumor spheroids has identified biological processes associated with a mixture of cell types and states, including protein markers for mesenchymal and stem-like cells. This 3D tumor model in which tumor heterogeneity is preserved may represent an advantageous model system to investigate novel therapeutic approaches.
Collapse
Affiliation(s)
- Uros Rajcevic
- NorLux Neuro-Oncology Laboratory, Department of Oncology, CRP-Santé, Luxembourg, Luxembourg ; Department of Research and Development, Blood Transfusion Center of Slovenia, Ljubljana, Slovenia ; Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Jaco C Knol
- OncoProteomics Laboratory, Department of Medical Oncology, VUmc-Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Sander Piersma
- OncoProteomics Laboratory, Department of Medical Oncology, VUmc-Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Sébastien Bougnaud
- NorLux Neuro-Oncology Laboratory, Department of Oncology, CRP-Santé, Luxembourg, Luxembourg
| | - Fred Fack
- NorLux Neuro-Oncology Laboratory, Department of Oncology, CRP-Santé, Luxembourg, Luxembourg
| | | | - Karl Søndenaa
- Department of Surgery, Haraldsplass Deaconal Hospital, University of Bergen, Bergen, Norway
| | | | - Thang V Pham
- OncoProteomics Laboratory, Department of Medical Oncology, VUmc-Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| | - Simone P Niclou
- NorLux Neuro-Oncology Laboratory, Department of Oncology, CRP-Santé, Luxembourg, Luxembourg
| | - Connie R Jiménez
- OncoProteomics Laboratory, Department of Medical Oncology, VUmc-Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
167
|
Dietlein F, Thelen L, Reinhardt HC. Cancer-specific defects in DNA repair pathways as targets for personalized therapeutic approaches. Trends Genet 2014; 30:326-39. [PMID: 25017190 DOI: 10.1016/j.tig.2014.06.003] [Citation(s) in RCA: 204] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/12/2014] [Accepted: 06/18/2014] [Indexed: 12/13/2022]
Abstract
Defects in DNA repair pathways enable cancer cells to accumulate genomic alterations that contribute to their aggressive phenotype. However, tumors rely on residual DNA repair capacities to survive the damage induced by genotoxic stress. This dichotomy might explain why only isolated DNA repair pathways are inactivated in cancer cells. Accordingly, synergism has been observed between DNA-damaging drugs and targeted inhibitors of DNA repair. DNA repair pathways are generally thought of as mutually exclusive mechanistic units handling different types of lesions in distinct cell cycle phases. Recent preclinical studies, however, provide strong evidence that multifunctional DNA repair hubs, which are involved in multiple conventional DNA repair pathways, are frequently altered in cancer. We therefore propose that targeted anticancer therapies should not only exploit synthetic lethal interactions between two single genes but also consider alterations in DNA repair hubs. Such a network-based approach considerably increases the opportunities for targeting DNA repair-defective tumors.
Collapse
Affiliation(s)
- Felix Dietlein
- Department of Internal Medicine, University Hospital of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany.
| | - Lisa Thelen
- Department of Internal Medicine, University Hospital of Cologne, 50931 Cologne, Germany
| | - H Christian Reinhardt
- Department of Internal Medicine, University Hospital of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases, University of Cologne, 50674 Cologne, Germany.
| |
Collapse
|
168
|
Freund G, Desplancq D, Stoessel A, Weinsanto R, Sibler AP, Robin G, Martineau P, Didier P, Wagner J, Weiss E. Generation of an intrabody-based reagent suitable for imaging endogenous proliferating cell nuclear antigen in living cancer cells. J Mol Recognit 2014; 27:549-58. [DOI: 10.1002/jmr.2378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/01/2014] [Accepted: 04/01/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Guillaume Freund
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| | - Dominique Desplancq
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| | - Audrey Stoessel
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| | - Robin Weinsanto
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| | - Annie-Paule Sibler
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| | - Gautier Robin
- Institut de Recherche en Cancérologie de Montpellier, U896; INSERM/Université Montpellier 1; Campus Val d'Aurelle Montpellier France
| | - Pierre Martineau
- Institut de Recherche en Cancérologie de Montpellier, U896; INSERM/Université Montpellier 1; Campus Val d'Aurelle Montpellier France
| | - Pascal Didier
- Faculté de Pharmacie, UMR 7213; CNRS/Université de Strasbourg; Route du Rhin Illkirch France
| | - Jérôme Wagner
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| | - Etienne Weiss
- Ecole Supérieure de Biotechnologie de Strasbourg, UMR 7242; CNRS/Université de Strasbourg; Boulevard Sébastien Brant Illkirch France
| |
Collapse
|
169
|
Zhong B, Wang T, Lun X, Zhang J, Zheng S, Yang W, Li W, Xiang AP, Chen Z. Contribution of nestin positive esophageal squamous cancer cells on malignant proliferation, apoptosis, and poor prognosis. Cancer Cell Int 2014; 14:57. [PMID: 24966803 PMCID: PMC4071021 DOI: 10.1186/1475-2867-14-57] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Accepted: 06/16/2014] [Indexed: 02/05/2023] Open
Abstract
Background The stem cell-associated intermediate filament nestin has recently been linked with neoplastic transformation, but the specific mechanism by which nestin positive tumor cells leads to malignant invasion and metastasis behaviors of esophageal squamous cell carcinoma (ESCC) remains unclear. Methods To obtain insight into the biological role of nestin in ESCC, we explored the association of the nestin phenotype with malignant proliferation and apoptosis in esophageal squamous cancer cells. Nestin expression was determined in ESCC specimens and cell lines, and correlated with clinicopathological properties, including clinical prognosis and proliferative markers. The association of the nestin phenotype with apoptotic indicators was also analyzed. Results Nestin was expressed in ESCC specimens and cell lines. ESCC patients with nestin-positive tumors had significantly shorter median survival and progression-free survival times than those with nestin-negative tumors. Positive staining for the proliferation markers Ki67 and PCNA (proliferating cell nuclear antigen) was detected in 56.9% and 60.2% of ESCC specimens, respectively, and was strongly correlated with the nestin phenotype. Notably, expression of cyclin dependent kinase-5 (CDK5) and P35 was detected in 53.8% and 48.4% of ESCC specimens, respectively, and was strongly associated with the nestin phenotype. Conclusion Our data demonstrated nestin expression in ESCC specimens and cell lines, and revealed a strong association of the nestin phenotype with poor prognosis in ESCC patients. Furthermore, we showed that nestin positive ESCC cells played an important role in the malignant proliferation and apoptosis.
Collapse
Affiliation(s)
- Beilong Zhong
- Department of Thoracic Surgery, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong 519000, China
| | - Tao Wang
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Guangzhou, Guangdong, China.,Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xueping Lun
- Department of Thoracic Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,Lung Cancer Research Center of Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,Department of Cardiothoracic Surgery of East Division, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Jinli Zhang
- Guangzhou Research Institute of Traumatic Surgery, the Fourth Affiliated Hospital, Ji'nan University, Guangzhou, Guangdong 510220, China
| | - Sannv Zheng
- Department of Anesthesiology and Operating Room of East Division, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Weilin Yang
- Department of Thoracic Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,Lung Cancer Research Center of Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,Department of Cardiothoracic Surgery of East Division, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Guangzhou, Guangdong, China.,Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Sun Yat-sen University, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Guangzhou, Guangdong, China.,Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhenguang Chen
- Department of Thoracic Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,Lung Cancer Research Center of Sun Yat-sen University, Guangzhou, Guangdong 510080, China.,Department of Cardiothoracic Surgery of East Division, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
170
|
Cazzalini O, Sommatis S, Tillhon M, Dutto I, Bachi A, Rapp A, Nardo T, Scovassi AI, Necchi D, Cardoso MC, Stivala LA, Prosperi E. CBP and p300 acetylate PCNA to link its degradation with nucleotide excision repair synthesis. Nucleic Acids Res 2014; 42:8433-48. [PMID: 24939902 PMCID: PMC4117764 DOI: 10.1093/nar/gku533] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The proliferating cell nuclear antigen (PCNA) protein serves as a molecular platform recruiting and coordinating the activity of factors involved in multiple deoxyribonucleic acid (DNA) transactions. To avoid dangerous genome instability, it is necessary to prevent excessive retention of PCNA on chromatin. Although PCNA functions during DNA replication appear to be regulated by different post-translational modifications, the mechanism regulating PCNA removal and degradation after nucleotide excision repair (NER) is unknown. Here we report that CREB-binding protein (CBP), and less efficiently p300, acetylated PCNA at lysine (Lys) residues Lys13,14,77 and 80, to promote removal of chromatin-bound PCNA and its degradation during NER. Mutation of these residues resulted in impaired DNA replication and repair, enhanced the sensitivity to ultraviolet radiation, and prevented proteolytic degradation of PCNA after DNA damage. Depletion of both CBP and p300, or failure to load PCNA on DNA in NER deficient cells, prevented PCNA acetylation and degradation, while proteasome inhibition resulted in accumulation of acetylated PCNA. These results define a CBP and p300-dependent mechanism for PCNA acetylation after DNA damage, linking DNA repair synthesis with removal of chromatin-bound PCNA and its degradation, to ensure genome stability.
Collapse
Affiliation(s)
- Ornella Cazzalini
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Sabrina Sommatis
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Micol Tillhon
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - Ilaria Dutto
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - Angela Bachi
- IFOM-FIRC Institute of Molecular Oncology, Milan 20100, Italy
| | - Alexander Rapp
- Technische Universität Darmstadt, Darmstadt 64287, Germany
| | - Tiziana Nardo
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - A Ivana Scovassi
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| | - Daniela Necchi
- Department of Drug Sciences, University of Pavia, Pavia 27100, Italy
| | | | - Lucia A Stivala
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Ennio Prosperi
- Institute of Molecular Genetics, National Research Council (CNR), Pavia 27100, Italy
| |
Collapse
|
171
|
Fattal I, Rimer J, Shental N, Molad Y, Gabrielli A, Livneh A, Sarig O, Goldberg I, Gafter U, Domany E, Cohen IR. Pemphigus vulgaris is characterized by low IgG reactivities to specific self-antigens along with high IgG reactivity to desmoglein 3. Immunology 2014; 143:374-80. [PMID: 24820664 DOI: 10.1111/imm.12316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 04/14/2014] [Accepted: 05/01/2014] [Indexed: 11/28/2022] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune skin disease, which has been characterized by IgG autoantibodies to desmoglein 3. Here we studied the antibody signatures of PV patients compared with healthy subjects and with patients with two other autoimmune diseases with skin manifestations (systemic lupus erythematosus and scleroderma), using an antigen microarray and informatics analysis. We now report a previously unobserved phenomenon--patients with PV, compared with the healthy subjects and the two other diseases, show a significant decrease in IgG autoantibodies to a specific set of self-antigens. This novel finding demonstrates that an autoimmune disease may be associated with a loss of specific, healthy IgG autoantibodies and not only with a gain of specific, pathogenic IgG autoantibodies.
Collapse
Affiliation(s)
- Ittai Fattal
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel; Department of Physics of Complex Systems, The Weizmann Institute of Science, Rehovot, Israel; Department of Nephrology, Rabin Medical Centre, Petach Tikva, Israel
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
172
|
The cutting edges in DNA repair, licensing, and fidelity: DNA and RNA repair nucleases sculpt DNA to measure twice, cut once. DNA Repair (Amst) 2014; 19:95-107. [PMID: 24754999 DOI: 10.1016/j.dnarep.2014.03.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
To avoid genome instability, DNA repair nucleases must precisely target the correct damaged substrate before they are licensed to incise. Damage identification is a challenge for all DNA damage response proteins, but especially for nucleases that cut the DNA and necessarily create a cleaved DNA repair intermediate, likely more toxic than the initial damage. How do these enzymes achieve exquisite specificity without specific sequence recognition or, in some cases, without a non-canonical DNA nucleotide? Combined structural, biochemical, and biological analyses of repair nucleases are revealing their molecular tools for damage verification and safeguarding against inadvertent incision. Surprisingly, these enzymes also often act on RNA, which deserves more attention. Here, we review protein-DNA structures for nucleases involved in replication, base excision repair, mismatch repair, double strand break repair (DSBR), and telomere maintenance: apurinic/apyrimidinic endonuclease 1 (APE1), Endonuclease IV (Nfo), tyrosyl DNA phosphodiesterase (TDP2), UV Damage endonuclease (UVDE), very short patch repair endonuclease (Vsr), Endonuclease V (Nfi), Flap endonuclease 1 (FEN1), exonuclease 1 (Exo1), RNase T and Meiotic recombination 11 (Mre11). DNA and RNA structure-sensing nucleases are essential to life with roles in DNA replication, repair, and transcription. Increasingly these enzymes are employed as advanced tools for synthetic biology and as targets for cancer prognosis and interventions. Currently their structural biology is most fully illuminated for DNA repair, which is also essential to life. How DNA repair enzymes maintain genome fidelity is one of the DNA double helix secrets missed by James Watson and Francis Crick, that is only now being illuminated though structural biology and mutational analyses. Structures reveal motifs for repair nucleases and mechanisms whereby these enzymes follow the old carpenter adage: measure twice, cut once. Furthermore, to measure twice these nucleases act as molecular level transformers that typically reshape the DNA and sometimes themselves to achieve extraordinary specificity and efficiency.
Collapse
|
173
|
Li Z, Huang RYC, Yopp DC, Hileman TH, Santangelo TJ, Hurwitz J, Hudgens JW, Kelman Z. A novel mechanism for regulating the activity of proliferating cell nuclear antigen by a small protein. Nucleic Acids Res 2014; 42:5776-89. [PMID: 24728986 PMCID: PMC4027161 DOI: 10.1093/nar/gku239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) forms a trimeric ring that associates with and influences the activity of many proteins participating in DNA metabolic processes and cell cycle progression. Previously, an uncharacterized small protein, encoded by TK0808 in the archaeon Thermococcus kodakarensis, was shown to stably interact with PCNA in vivo. Here, we show that this protein, designated Thermococcales inhibitor of PCNA (TIP), binds to PCNA in vitro and inhibits PCNA-dependent activities likely by preventing PCNA trimerization. Using hydrogen/deuterium exchange mass spectrometry and site-directed mutagenesis, the interacting regions of PCNA and TIP were identified. Most proteins bind to PCNA via a PCNA-interacting peptide (PIP) motif that interacts with the inter domain connecting loop (IDCL) on PCNA. TIP, however, lacks any known PCNA-interacting motif, suggesting a new mechanism for PCNA binding and regulation of PCNA-dependent activities, which may support the development of a new subclass of therapeutic biomolecules for inhibiting PCNA.
Collapse
Affiliation(s)
- Zhuo Li
- Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD 20850, USA
| | - Richard Y-C Huang
- Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD 20850, USA National Institute of Standards and Technology, 9600 Gudelsky Drive, Rockville, MD 20850, USA
| | - Daniel C Yopp
- Department of Microbiology and Center for RNA Biology, Ohio State University, Columbus, OH 43210, USA
| | - Travis H Hileman
- Department of Microbiology and Center for RNA Biology, Ohio State University, Columbus, OH 43210, USA
| | - Thomas J Santangelo
- Department of Microbiology and Center for RNA Biology, Ohio State University, Columbus, OH 43210, USA
| | - Jerard Hurwitz
- Program of Molecular Biology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Jeffrey W Hudgens
- Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD 20850, USA National Institute of Standards and Technology, 9600 Gudelsky Drive, Rockville, MD 20850, USA
| | - Zvi Kelman
- Institute for Bioscience and Biotechnology Research, 9600 Gudelsky Drive, Rockville, MD 20850, USA National Institute of Standards and Technology, 9600 Gudelsky Drive, Rockville, MD 20850, USA
| |
Collapse
|
174
|
A PCNA-derived cell permeable peptide selectively inhibits neuroblastoma cell growth. PLoS One 2014; 9:e94773. [PMID: 24728180 PMCID: PMC3984256 DOI: 10.1371/journal.pone.0094773] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 03/19/2014] [Indexed: 12/03/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA), through its interaction with various proteins involved in DNA synthesis, cell cycle regulation, and DNA repair, plays a central role in maintaining genome stability. We previously reported a novel cancer associated PCNA isoform (dubbed caPCNA), which was significantly expressed in a broad range of cancer cells and tumor tissues, but not in non-malignant cells. We found that the caPCNA-specific antigenic site lies between L126 and Y133, a region within the interconnector domain of PCNA that is known to be a major binding site for many of PCNA's interacting proteins. We hypothesized that therapeutic agents targeting protein-protein interactions mediated through this region may confer differential toxicity to normal and malignant cells. To test this hypothesis, we designed a cell permeable peptide containing the PCNA L126-Y133 sequence. Here, we report that this peptide selectively kills human neuroblastoma cells, especially those with MYCN gene amplification, with much less toxicity to non-malignant human cells. Mechanistically, the peptide is able to block PCNA interactions in cancer cells. It interferes with DNA synthesis and homologous recombination-mediated double-stranded DNA break repair, resulting in S-phase arrest, accumulation of DNA damage, and enhanced sensitivity to cisplatin. These results demonstrate conceptually the utility of this peptide for treating neuroblastomas, particularly, the unfavorable MYCN-amplified tumors.
Collapse
|
175
|
Li J, Qu W, Cheng Y, Sun Y, Jiang Y, Zou T, Wang Z, Xu Y, Zhao H. The Inhibitory Effect of Intravesical Fisetin against Bladder Cancer by Induction of p53 and Down-Regulation of NF-kappa B Pathways in a Rat Bladder Carcinogenesis Model. Basic Clin Pharmacol Toxicol 2014; 115:321-9. [PMID: 24646039 DOI: 10.1111/bcpt.12229] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/26/2014] [Indexed: 12/12/2022]
Affiliation(s)
- Jing Li
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Weixing Qu
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Yongyi Cheng
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Yi Sun
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Yazhuo Jiang
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Tiejun Zou
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Zhiping Wang
- Institute of Urology; The Second Hospital of Lanzhou University; Lanzhou China
| | - Yonggang Xu
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| | - Huacai Zhao
- Department of Urology; Shaanxi Provincial People's Hospital; The Third Affiliated Hospital of the Medical College of Xi'an Jiaotong University; Xi'an China
| |
Collapse
|
176
|
Wang SC. PCNA: a silent housekeeper or a potential therapeutic target? Trends Pharmacol Sci 2014; 35:178-86. [PMID: 24655521 DOI: 10.1016/j.tips.2014.02.004] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 02/17/2014] [Accepted: 02/18/2014] [Indexed: 11/18/2022]
Abstract
Proliferating cell nuclear antigen (PCNA) is known as a molecular marker for proliferation given its role in replication. Three identical molecules of PCNA form a molecular sliding clamp around the DNA double helix. This provides an essential platform on which multiple proteins are dynamically recruited and coordinately regulated. Over the past decade, new research has provided a deeper comprehension of PCNA as a coordinator of essential cellular functions for cell growth, death, and maintenance. Although the biology of PCNA in proliferation has been comprehensively reviewed, research progress in unveiling the potential of targeting PCNA for disease treatment has not been systematically discussed. Here we briefly summarize the basic structural and functional characteristics of PCNA, and then discuss new developments in its protein interactions, trimer formation, and signaling regulation that open the door to possible therapeutic targeting of PCNA.
Collapse
Affiliation(s)
- Shao-Chun Wang
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
177
|
Abstract
Faithful bypass of replication forks encountering obstructive DNA lesions is essential to prevent fork collapse and cell death. PrimPol is a new human primase and translesion polymerase that is able to bypass fork-blocking UV-induced lesions and to restart replication by origin-independent repriming.
Collapse
|
178
|
Natural cytotoxicity receptors and their ligands. Immunol Cell Biol 2013; 92:221-9. [PMID: 24366519 DOI: 10.1038/icb.2013.98] [Citation(s) in RCA: 198] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 11/14/2013] [Accepted: 11/16/2013] [Indexed: 12/15/2022]
Abstract
Natural killer (NK) cells are innate lymphoid cells (ILCs) that participate to the clearance of pathogen-infected cells and tumour cells. NK cells and subsets of ILCs express the natural cytotoxicity receptors (NCRs) NKp46, NKp44 and NKp30 at their surface. NCRs have been shown to recognize a broad spectrum of ligands ranging from viral-, parasite- and bacterial-derived ligands to cellular ligands; however, the full identification of NCR ligands remains to be performed and will undoubtedly contribute to a better understanding of NK cell and ILC biology.
Collapse
|
179
|
Hu WT, Li MQ, Liu W, Jin LP, Li DJ, Zhu XY. IL-33 enhances proliferation and invasiveness of decidual stromal cells by up-regulation of CCL2/CCR2 via NF-κB and ERK1/2 signaling. Mol Hum Reprod 2013; 20:358-72. [PMID: 24344240 DOI: 10.1093/molehr/gat094] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Interleukin (IL)-33, a newly described member of the IL-1 family, has been reported to facilitate primary tumor progression and metastatic dissemination. However, its biological function on decidual stromal cells (DSCs) remains unclear. In this study, we tested the hypothesis whether IL-33 promotes proliferation and invasion of DSCs, and the possible mechanism. IL-33 and its orphan receptor ST2 was found to be co-expressed by DSCs in human first-trimester pregnancy. Addition of IL-33, enhanced the proliferation and invasion of DSCs in a dosage-dependent manner, concomitantly with increasing expression of proliferation relative gene (PCNA, survivin) and invasion relative gene (titin, MMP2). Blocking IL-33/ST2 signaling by soluble sST2 apparently abolished the stimulatory effect on the proliferation, invasiveness and related gene expression in DSCs. We also demonstrated that chemokines CCL2/CCR2 was significantly increased with IL-33 administration. Moreover, inhibition of CCL2/CCR2 activation using CCL2 neutralizing antibody or CCR2 blocker prevented IL-33-stimulated proliferation and invasiveness capacity of DSCs. Increasing phosphorylation of nuclear factor NF-κB p65 and extracellular signal-regulated kinases ERK1/2 after treatment with IL-33 was confirmed by western blotting. And the IL-33-induced CCL2/CCR2 expression was abrogated by treatment with the NF-κB inhibitor BAY 11-7082 or ERK1/2 inhibitor U0126. Finally, we showed that decreased IL-33/ST2 expression was observed in DSCs from spontaneous abortion compared with normal pregnancy at both gene and protein levels. This study provides evidence for the molecular mechanism of IL-33 in promoting proliferation and invasiveness of DSCs by up-regulation of CCL2/CCR2 via NF-κB and ERK1/2 signal pathways and thus contributes insight to the potential of IL-33 involved in successful pregnancy via inducing DSCs mitosis and invasion.
Collapse
Affiliation(s)
- Wen-Ting Hu
- Laboratory for Reproductive Immunology, Hospital & Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai 200011, China
| | | | | | | | | | | |
Collapse
|
180
|
Xu X, Guardiani C, Yan C, Ivanov I. Opening pathways of the DNA clamps proliferating cell nuclear antigen and Rad9-Rad1-Hus1. Nucleic Acids Res 2013; 41:10020-31. [PMID: 24038358 PMCID: PMC3905852 DOI: 10.1093/nar/gkt810] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Proliferating cell nuclear antigen and the checkpoint clamp Rad9-Rad1-Hus1 topologically encircle DNA and act as mobile platforms in the recruitment of proteins involved in DNA damage response and cell cycle regulation. To fulfill these vital cellular functions, both clamps need to be opened and loaded onto DNA by a clamp loader complex—a process, which involves disruption of the DNA clamp’s subunit interfaces. Herein, we compare the relative stabilities of the interfaces using the molecular mechanics Poisson−Boltzmann solvent accessible surface method. We identify the Rad9-Rad1 interface as the weakest and, therefore, most likely to open during clamp loading. We also delineate the dominant interface disruption pathways under external forces in multiple-trajectory steered molecular dynamics runs. We show that, similar to the case of protein folding, clamp opening may not proceed through a single interface breakdown mechanism. Instead, we identify an ensemble of opening pathways, some more prevalent than others, characterized by specific groups of contacts that differentially stabilize the regions of the interface and determine the spatial and temporal patterns of breakdown. In Rad9-Rad1-Hus1, the Rad9-Rad1 and Rad9-Hus1 interfaces share the same dominant unzipping pathway, whereas the Hus1-Rad1 interface is disrupted concertedly with no preferred directionality.
Collapse
Affiliation(s)
- Xiaojun Xu
- Department of Chemistry, Center for Diagnostics and Therapeutics, Georgia State University, GA 30302, USA
| | | | | | | |
Collapse
|
181
|
Sutherland GT, Sheahan PJ, Matthews J, Dennis CVP, Sheedy DS, McCrossin T, Curtis MA, Kril JJ. The effects of chronic alcoholism on cell proliferation in the human brain. Exp Neurol 2013; 247:9-18. [PMID: 23541433 PMCID: PMC4709019 DOI: 10.1016/j.expneurol.2013.03.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Revised: 03/18/2013] [Accepted: 03/20/2013] [Indexed: 01/19/2023]
Abstract
Neurogenesis continues in the human subventricular zone and to a lesser extent in the hippocampal subgranular zone throughout life. Subventricular zone-derived neuroblasts migrate to the olfactory bulb where survivors become integrated as interneurons and are postulated to contribute to odor discrimination. Adult neurogenesis is dysregulated in many neurological, neurovascular and neurodegenerative diseases. Alcohol abuse can result in a neurodegenerative condition called alcohol-related brain damage. Alcohol-related brain damage manifests clinically as cognitive dysfunction and the loss of smell sensation (hyposmia) and pathologically as generalized white matter atrophy and focal neuronal loss. The exact mechanism linking chronic alcohol intoxication with alcohol-related brain damage remains largely unknown but rodent models suggest that decreased neurogenesis is an important component. We investigated this idea by comparing proliferative events in the subventricular zone and olfactory bulb of a well-characterized cohort of 15 chronic alcoholics and 16 age-matched controls. In contrast to the findings in animal models there was no difference in the number of proliferative cell nuclear antigen-positive cells in the subventricular zone of alcoholics (mean±SD=28.7±20.0) and controls (27.6±18.9, p=1.0). There were also no differences in either the total (p=0.89) or proliferative cells (p=0.98) in the granular cell layer of the olfactory bulb. Our findings show that chronic alcohol consumption does not affect cell proliferation in the human SVZ or olfactory bulb. In fact only microglial proliferation could be demonstrated in the latter. Therefore neurogenic deficits are unlikely to contribute to hyposmia in chronic alcoholics.
Collapse
Affiliation(s)
- G T Sutherland
- Discipline of Pathology, Sydney Medical School, The University of Sydney, NSW 2006, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
182
|
Müller R, Misund K, Holien T, Bachke S, Gilljam KM, Våtsveen TK, Rø TB, Bellacchio E, Sundan A, Otterlei M. Targeting proliferating cell nuclear antigen and its protein interactions induces apoptosis in multiple myeloma cells. PLoS One 2013; 8:e70430. [PMID: 23936203 PMCID: PMC3729839 DOI: 10.1371/journal.pone.0070430] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 06/18/2013] [Indexed: 11/18/2022] Open
Abstract
Multiple myeloma is a hematological cancer that is considered incurable despite advances in treatment strategy during the last decade. Therapies targeting single pathways are unlikely to succeed due to the heterogeneous nature of the malignancy. Proliferating cell nuclear antigen (PCNA) is a multifunctional protein essential for DNA replication and repair that is often overexpressed in cancer cells. Many proteins involved in the cellular stress response interact with PCNA through the five amino acid sequence AlkB homologue 2 PCNA-interacting motif (APIM). Thus inhibiting PCNA’s protein interactions may be a good strategy to target multiple pathways simultaneously. We initially found that overexpression of peptides containing the APIM sequence increases the sensitivity of cancer cells to contemporary therapeutics. Here we have designed a cell-penetrating APIM-containing peptide, ATX-101, that targets PCNA and show that it has anti-myeloma activity. We found that ATX-101 induced apoptosis in multiple myeloma cell lines and primary cancer cells, while bone marrow stromal cells and primary healthy lymphocytes were much less sensitive. ATX-101-induced apoptosis was caspase-dependent and cell cycle phase-independent. ATX-101 also increased multiple myeloma cells’ sensitivity against melphalan, a DNA damaging agent commonly used for treatment of multiple myeloma. In a xenograft mouse model, ATX-101 was well tolerated and increased the anti-tumor activity of melphalan. Therefore, targeting PCNA by ATX-101 may be a novel strategy in multiple myeloma treatment.
Collapse
Affiliation(s)
- Rebekka Müller
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristine Misund
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Myeloma Research, Trondheim, Norway
| | - Toril Holien
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Myeloma Research, Trondheim, Norway
| | - Siri Bachke
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Karin M. Gilljam
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Thea K. Våtsveen
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Myeloma Research, Trondheim, Norway
| | - Torstein B. Rø
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Myeloma Research, Trondheim, Norway
| | | | - Anders Sundan
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Myeloma Research, Trondheim, Norway
| | - Marit Otterlei
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- APIM Therapeutics AS, Trondheim, Norway
- * E-mail:
| |
Collapse
|
183
|
Wedgwood S, Lakshminrusimha S, Czech L, Schumacker PT, Steinhorn RH. Increased p22(phox)/Nox4 expression is involved in remodeling through hydrogen peroxide signaling in experimental persistent pulmonary hypertension of the newborn. Antioxid Redox Signal 2013; 18:1765-76. [PMID: 23244636 PMCID: PMC3619152 DOI: 10.1089/ars.2012.4766] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AIM To determine if the NADPH oxidase isoform Nox4 contributes to increased H(2)O(2) generation in persistent pulmonary hypertension of the newborn (PPHN) pulmonary arteries (PA), and to identify downstream signaling targets of Nox4 that contribute to vascular remodeling and vasoconstriction. RESULTS PPHN was induced in lambs by antenatal ligation of the ductus arteriosus at 128 days gestation. After 9 days, lungs, PA, and PA smooth muscle cells (PASMC) were isolated from control and PPHN lambs. Increased expression of p22(phox) and Nox4 in PPHN lungs, PA, and PASMC was associated with increased reactive oxygen species in PPHN PA, increased protein thiol oxidation in PPHN PASMC, and a decreased activity of extracellular superoxide dismutase (ecSOD) in the lungs and PASMC. Nox4 small interfering RNA (siRNA) decreased Nox4 expression and thiol oxidation and increased the ecSOD activity in PPHN PASMC. An increased activity of nuclear factor-kappa B (NFκB) and expression of its target gene cyclin D1 were detected in PPHN lungs, PA, and PASMC. Nox4 siRNA and catalase attenuated these increases in PASMC, and catalase decreased cyclin D1 expression in PPHN lungs. INNOVATION This study demonstrates for the first time that Nox4 expression is elevated in a lamb model of neonatal pulmonary hypertension. It identifies increased NFκB and cyclin D1 expression and a decreased ecSOD activity as targets of increased Nox4 signaling. CONCLUSION PPHN increases p22(phox) and Nox4 expression and activity resulting in elevated H(2)O(2) levels in PPHN PA. Increased H(2)O(2) induces vasoconstriction via mechanisms involving ecSOD inactivation, and stimulates vascular remodeling via NFκB activation and increased cyclin D1 expression. Approaches that inhibit the pulmonary arterial Nox4 activity may attenuate vasoconstriction and vascular remodeling in PPHN.
Collapse
Affiliation(s)
- Stephen Wedgwood
- Department of Pediatrics, Northwestern University, Chicago, IL 60611, USA.
| | | | | | | | | |
Collapse
|
184
|
Actis M, Inoue A, Evison B, Perry S, Punchihewa C, Fujii N. Small molecule inhibitors of PCNA/PIP-box interaction suppress translesion DNA synthesis. Bioorg Med Chem 2013; 21:1972-7. [DOI: 10.1016/j.bmc.2013.01.022] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 01/02/2013] [Accepted: 01/11/2013] [Indexed: 12/19/2022]
|
185
|
Horton NC, Mathew SO, Mathew PA. Novel interaction between proliferating cell nuclear antigen and HLA I on the surface of tumor cells inhibits NK cell function through NKp44. PLoS One 2013; 8:e59552. [PMID: 23527218 PMCID: PMC3602199 DOI: 10.1371/journal.pone.0059552] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 02/18/2013] [Indexed: 01/17/2023] Open
Abstract
NK cell function is closely regulated by numerous inhibitory and activating receptors binding corresponding ligands on the surface of target cells, providing vital first line defenses against infections and cancer. NKp44, originally discovered as an activating NK cell receptor, was recently found to elicit inhibitory effects on NK cell effector function through recognition of cell surface PCNA. Other reports have pointed to potential associations between NKp44 and HLA I molecules, as well as HLA I and Damage Associated Molecular Pattern molecules (DAMPs) on the surface of tumor cells. In this report, we have identified novel interaction between HLA I and PCNA on the surface of human tumor cells by confocal microscopy and immunoprecipitation. In addition to previous reports, we show PCNA on the cell surface where novel association with HLA I does not require the presence of NKp44 expressing NK cells and occurs with endogenous PCNA. The association of HLA I and PCNA forms the inhibitory ligand for NKp44, resulting in inhibition of NK cell cytotoxicity. We further postulate NCR ligands are composed of DAMP molecules localized to the cell surface, colocalizing with HLA I, and potentially heparin sulfate proteoglycans.
Collapse
Affiliation(s)
- Nathan C. Horton
- Department of Molecular Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Stephen O. Mathew
- Department of Molecular Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| | - Porunelloor A. Mathew
- Department of Molecular Biology and Immunology and Institute for Cancer Research, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- * E-mail:
| |
Collapse
|
186
|
Trillo MÁ, Martínez MA, Cid MA, Úbeda A. Retinoic acid inhibits the cytoproliferative response to weak 50‑Hz magnetic fields in neuroblastoma cells. Oncol Rep 2013; 29:885-94. [PMID: 23292364 PMCID: PMC3597587 DOI: 10.3892/or.2012.2212] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 11/22/2012] [Indexed: 12/23/2022] Open
Abstract
We previously reported that intermittent exposure to a 50‑Hz magnetic field (MF) at 100 µT stimulates cell proliferation in the human neuroblastoma cell line NB69. The present study aimed to investigate whether the magnetic field-induced growth promotion also occurs at a lower magnetic flux density of 10 µT. To this purpose, NB69 cells were subjected for 42 h to intermittent exposure, 3 h on/3 h off, to a 50‑Hz MF at a 10 or 100 µT magnetic flux density. The field exposure took place either in the presence or in the absence of the antiproliferative agent retinoic acid. At the end of the treatment and/or incubation period, the cell growth was estimated by hemocytometric counting and spectrophotometric analysis of total protein and DNA contents. Potential changes in DNA synthesis were also assessed through proliferating cell nuclear antigen (PCNA) immunolabeling. The results confirmed previously reported data that a 42-h exposure to a 50‑Hz sine wave MF at 100 µT promotes cell growth in the NB69 cell line, and showed that 10 µT induces a similar proliferative response. This effect, which was significantly associated and linearly correlated with PCNA expression, was abolished by the presence of retinoic acid in the culture medium.
Collapse
Affiliation(s)
- María Ángeles Trillo
- Department of Research-BEM, IRYCIS, Hospital Ramon y Cajal, 28034 Madrid, Spain.
| | | | | | | |
Collapse
|
187
|
Gopalakrishnan R, Sundaram J, Sattu K, Pandi A, Thiruvengadam D. Dietary supplementation of silymarin is associated with decreased cell proliferation, increased apoptosis, and activation of detoxification system in hepatocellular carcinoma. Mol Cell Biochem 2013; 377:163-76. [PMID: 23397134 DOI: 10.1007/s11010-013-1582-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 01/30/2013] [Indexed: 11/29/2022]
Abstract
Hepatocellular carcinoma (HCC) incidence rates are increasing in many parts of the world. HCC's limited treatment remedies and the poor prognosis emphasize the importance in developing an effective chemoprevention for this disease. Here, we investigated the molecular mechanisms involved in the chemoprevention of silymarin in N-nitrosodiethylamine (NDEA)-induced rat model of HCC. Liver of the rats treated with NDEA showed higher proliferation index and glycoconjugates. NDEA treatment also increased the level of anti-apoptotic proteins with simultaneous decrease in the level of pro-apoptotic proteins along with increased accumulation of Cytochrome c in mitochondria. The carcinogenic insult also increased microsomal phase I metabolizing enzymes with a simultaneous decrease in the Phase II detoxifying enzyme glutathione-S-transferase (GST). Whereas dietary silymarin administration along with NDEA treatment significantly decreased the proliferation and down regulated the expression of anti-apoptotic proteins with simultaneously increased expression of pro-apoptotic proteins along with the release of Cytochrome c to cytosol there by activating the intrinsic apoptotic pathway. Silymarin administration also decreased the level of glycoproteins and activated the phase II detoxifying enzyme GST. These results demonstrate that suppression of HCC by silymarin in vivo involves inhibition of proliferation, activation of apoptosis, and efficient detoxification.
Collapse
|
188
|
Strzalka W, Bartnicki F, Pels K, Jakubowska A, Tsurimoto T, Tanaka K. RAD5a ubiquitin ligase is involved in ubiquitination of Arabidopsis thaliana proliferating cell nuclear antigen. JOURNAL OF EXPERIMENTAL BOTANY 2013; 64:859-69. [PMID: 23314815 DOI: 10.1093/jxb/ers368] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
The proliferating cell nuclear antigen (PCNA) is post-translationally modified by ubiquitin in yeast and mammalian cells. It is widely accepted that in yeast mono- and polyubiquitinated PCNA is involved in distinct pathways of DNA postreplication repair. This study showed an interaction between plant ubiquitin and PCNA in the plant cell. Using different approaches, it was demonstrated that Arabidopsis RAD5a ubiquitin ligase is involved in the post-translational modification of plant PCNA. A detailed analysis of the properties of selected Arabidopsis ubiquitin-conjugating enzymes (AtUBC) has shown that a plant homologue of yeast RAD6 (AtUBC2) is sufficient to monoubiquitinate AtPCNA in the absence of ubiquitin ligase. Using different combinations of selected AtUBC proteins together with AtRAD5a, it was demonstrated that plants have potential to use different pathways to ubiquitinate PCNA. The analysis of Arabidopsis PCNA1 and PCNA2 did not demonstrate substantial differences in the ubiquitination pattern between these two proteins. The major ubiquitination target of Arabidopsis PCNA, conserved in eukaryotes, is lysine 164. Taken together, the presented results clearly demonstrate the involvement of Arabidopsis UBC and RAD5a proteins in the ubiquitination of plant PCNA at lysine 164. The data show the complexity of the plant ubiquitination system and open new questions about its regulation in the plant cell.
Collapse
Affiliation(s)
- Wojciech Strzalka
- Department of Plant Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow, Poland.
| | | | | | | | | | | |
Collapse
|
189
|
Loo LWM, Tiirikainen M, Cheng I, Lum-Jones A, Seifried A, Church JM, Gryfe R, Weisenberger DJ, Lindor NM, Gallinger S, Haile RW, Duggan DJ, Thibodeau SN, Casey G, Le Marchand L. Integrated analysis of genome-wide copy number alterations and gene expression in microsatellite stable, CpG island methylator phenotype-negative colon cancer. Genes Chromosomes Cancer 2013; 52:450-66. [PMID: 23341073 DOI: 10.1002/gcc.22043] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 12/04/2012] [Accepted: 12/16/2012] [Indexed: 12/11/2022] Open
Abstract
Microsatellite stable (MSS), CpG island methylator phenotype (CIMP)-negative colorectal tumors, the most prevalent molecular subtype of colorectal cancer, are associated with extensive copy number alteration (CNA) events and aneuploidy. We report on the identification of characteristic recurrent CNA (with frequency >25%) events and associated gene expression profiles for a total of 40 paired tumor and adjacent normal colon tissues using genome-wide microarrays. We observed recurrent CNAs, namely gains at 1q, 7p, 7q, 8p12-11, 8q, 12p13, 13q, 20p, 20q, Xp, and Xq and losses at 1p36, 1p31, 1p21, 4p15-12, 4q12-35, 5q21-22, 6q26, 8p, 14q, 15q11-12, 17p, 18p, 18q, 21q21-22, and 22q. Within these genomic regions we identified 356 genes with significant differential expression (P < 0.0001 and ±1.5-fold change) in the tumor compared to adjacent normal tissue. Gene ontology and pathway analyses indicated that many of these genes were involved in functional mechanisms that regulate cell cycle, cell death, and metabolism. An amplicon present in >70% of the tumor samples at 20q11-20q13 contained several cancer-related genes (AHCY, POFUT1, RPN2, TH1L, and PRPF6) that were upregulated and demonstrated a significant linear correlation (P < 0.05) for gene dosage and gene expression. Copy number loss at 8p, a CNA associated with adenocarcinoma and poor prognosis, was observed in >50% of the tumor samples and demonstrated a significant linear correlation for gene dosage and gene expression for two potential tumor suppressor genes, MTUS1 (8p22) and PPP2CB (8p12). The results from our integration analysis illustrate the complex relationship between genomic alterations and gene expression in colon cancer.
Collapse
Affiliation(s)
- Lenora W M Loo
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI 96813, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
De Biasio A, Blanco FJ. Proliferating Cell Nuclear Antigen Structure and Interactions. PROTEIN-NUCLEIC ACIDS INTERACTIONS 2013; 91:1-36. [DOI: 10.1016/b978-0-12-411637-5.00001-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
191
|
Desbois M, Rusakiewicz S, Locher C, Zitvogel L, Chaput N. Natural killer cells in non-hematopoietic malignancies. Front Immunol 2012; 3:395. [PMID: 23269924 PMCID: PMC3529393 DOI: 10.3389/fimmu.2012.00395] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 12/06/2012] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells belong to the innate immune system and were initially described functionallywise by their spontaneous cytotoxic potential against transformed or virus-infected cells. A delicate balance between activating and inhibiting receptors regulates NK cell tolerance. A better understanding of tissue resident NK cells, of NK cell maturation stages and migration patterns has evolved allowing a thoughtful evaluation of their modus operandi. While evidence has been brought up for their relevance as gate keepers in some hematopoietic malignancies, the role of NK cells against progression and dissemination of solid tumors remains questionable. Hence, many studies pointed out the functional defects of the rare NK cell infiltrates found in tumor beds and the lack of efficacy of adoptively transferred NK cells in patients. However, several preclinical evidences suggest their anti-metastatic role in a variety of mouse tumor models. In the present review, we discuss NK cell functions according to their maturation stage and environmental milieu, the receptor/ligand interactions dictating tumor cell recognition and recapitulate translational studies aimed at deciphering their prognostic or predictive role against human solid malignancies.
Collapse
Affiliation(s)
- Mélanie Desbois
- Institut de Cancérologie Gustave Roussy Villejuif, France ; Centre d'Investigation Clinique Biothérapie 507, Institut de cancérologie Gustave Roussy Villejuif, France ; Faculté de Médecine, Université Paris-Sud Le Kremlin-Bicȴtre, France xs
| | | | | | | | | |
Collapse
|
192
|
Cid MA, Ubeda A, Hernández-Bule ML, Martínez MA, Trillo MÁ. Antagonistic effects of a 50 Hz magnetic field and melatonin in the proliferation and differentiation of hepatocarcinoma cells. Cell Physiol Biochem 2012; 30:1502-16. [PMID: 23235525 DOI: 10.1159/000343338] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2012] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND/AIMS Epidemiological and experimental evidence exists indicating that exposure to weak, extremely low frequency magnetic fields (ELF - MF) could affect cancer progression. It has been proposed that such hypothetical action could be mediated by MF-induced effects on the cellular response to melatonin (MEL), a potentially oncostatic neurohormone. The present study investigates the response of HepG2 cells to intermittent exposure to a 50 Hz, 10 µT MF, in the presence or absence of MEL at physiological (10 nM) or pharmacological doses (1 µM). METHODS The Trypan blue cell exclusion test, BrdU incorporation and PCNA expression assays were carried out to assess the cellular response in terms of viability and proliferation. In addition, albumin and alpha-fetoprotein, were analyzed as specific hepatocellular differentiation markers. RESULTS The results indicate that the MF exerts significant cytoproliferative and dedifferentiating effects that can be prevented by 10 nM MEL. Conversely, MEL exerts cytostatic and differentiating effects on HepG2 that are abolished by simultaneous exposure to MF. CONCLUSION As a whole, these results support the hypothesis that ELF - MF and MEL exert opposite, mutually counteracting effects on cell proliferation and differentiation.
Collapse
Affiliation(s)
- María Antonia Cid
- Dept. Investigación-BEM, Hospital Ramón y Cajal-IRYCIS, Madrid, Spain.
| | | | | | | | | |
Collapse
|
193
|
Subramanian P, Arul D. Attenuation of NDEA-induced hepatocarcinogenesis by naringenin in rats. Cell Biochem Funct 2012; 31:511-7. [PMID: 23172681 DOI: 10.1002/cbf.2929] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 10/13/2012] [Accepted: 10/17/2012] [Indexed: 12/31/2022]
Abstract
Chemoprevention is one of the most promising and realistic approaches in the prevention of cancer. Several bioactive compounds present in fruits and vegetables have revealed their cancer curative potential on hepatocellular carcinoma. Naringenin is one such naturally occurring flavonoid widely found in citrus fruits. In this study, we examined the molecular mechanisms by which naringenin inhibited NDEA-induced hepatocellular carcinoma in rats by analysing the expression patterns of proliferating cell nuclear antigen, Bcl-2, NF-κB, VEGF and MMP-2/9. Enhanced cell proliferation and apoptotic evasion in NDEA-induced hepatocarcinogenesis was associated with imbalance in pro-apoptotic and anti-apoptotic proteins together with upregulation of proliferating cell nuclear antigen (PCNA) and downregulation of caspase-3. Administration of pretreatment and posttreatment of naringenin decreased the expression of PCNA and Bcl-2 and increased the expression of Bax and caspase-3, indicating antiproliferative and apoptotic effects, respectively. Administration of NDEA increased the tumour expression of NF-κB, COX-2, VEGF, MMP-2 and MMP-9 that was correlated with more aggressive lesions and tumour growth. Downregulation of NF-κB, VEGF and MMPs by naringenin seen in the present study were correlated with the inhibition of liver tumour induced by NDEA. Our results suggest that naringenin could act as a legitimate agent by inhibiting cancer processes.
Collapse
Affiliation(s)
- Perumal Subramanian
- Department of Biochemistry and Biotechnology, Faculty of Science, Annamalai University, Tamil Nadu, India.
| | | |
Collapse
|
194
|
Chiara AD, Pederzoli-Ribeil M, Burgel PR, Danel C, Witko-Sarsat V. Targeting cytosolic proliferating cell nuclear antigen in neutrophil-dominated inflammation. Front Immunol 2012; 3:311. [PMID: 23181059 PMCID: PMC3501000 DOI: 10.3389/fimmu.2012.00311] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 09/17/2012] [Indexed: 12/12/2022] Open
Abstract
New therapeutic approaches that can accelerate neutrophil apoptosis under inflammatory conditions to enhance the resolution of inflammation are now under study. Neutrophils are deprived of proliferative capacity and have a tightly controlled lifespan to avoid their persistence at the site of injury. We have recently described that the proliferating cell nuclear antigen (PCNA), a nuclear factor involved in DNA replication and repair of proliferating cells is a key regulator of neutrophil survival. The nuclear-to-cytoplasmic relocalization occurred during granulocytic differentiation and is dependent on a nuclear export sequence thus strongly suggesting that PCNA has physiologic cytoplasmic functions. In this review, we will try to put into perspective the physiologic relevance of PCNA in neutrophils. We will discuss key issues such as molecular structure, post-translational modifications, based on our knowledge of nuclear PCNA, assuming that similar principles governing its function are conserved between nuclear and cytosolic PCNA. The example of cystic fibrosis that features one of the most intense neutrophil-dominated pulmonary inflammation will be discussed. We believe that through an intimate comprehension of the cytosolic PCNA scaffold based on nuclear PCNA knowledge, novel pathways regulating neutrophil survival can be unraveled and innovative agents can be developed to dampen inflammation where it proves detrimental.
Collapse
Affiliation(s)
- Alessia De Chiara
- Department of Immunology and Hematology, INSERM U1016, Cochin Institute ParisFrance
- Paris Descartes UniversityParis, France
- CNRS-UMR 8104Paris, France
| | - Magali Pederzoli-Ribeil
- Department of Immunology and Hematology, INSERM U1016, Cochin Institute ParisFrance
- Paris Descartes UniversityParis, France
- CNRS-UMR 8104Paris, France
| | - Pierre-Régis Burgel
- Paris Descartes UniversityParis, France
- Department of Pneumology, Cochin HospitalParis, France
| | - Claire Danel
- Paris Diderot UniversityParis, France
- Department of Pneumology, Bichat HospitalParis, France
| | - Véronique Witko-Sarsat
- Department of Immunology and Hematology, INSERM U1016, Cochin Institute ParisFrance
- Paris Descartes UniversityParis, France
- CNRS-UMR 8104Paris, France
| |
Collapse
|
195
|
Downregulation of NF-κB and PCNA in the regulatory pathways of apoptosis by cyclooxygenase-2 inhibitors in experimental lung cancer. Mol Cell Biochem 2012; 369:75-86. [PMID: 22752388 DOI: 10.1007/s11010-012-1370-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/14/2012] [Indexed: 10/28/2022]
|
196
|
Takawa M, Cho HS, Hayami S, Toyokawa G, Kogure M, Yamane Y, Iwai Y, Maejima K, Ueda K, Masuda A, Dohmae N, Field HI, Tsunoda T, Kobayashi T, Akasu T, Sugiyama M, Ohnuma SI, Atomi Y, Ponder BAJ, Nakamura Y, Hamamoto R. Histone lysine methyltransferase SETD8 promotes carcinogenesis by deregulating PCNA expression. Cancer Res 2012; 72:3217-27. [PMID: 22556262 DOI: 10.1158/0008-5472.can-11-3701] [Citation(s) in RCA: 156] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Although the physiologic significance of lysine methylation of histones is well known, whether lysine methylation plays a role in the regulation of nonhistone proteins has not yet been examined. The histone lysine methyltransferase SETD8 is overexpressed in various types of cancer and seems to play a crucial role in S-phase progression. Here, we show that SETD8 regulates the function of proliferating cell nuclear antigen (PCNA) protein through lysine methylation. We found that SETD8 methylated PCNA on lysine 248, and either depletion of SETD8 or substitution of lysine 248 destabilized PCNA expression. Mechanistically, lysine methylation significantly enhanced the interaction between PCNA and the flap endonuclease FEN1. Loss of PCNA methylation retarded the maturation of Okazaki fragments, slowed DNA replication, and induced DNA damage, and cells expressing a methylation-inactive PCNA mutant were more susceptible to DNA damage. An increase of methylated PCNA was found in cancer cells, and the expression levels of SETD8 and PCNA were correlated in cancer tissue samples. Together, our findings reveal a function for lysine methylation on a nonhistone protein and suggest that aberrant lysine methylation of PCNA may play a role in human carcinogenesis.
Collapse
Affiliation(s)
- Masashi Takawa
- Laboratory of Molecular Medicine, Human Genome Center, Institute of Medical Science, The University of Tokyo, and National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
197
|
Lo YH, Ho PC, Wang SC. Epidermal growth factor receptor protects proliferating cell nuclear antigen from cullin 4A protein-mediated proteolysis. J Biol Chem 2012; 287:27148-57. [PMID: 22692198 DOI: 10.1074/jbc.m112.388843] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is an essential component for DNA synthesis upon growth stimulation. It has been shown that phosphorylation of PCNA at Tyr-211 by the EGF receptor (EGFR) protects PCNA from polyubiquitylation and degradation, whereas blocking phosphorylation induces ubiquitylation-mediated degradation of the chromatin-bound, but not the -unbound, PCNA, and suppresses cell proliferation. However, the ubiquitin E3 ligase linking growth signaling to the proteolysis of PCNA and the underlying regulatory mechanism remain to be identified. Here we show that, in the absence of Tyr-211 phosphorylation, PCNA is subject to polyubiquitylation at Lys-164 by the CUL4A E3 ligase, resulting in the degradation of PCNA. Mutation of Lys-164 to arginine prevents PCNA ubiquitylation and rescues the degradation of the K164R/Y211F PCNA double mutant. Activation of EGFR inhibits the interaction of PCNA with CUL4A, whereas inhibition of EGFR leads to increased CUL4A-PCNA interaction and CUL4A-dependent ubiquitin-mediated degradation of PCNA. Substitution of endogenous PCNA with the Y211F mutant PCNA conveys enhanced sensitization to EGFR inhibition. Our findings identify CUL4A as the ubiquitin ligase linking the down-regulation of cell surface receptor tyrosine kinase to the nuclear DNA replication machinery in cancer cells.
Collapse
Affiliation(s)
- Yuan-Hung Lo
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267-0521, USA
| | | | | |
Collapse
|
198
|
Tamura RE, de Vasconcellos JF, Sarkar D, Libermann TA, Fisher PB, Zerbini LF. GADD45 proteins: central players in tumorigenesis. Curr Mol Med 2012; 12:634-51. [PMID: 22515981 PMCID: PMC3797964 DOI: 10.2174/156652412800619978] [Citation(s) in RCA: 246] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 11/23/2011] [Accepted: 12/15/2011] [Indexed: 12/18/2022]
Abstract
The Growth Arrest and DNA Damage-inducible 45 (GADD45) proteins have been implicated in regulation of many cellular functions including DNA repair, cell cycle control, senescence and genotoxic stress. However, the pro-apoptotic activities have also positioned GADD45 as an essential player in oncogenesis. Emerging functional evidence implies that GADD45 proteins serve as tumor suppressors in response to diverse stimuli, connecting multiple cell signaling modules. Defects in the GADD45 pathway can be related to the initiation and progression of malignancies. Moreover, induction of GADD45 expression is an essential step for mediating anti-cancer activity of multiple chemotherapeutic drugs and the absence of GADD45 might abrogate their effects in cancer cells. In this review, we present a comprehensive discussion of the functions of GADD45 proteins, linking their regulation to effectors of cell cycle arrest, DNA repair and apoptosis. The ramifications regarding their roles as essential and central players in tumor growth suppression are also examined. We also extensively review recent literature to clarify how different chemotherapeutic drugs induce GADD45 gene expression and how its up-regulation and interaction with different molecular partners may benefit cancer chemotherapy and facilitate novel drug discovery.
Collapse
Affiliation(s)
- Rodrigo Esaki Tamura
- International Centre for Genetic Engineering and Biotechnology, and Medical Biochemistry Division, University of Cape Town, Cape Town, South Africa
| | - Jaíra Ferreira de Vasconcellos
- Centro Infantil Boldrini, Molecular Biology Laboratory, Campinas, Brazil
- State University of Campinas, Faculty of Medical Sciences, Department of Medical Genetics, Campinas, Brazil
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA
| | - Towia A Libermann
- BIDMC Genomics and Proteomics Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Paul B Fisher
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA
| | - Luiz Fernando Zerbini
- International Centre for Genetic Engineering and Biotechnology, and Medical Biochemistry Division, University of Cape Town, Cape Town, South Africa
- BIDMC Genomics and Proteomics Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
199
|
Tan Z, Wortman M, Dillehay KL, Seibel WL, Evelyn CR, Smith SJ, Malkas LH, Zheng Y, Lu S, Dong Z. Small-molecule targeting of proliferating cell nuclear antigen chromatin association inhibits tumor cell growth. Mol Pharmacol 2012; 81:811-9. [PMID: 22399488 PMCID: PMC3362894 DOI: 10.1124/mol.112.077735] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 03/07/2012] [Indexed: 01/30/2023] Open
Abstract
Proliferating cell nuclear antigen (PCNA), a potential anticancer target, forms a homotrimer and is required for DNA replication and numerous other cellular processes. The purpose of this study was to identify novel small molecules that modulate PCNA activity to affect tumor cell proliferation. An in silico screen of a compound library against a crystal structure of PCNA and a subsequent structural similarity search of the ZINC chemical database were carried out to derive relevant docking partners. Nine compounds, termed PCNA inhibitors (PCNA-Is), were selected for further characterization. PCNA-I1 selectively bound to PCNA trimers with a dissociation constant (K(d)) of ~0.2 to 0.4 μM. PCNA-Is promoted the formation of SDS-refractory PCNA trimers. PCNA-I1 dose- and time-dependently reduced the chromatin-associated PCNA in cells. Consistent with its effects on PCNA trimer stabilization, PCNA-I1 inhibited the growth of tumor cells of various tissue types with an IC(50) of ~0.2 μM, whereas it affected the growth of nontransformed cells at significantly higher concentrations (IC(50), ~1.6 μM). Moreover, uptake of BrdU was dose-dependently reduced in cells treated with PCNA-I1. Mechanistically the PCNA-Is mimicked the effect of PCNA knockdown by siRNA, inducing cancer cell arrest at both the S and G(2)/M phases. Thus, we have identified a class of compounds that can directly bind to PCNA, stabilize PCNA trimers, reduce PCNA association with chromatin, and inhibit tumor cell growth by inducing a cell cycle arrest. They are valuable tools in studying PCNA function and may be useful for future PCNA-targeted cancer therapy.
Collapse
Affiliation(s)
- Zongqing Tan
- Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
Epstein-Barr virus BPLF1 deubiquitinates PCNA and attenuates polymerase η recruitment to DNA damage sites. J Virol 2012; 86:8097-106. [PMID: 22623772 DOI: 10.1128/jvi.00588-12] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PCNA is monoubiquitinated in response to DNA damage and fork stalling and then initiates recruitment of specialized polymerases in the DNA damage tolerance pathway, translesion synthesis (TLS). Since PCNA is reported to associate with Epstein-Barr virus (EBV) DNA during its replication, we investigated whether the EBV deubiquitinating (DUB) enzyme encoded by BPLF1 targets ubiquitinated PCNA and disrupts TLS. An N-terminal BPLF1 fragment (a BPLF1 construct containing the first 246 amino acids [BPLF1 1-246]) associated with PCNA and attenuated its ubiquitination in response to fork-stalling agents UV and hydroxyurea in cultured cells. Moreover, monoubiquitinated PCNA was deubiquitinated after incubation with purified BPLF1 1-246 in vitro. BPLF1 1-246 dysregulated TLS by reducing recruitment of the specialized repair polymerase polymerase η (Polη) to the detergent-resistant chromatin compartment and virtually abolished localization of Polη to nuclear repair foci, both hallmarks of TLS. Expression of BPLF1 1-246 decreased viability of UV-treated cells and led to cell death, presumably through deubiquitination of PCNA and the inability to repair damaged DNA. Importantly, deubiquitination of PCNA could be detected endogenously in EBV-infected cells in comparison with samples expressing short hairpin RNA (shRNA) against BPLF1. Further, the specificity of the interaction between BPLF1 and PCNA was dependent upon a PCNA-interacting peptide (PIP) domain within the N-terminal region of BPLF1. Both DUB activity and PIP sequence are conserved in the members of the family Herpesviridae. Thus, deubiquitination of PCNA, normally deubiquitinated by cellular USP1, by the viral DUB can disrupt repair of DNA damage by compromising recruitment of TLS polymerase to stalled replication forks. PCNA is the first cellular target identified for BPLF1 and its deubiquitinating activity.
Collapse
|