1
|
Mukherjee S, Barua A, Wang L, Tian B, Moore CL. The alternative polyadenylation regulator CFIm25 promotes macrophage differentiation and activates the NF-κB pathway. Cell Commun Signal 2025; 23:115. [PMID: 40022203 PMCID: PMC11871739 DOI: 10.1186/s12964-025-02114-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Macrophages are required for development and tissue repair and protect against microbial attacks. In response to external signals, monocytes differentiate into macrophages, but our knowledge of changes that promote this transition at the level of mRNA processing, in particular mRNA polyadenylation, needs advancement if it is to inform new disease treatments. Here, we identify CFIm25, a well-documented regulator of poly(A) site choice, as a novel mediator of macrophage differentiation. METHODS CFIm25 expression was analyzed in differentiating primary human monocytes and monocytic cell lines. Overexpression and depletion experiments were performed to assess CFIm25's role in differentiation, NF-κB signaling, and alternative polyadenylation (APA). mRNA 3' end-focused sequencing was conducted to identify changes in poly(A) site use of genes involved in macrophage differentiation and function. Cell cycle markers, NF-κB pathway components, and their targets were examined. The role of CFIm25 in NF-κB signaling was further evaluated through chemical inhibition and knockdown of pathway regulators. RESULTS CFIm25 showed a striking increase upon macrophage differentiation, suggesting it promotes this process. Indeed, CFIm25 overexpression during differentiation amplified the acquisition of macrophage characteristics and caused an earlier slowing of the cell cycle, a hallmark of this transition, along with APA-mediated downregulation of cyclin D1. The NF-κB signaling pathway plays a major role in maturation of monocytes to macrophages, and the mRNAs of null, TBL1XR1, and NFKB1, all positive regulators of NF-κB signaling, underwent 3'UTR shortening, coupled with an increase in the corresponding proteins. CFIm25 overexpression also elevated phosphorylation of the NF-κB-p65 transcription activator, produced an earlier increase in the NF-κB targets p21, Bcl-XL, ICAM1 and TNF-α, and resulted in greater resistance to NF-κB chemical inhibition. Knockdown of Tables 2 and TBL1XR1 in CFIm25-overexpressing cells attenuated these effects, reinforcing the mechanistic link between CFIm25-regulated APA and NF-κB activation. Conversely, depletion of CFIm25 hindered differentiation and led to lengthening of NFKB1, TAB2, and TBL1XR1 3' UTRs. CONCLUSIONS Our study establishes CFIm25 as a key mediator of macrophage differentiation that operates through a coordinated control of cell cycle progression and NF-κB signaling. This linkage of mRNA processing and immune cell function also expands our understanding of the role of alternative polyadenylation in regulating cell signaling.
Collapse
Affiliation(s)
- Srimoyee Mukherjee
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Atish Barua
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Luyang Wang
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Bin Tian
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Claire L Moore
- Department of Developmental, Molecular, and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA.
| |
Collapse
|
2
|
Mukherjee S, Barua A, Wang L, Tian B, Moore CL. The alternative polyadenylation regulator CFIm25 promotes macrophage differentiation and activates the NF-κβ pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.03.611136. [PMID: 39282342 PMCID: PMC11398326 DOI: 10.1101/2024.09.03.611136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Macrophages are required for our body's development and tissue repair and protect against microbial attacks. We previously reported a crucial role for regulation of mRNA 3'-end cleavage and polyadenylation (C/P) in monocyte to macrophage differentiation. The CFIm25 subunit of the C/P complex showed a striking increase upon differentiation of monocytes with Phorbol Myristate Acetate, suggesting that it promotes this process. To test this hypothesis, CFIm25 was overexpressed in two different monocytic cell lines, followed by differentiation. Both cell lines showed a significant increase in macrophage characteristics and an earlier slowing of the cell cycle. In contrast, depletion of CFIm25 hindered differentiation. Cell cycle slowing upon CFIm25 overexpression was consistent with a greater decrease in the proliferation markers PCNA and cyclin D1, coupled with increased 3'UTR lengthening of cyclin D1 mRNA. Since choice of other poly(A) sites could be affected by manipulating CFIm25, we identified additional genes with altered use of poly(A) sites during differentiation and examined how this changed upon CFIm25 overexpression. The mRNAs of positive regulators of NF-κB signaling, TAB2 and TBL1XR1, and NFKB1, which encodes the NF-κB p50 precursor, underwent 3'UTR shortening that was associated with increased protein expression compared to the control. Cells overexpressing CFIm25 also showed elevated levels of phosphorylated NF-κB-p65 and the NF-κB targets p21, Bcl-XL, ICAM1 and TNF-α at an earlier time and greater resistance to NF-κB chemical inhibition. In conclusion, our study supports a model in which CFIm25 accelerates the monocyte to macrophage transition by promoting alternative polyadenylation events which lead to activation of the NF-κB pathway.
Collapse
|
3
|
Nowak CM, Quarton T, Bleris L. Impact of variability in cell cycle periodicity on cell population dynamics. PLoS Comput Biol 2023; 19:e1011080. [PMID: 37339124 DOI: 10.1371/journal.pcbi.1011080] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 04/06/2023] [Indexed: 06/22/2023] Open
Abstract
The cell cycle consists of a series of orchestrated events controlled by molecular sensing and feedback networks that ultimately drive the duplication of total DNA and the subsequent division of a single parent cell into two daughter cells. The ability to block the cell cycle and synchronize cells within the same phase has helped understand factors that control cell cycle progression and the properties of each individual phase. Intriguingly, when cells are released from a synchronized state, they do not maintain synchronized cell division and rapidly become asynchronous. The rate and factors that control cellular desynchronization remain largely unknown. In this study, using a combination of experiments and simulations, we investigate the desynchronization properties in cervical cancer cells (HeLa) starting from the G1/S boundary following double-thymidine block. Propidium iodide (PI) DNA staining was used to perform flow cytometry cell cycle analysis at regular 8 hour intervals, and a custom auto-similarity function to assess the desynchronization and quantify the convergence to an asynchronous state. In parallel, we developed a single-cell phenomenological model the returns the DNA amount across the cell cycle stages and fitted the parameters using experimental data. Simulations of population of cells reveal that the cell cycle desynchronization rate is primarily sensitive to the variability of cell cycle duration within a population. To validate the model prediction, we introduced lipopolysaccharide (LPS) to increase cell cycle noise. Indeed, we observed an increase in cell cycle variability under LPS stimulation in HeLa cells, accompanied with an enhanced rate of cell cycle desynchronization. Our results show that the desynchronization rate of artificially synchronized in-phase cell populations can be used a proxy of the degree of variance in cell cycle periodicity, an underexplored axis in cell cycle research.
Collapse
Affiliation(s)
- Chance M Nowak
- Bioengineering Department, The University of Texas at Dallas, Richardson, Texas, United States of America
- Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, United States of America
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, United States of America
| | - Tyler Quarton
- Bioengineering Department, The University of Texas at Dallas, Richardson, Texas, United States of America
- Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, United States of America
| | - Leonidas Bleris
- Bioengineering Department, The University of Texas at Dallas, Richardson, Texas, United States of America
- Center for Systems Biology, The University of Texas at Dallas, Richardson, Texas, United States of America
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, United States of America
| |
Collapse
|
4
|
Muniyandi A, Martin M, Sishtla K, Motolani A, Sun M, Jensen NR, Qi X, Boulton ME, Prabhu L, Lu T, Corson TW. PRMT5 is a therapeutic target in choroidal neovascularization. Sci Rep 2023; 13:1747. [PMID: 36720900 PMCID: PMC9889383 DOI: 10.1038/s41598-023-28215-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 01/16/2023] [Indexed: 02/02/2023] Open
Abstract
Ocular neovascular diseases including neovascular age-related macular degeneration (nvAMD) are widespread causes of blindness. Patients' non-responsiveness to currently used biologics that target vascular endothelial growth factor (VEGF) poses an unmet need for novel therapies. Here, we identify protein arginine methyltransferase 5 (PRMT5) as a novel therapeutic target for nvAMD. PRMT5 is a well-known epigenetic enzyme. We previously showed that PRMT5 methylates and activates a proangiogenic and proinflammatory transcription factor, the nuclear factor kappa B (NF-κB), which has a master role in tumor progression, notably in pancreatic ductal adenocarcinoma and colorectal cancer. We identified a potent and specific small molecule inhibitor of PRMT5, PR5-LL-CM01, that dampens the methylation and activation of NF-κB. Here for the first time, we assessed the antiangiogenic activity of PR5-LL-CM01 in ocular cells. Immunostaining of human nvAMD sections revealed that PRMT5 is highly expressed in the retinal pigment epithelium (RPE)/choroid where neovascularization occurs, while mouse eyes with laser induced choroidal neovascularization (L-CNV) showed PRMT5 is overexpressed in the retinal ganglion cell layer and in the RPE/choroid. Importantly, inhibition of PRMT5 by PR5-LL-CM01 or shRNA knockdown of PRMT5 in human retinal endothelial cells (HRECs) and induced pluripotent stem cell (iPSC)-derived choroidal endothelial cells (iCEC2) reduced NF-κB activity and the expression of its target genes, such as tumor necrosis factor α (TNF-α) and VEGF-A. In addition to inhibiting angiogenic properties of proliferation and tube formation, PR5-LL-CM01 blocked cell cycle progression at G1/S-phase in a dose-dependent manner in these cells. Thus, we provide the first evidence that inhibition of PRMT5 impedes angiogenesis in ocular endothelial cells, suggesting PRMT5 as a potential therapeutic target to ameliorate ocular neovascularization.
Collapse
Affiliation(s)
- Anbukkarasi Muniyandi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Matthew Martin
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kamakshi Sishtla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Aishat Motolani
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mengyao Sun
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Nathan R Jensen
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiaoping Qi
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Michael E Boulton
- Department of Ophthalmology and Visual Sciences, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Lakshmi Prabhu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tao Lu
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Medical & Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Timothy W Corson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Pharmacology & Toxicology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
- Department of Biochemistry & Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
Druker J, Wilson JW, Child F, Shakir D, Fasanya T, Rocha S. Role of Hypoxia in the Control of the Cell Cycle. Int J Mol Sci 2021; 22:ijms22094874. [PMID: 34062959 PMCID: PMC8124716 DOI: 10.3390/ijms22094874] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/22/2022] Open
Abstract
The cell cycle is an important cellular process whereby the cell attempts to replicate its genome in an error-free manner. As such, mechanisms must exist for the cell cycle to respond to stress signals such as those elicited by hypoxia or reduced oxygen availability. This review focuses on the role of transcriptional and post-transcriptional mechanisms initiated in hypoxia that interface with cell cycle control. In addition, we discuss how the cell cycle can alter the hypoxia response. Overall, the cellular response to hypoxia and the cell cycle are linked through a variety of mechanisms, allowing cells to respond to hypoxia in a manner that ensures survival and minimal errors throughout cell division.
Collapse
Affiliation(s)
- Jimena Druker
- Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK;
| | - James W. Wilson
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Fraser Child
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Dilem Shakir
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Temitope Fasanya
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
| | - Sonia Rocha
- Department of Molecular Physiology and Cell Signalling, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK; (J.W.W.); (F.C.); (D.S.); (T.F.)
- Correspondence: ; Tel.: +44-(0)151-794-9084
| |
Collapse
|
6
|
de Jesús TJ, Ramakrishnan P. NF-κB c-Rel Dictates the Inflammatory Threshold by Acting as a Transcriptional Repressor. iScience 2020; 23:100876. [PMID: 32062419 PMCID: PMC7031323 DOI: 10.1016/j.isci.2020.100876] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 01/11/2020] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
NF-κB/Rel family of transcription factors plays a central role in initiation and resolution of inflammatory responses. Here, we identified a function of the NF-κB subunit c-Rel as a transcriptional repressor of inflammatory genes. Genetic deletion of c-Rel substantially potentiates the expression of several TNF-α-induced RelA-dependent mediators of inflammation. v-Rel, the viral homologue of c-Rel, but not RelB, also possesses this repressive function. Mechanistically, we found that c-Rel selectively binds to the co-repressor HDAC1 and competitively binds to the DNA mediating HDAC1 recruitment to the promoters of inflammatory genes. A specific point mutation at tyrosine25 in c-Rel's DNA-binding domain, for which a missense single nucleotide variation (Y25H) exists in humans, completely abrogated its ability to bind DNA and repress TNF-α-induced, RelA-mediated transcription. Our findings reveal that the transactivator NF-κB subunit c-Rel also plays a role as a transcriptional repressor in the maintenance of inflammatory homeostasis.
Collapse
Affiliation(s)
- Tristan James de Jesús
- Department of Pathology, School of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, 6526, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA
| | - Parameswaran Ramakrishnan
- Department of Pathology, School of Medicine, Case Western Reserve University and University Hospitals Cleveland Medical Center, 6526, Wolstein Research Building, 2103 Cornell Road, Cleveland, OH 44106, USA; Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; The Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
7
|
Manu KA, Cao PHA, Chai TF, Casey PJ, Wang M. p21cip1/waf1 Coordinate Autophagy, Proliferation and Apoptosis in Response to Metabolic Stress. Cancers (Basel) 2019; 11:cancers11081112. [PMID: 31382612 PMCID: PMC6721591 DOI: 10.3390/cancers11081112] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/25/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022] Open
Abstract
Cancer cells possess metabolic properties that are different from benign cells. These unique characteristics have become attractive targets that are being actively investigated for cancer therapy. p21cip1/waf1, also known as Cyclin-Dependent Kinase inhibitor 1A, is encoded by the CDKN1A gene. It is a major p53 target gene involved in cell cycle progression that has been extensively evaluated. To date, p21 has been reported to regulate various cell functions, both dependent and independent of p53. Besides regulating the cell cycle, p21 also modulates apoptosis, induces senescence, and maintains cellular quiescence in response to various stimuli. p21 transcription is induced in response to stresses, including those from oxidative and chemotherapeutic treatment. A recent study has shown that in response to metabolic stresses such as nutrient and energy depletion, p21 expression is induced to regulate various cell functions. Despite the biological significance, the mechanism of p21 regulation in cancer adaptation to metabolic stress is underexplored and thus represents an exciting field. This review focuses on the recent development of p21 regulation in response to metabolic stress and its impact in inducing cell cycle arrest and death in cancer cells.
Collapse
Affiliation(s)
- Kanjoormana Aryan Manu
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Pham Hong Anh Cao
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Tin Fan Chai
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Patrick J Casey
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mei Wang
- Program in Cancer and Stem Cell Biology, Duke-NUS Graduate Medical School, Singapore 169857, Singapore.
- Department of Biochemistry, National University of Singapore, Singapore 117596, Singapore.
| |
Collapse
|
8
|
Jiang Z, Yuan Y, Zheng H, Cui H, Sun X, Zhao W, Liu X. COMMD1 regulates cell proliferation and cell cycle progression by modulating p21 Cip1 levels. Biosci Biotechnol Biochem 2019; 83:845-850. [PMID: 30667321 DOI: 10.1080/09168451.2019.1569497] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Copper metabolism MURR1 domain-containing 1 (COMMD1) is a protein that participates in multiple cellular processes, including copper homeostasis and nuclear factor kappa B (NF-κB) and hypoxia-inducible factor 1α (HIF-1α) signaling. The COMMD1 upstream regulators X-linked inhibitor of apoptosis protein (XIAP) and p300 and downstream targets such as NF-κB and HIF-1α are involved in the regulation of cell proliferation and cell cycle progression. However, whether COMMD1 regulates cell proliferation and the cell cycle remains unclear. In the present study, we demonstrated that both overexpression and knockdown of COMMD1 affected the proliferation of HEK293 cells, and the cell cycle assay revealed that ectopic expression of COMMD1 arrested the cell cycle at the G1 phase. Furthermore, western blot analysis showed that COMMD1 affected p21 Cip1 levels. Taken together, these results suggest that COMMD1 regulates cell proliferation and cell cycle progression by modulating p21 Cip1 levels. Abbreviations COMMD1: Copper metabolism MURR1 domain containing 1; XIAP: X chromosome-linked inhibitor of apoptosis protein; FCS: Fetal calf serum; WCE: Whole cell extracts; RT-PCR: Reverse transcription-polymerase chain reaction; HEK293: Human embryonic kidney 293; ShRNA: Short hairpin RNA; NF-κB: Nuclear factor kappa-light-chain-enhancer of activated B cells; ARF: Alternate reading frame protein product of the CDKN2A locus.
Collapse
Affiliation(s)
- Zhiwen Jiang
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Yuan Yuan
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Huiling Zheng
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Hongjing Cui
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Xuerong Sun
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Wei Zhao
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China
| | - Xinguang Liu
- a Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Institute of Aging Research , Guangdong Medical University , Dongguan , China.,b Dongguan Scientific Research Center , Guangdong Medical University , Guangdong , China.,c Institute of Biochemistry and Molecular Biology , Guangdong Medical University , Dongguan , China
| |
Collapse
|
9
|
Priebe MK, Dewert N, Amschler K, Erpenbeck L, Heinzerling L, Schön MP, Seitz CS, Lorenz VN. c-Rel is a cell cycle modulator in human melanoma cells. Exp Dermatol 2018; 28:121-128. [PMID: 30466153 DOI: 10.1111/exd.13848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 11/13/2018] [Accepted: 11/19/2018] [Indexed: 12/13/2022]
Abstract
Melanoma progression and resistance to therapy are associated with faulty regulation of signalling molecules including the central transcription factor NF-κB. Increased expression of the c-Rel subunit of NF-κB has been described in progressing melanoma, though mechanistic implications of this upregulation remain unclear. To elucidate the functional role of c-Rel in melanoma biology, we have assessed its expression in human melanoma as well as in melanoma cell lines. Suppression of c-Rel expression in four melanoma cell lines resulted in reduced growth and altered cell cycle regulation, namely G2/M and polyploid phase induction. Moreover, mitotic spindle morphology was profoundly altered in three of the cell lines with a predominance of monopolar structures. These findings suggest that c-Rel is involved in G2/M phase regulation, prevention of polyploidy and, consequently, chromosomal stability. Our results highlight a novel tumor-promoting function of c-Rel in human melanoma cells through governing cell cycle regulation.
Collapse
Affiliation(s)
- Marie K Priebe
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| | - Nadin Dewert
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| | - Katharina Amschler
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| | - Luise Erpenbeck
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| | - Lucie Heinzerling
- Department of Dermatology, Friedrich Alexander University, Erlangen, Germany
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| | - Cornelia S Seitz
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| | - Verena N Lorenz
- Department of Dermatology, Venereology and Allergology, Georg-August-University, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
10
|
The role of NF-κB and miRNA in oral cancer and cancer stem cells with or without HPV16 infection. PLoS One 2018; 13:e0205518. [PMID: 30372446 PMCID: PMC6205583 DOI: 10.1371/journal.pone.0205518] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
A small subpopulation of cancer stem-like cells (CSCs) present in almost all tumors is responsible for drug resistance and tumor recurrence. The role of NF-kB and miRNA in close association with essential risk factors, tobacco, alcohol and high risk HPV infection during oral carcinogenesis and its prognosis is not well understood. We have isolated cancer stem like SP cells from both HPV+/-ve oral squamous cell carcinoma (OSCC) cell lines and primary tumors, which formed orospheres, expressed stemness markers Oct4, Sox-2, CD133 and CD117. These cells showed differentially upregulated expression of NF-kB proteins and selective overexpression of viral oncogenes E6/E7 only in HPV16+ve cells which formed higher number of orospheres, overexpressed c-Rel and selectively activated p65 that heterodimerized with p50 to show higher DNA binding activity. Further, selective over expression of miR-21 and miR-155 and downregulation of miR-34a were demonstrated by HPV+ve CSCs which overexpress HPV16 oncogene E6 that is responsible for the maintenance of stemness. While, HPV-ve CSCs show exclusively p50 homodimeriztion, poor differentiation and worst prognosis, HPV infection induced participation of p65 along with deregulated expression of specific miRNAs led to well differentiation of tumors and better prognosis.
Collapse
|
11
|
Tomas-Hernandez S, Garcia-Vallvé S, Pujadas G, Valls C, Ojeda-Montes MJ, Gimeno A, Cereto-Massagué A, Roca-Martinez J, Suárez M, Arola L, Blanco J, Mulero M, Beltran-Debón R. Anti-inflammatory and Proapoptotic Properties of the Natural Compound o-Orsellinaldehyde. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:10952-10963. [PMID: 30269491 DOI: 10.1021/acs.jafc.8b00782] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Metabolic syndrome is a cluster of medical conditions that increases the risk of developing cardiovascular disease and type 2 diabetes. Numerous studies have shown that inflammation is directly involved in the onset of metabolic syndrome and related pathologies. In this study, in silico techniques were applied to a natural products database containing molecules isolated from mushrooms from the Catalan forests to predict molecules that can act as human nuclear-factor κβ kinase 2 (IKK-2) inhibitors. IKK-2 is the main component responsible for activating the nuclear-factor κβ transcription factor (NF-κβ). One of these predicted molecules was o-orsellinaldehyde, a molecule present in the mushroom Grifola frondosa. This study shows that o-orsellinaldehyde presents anti-inflammatory and pro-apoptotic properties by acting as IKK-2 inhibitor. Additionally, we suggest that the anti-inflammatory and pro-apoptotic properties of Grifola frondosa mushroom could partially be explained by the presence of o-orsellinaldehyde on its composition.
Collapse
Affiliation(s)
- Sarah Tomas-Hernandez
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Santiago Garcia-Vallvé
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
- Technological Unit of Nutrition and Health , EURECAT-Technological Center of Catalonia , Avinguda Universitat, 1 , 43204 Reus , Spain
| | - Gerard Pujadas
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
- Technological Unit of Nutrition and Health , EURECAT-Technological Center of Catalonia , Avinguda Universitat, 1 , 43204 Reus , Spain
| | - Cristina Valls
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - María José Ojeda-Montes
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Aleix Gimeno
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Adrià Cereto-Massagué
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Joel Roca-Martinez
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Manuel Suárez
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili, (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Lluis Arola
- Technological Unit of Nutrition and Health , EURECAT-Technological Center of Catalonia , Avinguda Universitat, 1 , 43204 Reus , Spain
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili, (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Jordi Blanco
- Laboratory of Toxicology and Environmental Health, School of Medicine, IISPV , Universitat Rovira i Virgili , 43201 Reus , Spain
| | - Miquel Mulero
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| | - Raúl Beltran-Debón
- Cheminformatics and Nutrition Group, Department of Biochemistry and Biotechnology , Universitat Rovira i Virgili (URV), Campus de Sescelades , 43007 Tarragona , Spain
| |
Collapse
|
12
|
Lorenz VN, Schön MP, Seitz CS. c-Rel in Epidermal Homeostasis: A Spotlight on c-Rel in Cell Cycle Regulation. J Invest Dermatol 2016; 136:1090-1096. [PMID: 27032306 DOI: 10.1016/j.jid.2016.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/15/2016] [Accepted: 02/03/2016] [Indexed: 12/19/2022]
Abstract
To maintain proper skin barrier function, epidermal homeostasis requires a subtly governed balance of proliferating and differentiating keratinocytes. While differentiation takes place in the suprabasal layers, proliferation, including mitosis, is usually restricted to the basal layer. Only recently identified as an important regulator of epidermal homeostasis, c-Rel, an NF-κB transcription factor subunit, affects the viability and proliferation of epidermal keratinocytes. In human keratinocytes, decreased expression of c-Rel causes a plethora of dysregulated cellular functions including impaired cell viability, increased apoptosis, and abnormalities during mitosis and cell cycle regulation. On the other hand, c-Rel shows aberrant expression in many epidermal tumors. Here, in the context of its role in different cell types and compared with other NF-κB subunits, we discuss the putative function of c-Rel as a regulator of epidermal homeostasis and mitotic progression. In addition, implications for disease pathophysiology with perturbed c-Rel function and abnormal homeostasis, such as epidermal carcinogenesis, will be discussed.
Collapse
Affiliation(s)
- Verena N Lorenz
- Department of Dermatology, Venereology and Allergology, Georg August University, Göttingen, Germany.
| | - Michael P Schön
- Department of Dermatology, Venereology and Allergology, Georg August University, Göttingen, Germany
| | - Cornelia S Seitz
- Department of Dermatology, Venereology and Allergology, Georg August University, Göttingen, Germany
| |
Collapse
|
13
|
Haery L, Mussakhan S, Waxman DJ, Gilmore TD. Evidence for an oncogenic modifier role for mutant histone acetyltransferases in diffuse large B-cell lymphoma. Leuk Lymphoma 2016; 57:2661-71. [PMID: 27003102 DOI: 10.3109/10428194.2016.1160083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mutations in histone acetyltransferases (HATs) are among the most common mutations in diffuse large B-cell lymphoma (DLBCL). We previously showed that two human DLBCL cell lines, RC-K8 and SUDHL2, express C-terminally truncated, HAT domain-deficient p300 proteins (p300ΔC) that are required for optimal cell proliferation. Microarray analysis of mRNA expression in RC-K8 cells following p300ΔC knockdown shows upregulation of NF-κB and p53 gene expression programs and downregulation of a MYC gene expression program. Experiments indicate that these gene expression changes are due to inhibitory effects of p300ΔC on NF-κB activity and on p53 protein levels and stimulatory effects on MYC protein levels, suggesting that p300ΔC mutants enhance the proliferation of DLBCL cells by adjusting the transcriptional output of cell-specific oncoproteins. We propose that p300/CBP gene truncation represents a new class of oncogenic mutation that optimizes the activity of context-specific oncogenic transcription factors. We propose 'oncogenic modifier' to describe such mutations.
Collapse
Affiliation(s)
- Leila Haery
- a Department of Biology , Boston University , Boston , MA , USA
| | | | - David J Waxman
- a Department of Biology , Boston University , Boston , MA , USA
| | | |
Collapse
|
14
|
Kochupurakkal BS, Wang ZC, Hua T, Culhane AC, Rodig SJ, Rajkovic-Molek K, Lazaro JB, Richardson AL, Biswas DK, Iglehart JD. RelA-Induced Interferon Response Negatively Regulates Proliferation. PLoS One 2015; 10:e0140243. [PMID: 26460486 PMCID: PMC4604146 DOI: 10.1371/journal.pone.0140243] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 09/23/2015] [Indexed: 12/21/2022] Open
Abstract
Both oncogenic and tumor-suppressor activities are attributed to the Nuclear Factor kappa B (NF-kB) pathway. Moreover, NF-kB may positively or negatively regulate proliferation. The molecular determinants of these opposing roles of NF-kB are unclear. Using primary human mammary epithelial cells (HMEC) as a model, we show that increased RelA levels and consequent increase in basal transcriptional activity of RelA induces IRF1, a target gene. Induced IRF1 upregulates STAT1 and IRF7, and in consort, these factors induce the expression of interferon response genes. Activation of the interferon pathway down-regulates CDK4 and up-regulates p27 resulting in Rb hypo-phosphorylation and cell cycle arrest. Stimulation of HMEC with IFN-γ elicits similar phenotypic and molecular changes suggesting that basal activity of RelA and IFN-γ converge on IRF1 to regulate proliferation. The anti-proliferative RelA-IRF1-CDK4 signaling axis is retained in ER+/HER2- breast tumors analyzed by The Cancer Genome Atlas (TCGA). Using immuno-histochemical analysis of breast tumors, we confirm the negative correlation between RelA levels and proliferation rate in ER+/HER2- breast tumors. These findings attribute an anti-proliferative tumor-suppressor role to basal RelA activity. Inactivation of Rb, down-regulation of RelA or IRF1, or upregulation of CDK4 or IRF2 rescues the RelA-IRF1-CDK4 induced proliferation arrest in HMEC and are points of disruption in aggressive tumors. Activity of the RelA-IRF1-CDK4 axis may explain favorable response to CDK4/6 inhibition observed in patients with ER+ Rb competent tumors.
Collapse
Affiliation(s)
- Bose S. Kochupurakkal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- * E-mail: (JDI); (BSK)
| | - Zhigang C. Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Tony Hua
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Aedin C. Culhane
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Scott J. Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | | | - Jean-Bernard Lazaro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Andrea L. Richardson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
| | - Debajit K. Biswas
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - J. Dirk Iglehart
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, United States of America
- Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, United States of America
- * E-mail: (JDI); (BSK)
| |
Collapse
|
15
|
Abstract
The NF-κB (nuclear factor κB) transcription factor family is a pleiotropic regulator of many cellular pathways, providing a mechanism for the cell to respond to a wide variety of stimuli and environmental challenges. It is not surprising therefore that an important component of NF-κB's function includes regulation of the cell cycle. However, this aspect of its behaviour is often overlooked and receives less attention than its ability to induce inflammatory gene expression. In the present article, we provide an updated review of the current state of our knowledge about integration of NF-κB activity with cell cycle regulation, including newly characterized direct and indirect target genes in addition to the mechanisms through which NF-κB itself can be regulated by the cell cycle.
Collapse
|
16
|
Rahman M, Salajegheh A, Smith R, Lam AY. B-Raf mutation: A key player in molecular biology of cancer. Exp Mol Pathol 2013; 95:336-42. [DOI: 10.1016/j.yexmp.2013.10.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 10/15/2013] [Indexed: 12/21/2022]
|
17
|
Ansah C, Mensah KB. A review of the anticancer potential of the antimalarial herbal cryptolepis sanguinolenta and its major alkaloid cryptolepine. Ghana Med J 2013; 47:137-147. [PMID: 24391229 PMCID: PMC3875281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023] Open
Abstract
Cryptolepis sanguinolenta (Lindl.) Schltr (Periplocaceae), has a longstanding traditional use in the treatment of malaria in the West African region. Recent evidence suggests that the aqueous extract from the roots and the major alkaloid from the plant, cryptolepine, have prospects as cancer chemotherapeutic agents on account of their potent cytotoxicity to mammalian cells. Several mechanisms have been proposed to explain the cytotoxic activities of the agents. However, emerging evidence from their anti-inflammatory actions suggest that the mechanism of the cytotoxicity may be closely related to its anti-inflammatory activity. This review looks at the mechanisms of cryptolepis-induced cytotoxicity, its link with inflammation and its potential as anticancer agent. The elucidation of these interwoven mechanisms may be useful in the development of cryptolepine or other analogues as new anticancer agents.
Collapse
Affiliation(s)
- C Ansah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - K B Mensah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
18
|
Huang WC, Hung MC. Beyond NF-κB activation: nuclear functions of IκB kinase α. J Biomed Sci 2013; 20:3. [PMID: 23343355 PMCID: PMC3563485 DOI: 10.1186/1423-0127-20-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/11/2013] [Indexed: 12/15/2022] Open
Abstract
IκB kinase (IKK) complex, the master kinase for NF-κB activation, contains two kinase subunits, IKKα and IKKβ. In addition to mediating NF-κB signaling by phosphorylating IκB proteins during inflammatory and immune responses, the activation of the IKK complex also responds to various stimuli to regulate diverse functions independently of NF-κB. Although these two kinases share structural and biochemical similarities, different sub-cellular localization and phosphorylation targets between IKKα and IKKβ account for their distinct physiological and pathological roles. While IKKβ is predominantly cytoplasmic, IKKα has been found to shuttle between the cytoplasm and the nucleus. The nuclear-specific roles of IKKα have brought increasing complexity to its biological function. This review highlights major advances in the studies of the nuclear functions of IKKα and the mechanisms of IKKα nuclear translocation. Understanding the nuclear activity is essential for targeting IKKα for therapeutics.
Collapse
Affiliation(s)
- Wei-Chien Huang
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 40447, Taiwan.
| | | |
Collapse
|
19
|
To NFκB or not to NFκB: The Dilemma on How to Inhibit a Cancer Cell Fate Regulator. Transl Med UniSa 2012; 4:73-85. [PMID: 23905066 PMCID: PMC3728801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Nuclear factor κB (NFκB) is a transcription factor that plays an important role in carcinogenesis as well as in the regulation of inflammatory response. NFκB is constitutively expressed in tumours where it induces the expression of genes which promote cell proliferation, apoptotic events, angiogenesis, invasion and metastasis. Furthermore, many cancer cells show aberrant or constitutive NFκB activation that mediates resistance to chemo- and radio-therapy. Therefore, the inhibition of NFκB activity appears a potential therapeutic strategy for cancer treatment. In this review, we focus on the role of NFκB in carcinogenesis and summarize actual inhibitors of NFκB that could be potential therapeutic target in cancer therapy.
Collapse
|
20
|
Gilmore TD, Gerondakis S. The c-Rel Transcription Factor in Development and Disease. Genes Cancer 2012; 2:695-711. [PMID: 22207895 DOI: 10.1177/1947601911421925] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2011] [Accepted: 08/08/2011] [Indexed: 12/21/2022] Open
Abstract
c-Rel is a member of the nuclear factor κB (NF-κB) transcription factor family. Unlike other NF-κB proteins that are expressed in a variety of cell types, high levels of c-Rel expression are found primarily in B and T cells, with many c-Rel target genes involved in lymphoid cell growth and survival. In addition to c-Rel playing a major role in mammalian B and T cell function, the human c-rel gene (REL) is a susceptibility locus for certain autoimmune diseases such as arthritis, psoriasis, and celiac disease. The REL locus is also frequently altered (amplified, mutated, rearranged), and expression of REL is increased in a variety of B and T cell malignancies and, to a lesser extent, in other cancer types. Thus, agents that modulate REL activity may have therapeutic benefits for certain human cancers and chronic inflammatory diseases.
Collapse
|
21
|
Ben-Abdallah M, Sturny-Leclère A, Avé P, Louise A, Moyrand F, Weih F, Janbon G, Mémet S. Fungal-induced cell cycle impairment, chromosome instability and apoptosis via differential activation of NF-κB. PLoS Pathog 2012; 8:e1002555. [PMID: 22396644 PMCID: PMC3291658 DOI: 10.1371/journal.ppat.1002555] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2011] [Accepted: 01/12/2012] [Indexed: 12/15/2022] Open
Abstract
Microbial pathogens have developed efficient strategies to compromise host immune responses. Cryptococcus neoformans is a facultative intracellular pathogen, recognised as the most common cause of systemic fungal infections leading to severe meningoencephalitis, mainly in immunocompromised patients. This yeast is characterized by a polysaccharide capsule, which inhibits its phagocytosis. Whereas phagocytosis escape and macrophage intracellular survival have been intensively studied, extracellular survival of this yeast and restraint of host innate immune response are still poorly understood. In this study, we have investigated whether C. neoformans affected macrophage cell viability and whether NF-κB (nuclear factor-κB), a key regulator of cell growth, apoptosis and inflammation, was involved. Using wild-type (WT) as well as mutant strains of C. neoformans for the pathogen side, and WT and mutant cell lines with altered NF-κB activity or signalling as well as primary macrophages for the host side, we show that C. neoformans manipulated NF-κB-mediated signalling in a unique way to regulate macrophage cell fate and viability. On the one hand, serotype A strains reduced macrophage proliferation in a capsule-independent fashion. This growth decrease, which required a critical dosage of NF-κB activity, was caused by cell cycle disruption and aneuploidy, relying on fungal-induced modification of expression of several cell cycle checkpoint regulators in S and G2/M phases. On the other hand, C. neoformans infection induced macrophage apoptosis in a capsule-dependent manner with a differential requirement of the classical and alternative NF-κB signalling pathways, the latter one being essential. Together, these findings shed new light on fungal strategies to subvert host response through uncoupling of NF-κB activity in pathogen-controlled apoptosis and impairment of cell cycle progression. They also provide the first demonstration of induction of aneuploidy by a fungal pathogen, which may have wider implications for human health as aneuploidy is proposed to promote tumourigenesis.
Collapse
Affiliation(s)
- Mariem Ben-Abdallah
- Institut Pasteur, Unité de Mycologie Moléculaire, Département Infection et Epidémiologie, Paris, France
- CNRS, URA3012, Paris, France
| | - Aude Sturny-Leclère
- Institut Pasteur, Unité de Mycologie Moléculaire, Département Infection et Epidémiologie, Paris, France
- CNRS, URA3012, Paris, France
| | - Patrick Avé
- Institut Pasteur, Unité d'Histopathologie, Département Infection et Epidémiologie, Paris, France
| | - Anne Louise
- Institut Pasteur, Plateforme d'Imagerie Dynamique, Paris, France
| | - Frédérique Moyrand
- Institut Pasteur, Unité des Aspergillus, Département de Parasitologie et Mycologie, Paris, France
| | - Falk Weih
- Leibniz-Institute for Age Research - Fritz-Lipmann-Institute, Research Group Immunology, Jena, Germany
| | - Guilhem Janbon
- Institut Pasteur, Unité des Aspergillus, Département de Parasitologie et Mycologie, Paris, France
| | - Sylvie Mémet
- Institut Pasteur, Unité de Mycologie Moléculaire, Département Infection et Epidémiologie, Paris, France
- CNRS, URA3012, Paris, France
- * E-mail:
| |
Collapse
|
22
|
NF-κB hyper-activation by HTLV-1 tax induces cellular senescence, but can be alleviated by the viral anti-sense protein HBZ. PLoS Pathog 2011; 7:e1002025. [PMID: 21552325 PMCID: PMC3084201 DOI: 10.1371/journal.ppat.1002025] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 01/28/2011] [Indexed: 11/30/2022] Open
Abstract
Activation of I-κB kinases (IKKs) and NF-κB by the human T lymphotropic virus type 1 (HTLV-1) trans-activator/oncoprotein, Tax, is thought to promote cell proliferation and transformation. Paradoxically, expression of Tax in most cells leads to drastic up-regulation of cyclin-dependent kinase inhibitors, p21CIP1/WAF1 and p27KIP1, which cause p53-/pRb-independent cellular senescence. Here we demonstrate that p21CIP1/WAF1-/p27KIP1-mediated senescence constitutes a checkpoint against IKK/NF-κB hyper-activation. Senescence induced by Tax in HeLa cells is attenuated by mutations in Tax that reduce IKK/NF-κB activation and prevented by blocking NF-κB using a degradation-resistant mutant of I-κBα despite constitutive IKK activation. Small hairpin RNA-mediated knockdown indicates that RelA induces this senescence program by acting upstream of the anaphase promoting complex and RelB to stabilize p27KIP1 protein and p21CIP1/WAF1 mRNA respectively. Finally, we show that down-regulation of NF-κB by the HTLV-1 anti-sense protein, HBZ, delay or prevent the onset of Tax-induced senescence. We propose that the balance between Tax and HBZ expression determines the outcome of HTLV-1 infection. Robust HTLV-1 replication and elevated Tax expression drive IKK/NF-κB hyper-activation and trigger senescence. HBZ, however, modulates Tax-mediated viral replication and NF-κB activation, thus allowing HTLV-1-infected cells to proliferate, persist, and evolve. Finally, inactivation of the senescence checkpoint can facilitate persistent NF-κB activation and leukemogenesis. Transcription factors of the NF-κB/Rel family are critical for the proliferation of lymphocytes and the expression of genes that mediate inflammatory and immune responses. They are often aberrantly activated in human cancers, especially leukemia, where they confer survival and proliferation advantages. Through the study of the trans-activator/oncoprotein, Tax, of the human T-lymphotropic virus type 1 (HTLV-1), we have found that persistent and potentially oncogenic activation of NF-κB triggers a defense mechanism that commits cells into senescence, an irreversible state of cell cycle arrest. This checkpoint is turned on by hyper-activated p65/RelA and is mediated by two cyclin-dependent kinase inhibitors, p21 and p27, in a p53- and pRb-independent manner. It is often impaired in cancer cells with constitutively active NF-κB. Our results anticipate that the anti-sense protein of HTLV-1, HBZ, which down-regulates NF-κB and HTLV-1 trans-activation by Tax, would mitigate or prevent Tax-induced senescence. This prediction has been borne out experimentally. Thus, Tax promotes robust HTLV-1 replication, potent NF-κB activation and senescence, while HBZ attenuates Tax-driven viral replication and NF-κB activation to allow proliferation of infected cells and persistent infection. Finally, our data support the notion that inactivation of the senescence checkpoint facilitates chronic NF-κB hyper-activation, a critical step in leukemia development.
Collapse
|
23
|
A rearranged EP300 gene in the human B-cell lymphoma cell line RC-K8 encodes a disabled transcriptional co-activator that contributes to cell growth and oncogenicity. Cancer Lett 2011; 302:76-83. [PMID: 21232847 DOI: 10.1016/j.canlet.2010.12.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 12/12/2010] [Accepted: 12/18/2010] [Indexed: 12/27/2022]
Abstract
Human diffuse large B-cell lymphoma cell line RC-K8 has an altered EP300 locus that encodes a C-terminally truncated histone acetyltransferase (HAT) protein (p300ΔC). We now show that p300ΔC contains 1047N-terminal amino acids of p300 fused to 25 amino acids encoded by sequences from chromosome 6. Over-expressed p300ΔC localized to nuclear subdomains and interacted with transcription factor REL. p300ΔC did not function as a co-activator for REL-directed transactivation, and blocked the ability of wild-type p300 to enhance transcriptional activation by REL. Knock down of p300ΔC in RC-K8 cells reduced their growth in both liquid culture and soft agar. Truncations of p300 were not found in eight other B-lymphoma cell lines. These results suggest that p300ΔC contributes to the oncogenic state of RC-K8 cells by acting as a defective co-activator.
Collapse
|
24
|
Liu JJ, Lin M, Yu JY, Liu B, Bao JK. Targeting apoptotic and autophagic pathways for cancer therapeutics. Cancer Lett 2011; 300:105-14. [DOI: 10.1016/j.canlet.2010.10.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2010] [Revised: 09/07/2010] [Accepted: 10/04/2010] [Indexed: 12/19/2022]
|
25
|
Bao BY, Ting HJ, Hsu JW, Yasmin-Karim S, Messing E, Lee YF. Down-regulation of NF-kappaB signals is involved in loss of 1alpha,25-dihydroxyvitamin D3 responsiveness. J Steroid Biochem Mol Biol 2010; 120:11-21. [PMID: 20206692 DOI: 10.1016/j.jsbmb.2010.02.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 02/11/2010] [Accepted: 02/14/2010] [Indexed: 01/02/2023]
Abstract
Vitamin D anti-tumor effect is often found reduced in the late stages of cancer. To uncover vitamin D resistance mechanism, we established a vitamin D-resistant human prostate cancer LNCaP cell line, LNCaP-R, by chronic exposure of cells to 1alpha,25-dihydroxyvitamin D(3) (1,25-VD). The vitamin D receptor (VDR)-mediated transcriptional activity was reduced in LNCaP-R, whereas VDR expression level and DNA-binding capacity were similar compared to parental cells (LNCaP-P). The expressions of the key factors involved in VDR transactivity, including CYP24A1 and VDR-associated proteins are all increased in LNCaP-R cells, and yet treatment with ketoconazole, P450 enzymes inhibitor, as well as trichostatin A (TSA), a histone deacetylase inhibitor, did not sensitize LNCaP-R cells response to vitamin D, suggesting that neither a local 1,25-VD availability, nor VDR-associated proteins are responsible for the vitamin D resistance. Interestingly, nuclear factor-kappaB (NF-kappaB) signaling, which is critical for 1,25-VD/VDR activity was found reduced in LNCaP-R cells, thereby treatment with NF-kappaB activator, 12-O-tetradecanoylphorbol-13-acetate (TPA), can sensitize LNCaP-R vitamin D response. Together, we conclude that NF-kappaB signaling is critical for vitamin D sensitivity, and dysregulation of this pathway would result in vitamin D resistance and disease progression.
Collapse
Affiliation(s)
- Bo-Ying Bao
- Department of Urology and Pathology & Laboratory Medicine, Rochester, NY 14642, USA
| | | | | | | | | | | |
Collapse
|
26
|
Janbandhu VC, Singh AK, Mukherji A, Kumar V. p65 Negatively regulates transcription of the cyclin E gene. J Biol Chem 2010; 285:17453-64. [PMID: 20385564 DOI: 10.1074/jbc.m109.058974] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
NF-kappaB family members play a pivotal role in many cellular and organismal functions, including the cell cycle. As an activator of cyclin D1 and p21(Waf1) genes, NF-kappaB has been regarded as a critical modulator of cell cycle. To study the involvement of NF-kappaB in G(1)/S phase regulation, the levels of selected transcriptional regulators were monitored following overexpression of NF-kappaB or its physiological induction by tumor necrosis factor-alpha. Cyclin E gene was identified as a major transcriptional target of NF-kappaB. Recruitment of NF-kappaB to the cyclin E promoter was correlated with the transrepression of cyclin E gene. Ligation-mediated PCR and micrococcal nuclease-Southern assays suggested the nucleosomal nature of this region while chromatin immunoprecipitation analysis confirmed the exchange of cofactors following tumor necrosis factor-alpha treatment or release from serum starvation. There was a progressive reduction in cyclin E transcription along with the accumulation of catalytically inactive cyclin E-cdk2 complexes and arrest of cells in G(1)/S-phase. Thus, our study clearly establishes NF-kappaB as a negative regulator of cell cycle through transcriptional repression of cyclin E.
Collapse
Affiliation(s)
- Vaibhao C Janbandhu
- Virology Group, International Centre for Genetic Engineering and Biotechnology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | | | | | | |
Collapse
|
27
|
NF-kappaB and cancer: how intimate is this relationship. Mol Cell Biochem 2010; 336:25-37. [PMID: 19823771 PMCID: PMC3148942 DOI: 10.1007/s11010-009-0267-2] [Citation(s) in RCA: 314] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 09/15/2009] [Indexed: 02/03/2023]
Abstract
NF-kappaB, a transcription factor first discovered in 1986, is now known to be closely connected to the process of tumorogenesis based on a multiplicity of evidence. (1) NF-kappaB is activated in response to tobacco, stress, dietary agents, obesity, alcohol, infectious agents, irradiation, and environmental stimuli that account for as much as 95% of all cancers. (2) The transcription factor has been linked with transformation of cells. (3) It is constitutively active in most tumor cells. (4) It has also been linked with the survival of cancer stem cells, an early progenitor cell that has acquired self-renewal potential. (5) NF-kappaB regulates the expression of most anti-apoptotic gene products associated with the survival of the tumor. (6) It also regulates the gene products linked with proliferation of tumors. (7) The transcription factor controls the expression of gene products linked with invasion, angiogenesis, and metastasis of cancer. (8) While most carcinogens activate NF-kappaB, most chemopreventive agents suppress its activation. These observations suggest that NF-kappaB is intimately intertwined with cancer growth and metastasis. The mechanism that leads to constitutive activation of NF-kappaB in hematological, gastrointestinal, genitourinary, gynecological, thoracic head and neck, breast, and skin cancers, and the ways NF-kappaB is activated are the topics of discussion in this review.
Collapse
|
28
|
Garbati MR, Alço G, Gilmore TD. Histone acetyltransferase p300 is a coactivator for transcription factor REL and is C-terminally truncated in the human diffuse large B-cell lymphoma cell line RC-K8. Cancer Lett 2009; 291:237-45. [PMID: 19948376 DOI: 10.1016/j.canlet.2009.10.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2009] [Revised: 10/22/2009] [Accepted: 10/26/2009] [Indexed: 01/03/2023]
Abstract
Human c-Rel (REL) is a member of the NF-kappaB family of transcription factors. REL's normal physiological role is in the regulation of B-cell proliferation and survival. The REL gene is amplified in many human B-cell lymphomas and overexpression of REL can transform chicken lymphoid cells. In this report, histone acetyltransferase p300 enhanced REL-induced transactivation and interacted with REL both in vitro and in REL-transformed chicken spleen cells and the B-lymphoma cell line RC-K8, in which REL is constitutively active and required for proliferation. However, due to a deletion in the EP300 locus, only a C-terminally truncated form of p300 is expressed in RC-K8 cells. These results suggest a role for p300 in REL-mediated oncogenic activity in B lymphoma.
Collapse
|
29
|
Chen F, Beezhold K, Castranova V. Tumor Promoting or Tumor Suppressing of NF-κ B, a Matter of Cell Context Dependency. Int Rev Immunol 2009; 27:183-204. [DOI: 10.1080/08830180802130327] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
30
|
Dahlman JM, Wang J, Bakkar N, Guttridge DC. The RelA/p65 subunit of NF-κB specifically regulates cyclin D1 protein stability: Implications for cell cycle withdrawal and skeletal myogenesis. J Cell Biochem 2009; 106:42-51. [DOI: 10.1002/jcb.21976] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
31
|
Penzo M, Massa PE, Olivotto E, Bianchi F, Borzi RM, Hanidu A, Li X, Li J, Marcu KB. Sustained NF-kappaB activation produces a short-term cell proliferation block in conjunction with repressing effectors of cell cycle progression controlled by E2F or FoxM1. J Cell Physiol 2008; 218:215-27. [PMID: 18803232 DOI: 10.1002/jcp.21596] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
NF-kappaB transcription factors induce a host of genes involved in pro-inflammatory/stress-like responses; but the collateral effects and consequences of sustained NF-kappaB activation on other cellular gene expression programming remain less well understood. Here enforced expression of a constitutively active IKKbeta T-loop mutant (IKKbetaca) drove murine fibroblasts into transient growth arrest that subsided within 2-3 weeks of continuous culture. Proliferation arrest was associated with a G1/S phase block in immortalized and primary early passage MEFs. Molecular analysis in immortalized MEFs revealed that inhibition of cell proliferation in the initial 1-2 weeks after their IKKbetaca retroviral infection was linked to the transient, concerted repression of essential cell cycle effectors that are known targets of either E2F or FoxM1. Co-expression of a phosphorylation resistant IkappaBalpha super repressor and IKKbetaca abrogated growth arrest and cell cycle effector repression, thereby linking IKKbetaca's effects to canonical NF-kappaB activation. Transient growth arrest of IKKbetaca cells was associated with enhanced p21 (cyclin-dependent kinase inhibitor 1A) protein expression, due in part to transcriptional activation by NF-kappaB and also likely due to strong repression of Skp2 and Csk1, both of which are FoxM1 direct targets mediating proteasomal dependent p21 turnover. Ablation of p21 in immortalized MEFs reduced their IKKbetaca mediated growth suppression. Moreover, trichostatin A inhibition of HDACs alleviated the repression of E2F and FoxM1 targets induced by IKKbetaca, suggesting chromatin mediated gene silencing in IKKbetaca's short term repressive effects on E2F and FoxM1 target gene expression.
Collapse
Affiliation(s)
- Marianna Penzo
- Centro Ricerca Biomedica Applicata (CRBA), S. Orsola-Malpighi University Hospital, University of Bologna, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abdel-Latif MMM, Kelleher D, Reynolds JV. Potential role of NF-kappaB in esophageal adenocarcinoma: as an emerging molecular target. J Surg Res 2008; 153:172-80. [PMID: 18533190 DOI: 10.1016/j.jss.2007.12.755] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 09/10/2007] [Accepted: 12/06/2007] [Indexed: 12/17/2022]
Abstract
Esophageal adenocarcinoma is increasing in incidence and arises in a background of reflux induced inflammation, metaplasia, and dysplasia. The proinflammatory transcription factor nuclear factor-kappa B (NF-kappaB) has a central role in inflammation and tumorigenesis. Because a role for NF-kappaB has been implicated in the pathogenesis of esophageal cancer, this transcription factor has been the focus of the current research of this devastating disease. NF-kappaB blocks apoptosis, mediates tumor cell proliferation, and induces resistance to chemotherapeutic drugs. Research efforts to improve the effect of chemotherapy have led to an improvement in patient survival but there is still a need for improvement, and NF-kappaB is a potential target for cancer drug development. In this review, we have attempted to highlight the possible role of NF-kappaB in esophageal adenocarcinoma and discuss the anticancer strategy with NF-kappaB as a promising molecular target in esophageal cancer therapy.
Collapse
Affiliation(s)
- Mohamed M M Abdel-Latif
- Department of Clinical Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Dublin 8, Ireland.
| | | | | |
Collapse
|
33
|
Fujita A, Sato JR, Garay-Malpartida HM, Morettin PA, Sogayar MC, Ferreira CE. Time-varying modeling of gene expression regulatory networks using the wavelet dynamic vector autoregressive method. ACTA ACUST UNITED AC 2007; 23:1623-30. [PMID: 17463021 DOI: 10.1093/bioinformatics/btm151] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
MOTIVATION A variety of biological cellular processes are achieved through a variety of extracellular regulators, signal transduction, protein-protein interactions and differential gene expression. Understanding of the mechanisms underlying these processes requires detailed molecular description of the protein and gene networks involved. To better understand these molecular networks, we propose a statistical method to estimate time-varying gene regulatory networks from time series microarray data. One well known problem when inferring connectivity in gene regulatory networks is the fact that the relationships found constitute correlations that do not allow inferring causation, for which, a priori biological knowledge is required. Moreover, it is also necessary to know the time period at which this causation occurs. Here, we present the Dynamic Vector Autoregressive model as a solution to these problems. RESULTS We have applied the Dynamic Vector Autoregressive model to estimate time-varying gene regulatory networks based on gene expression profiles obtained from microarray experiments. The network is determined entirely based on gene expression profiles data, without any prior biological knowledge. Through construction of three gene regulatory networks (of p53, NF-kappaB and c-myc) for HeLa cells, we were able to predict the connectivity, Granger-causality and dynamics of the information flow in these networks. SUPPLEMENTARY INFORMATION Additional figures may be found at http://mariwork.iq.usp.br/dvar/.
Collapse
Affiliation(s)
- A Fujita
- Institute of Mathematics and Statistics, University of São Paulo, Rua do Matão, 1010-São Paulo, 05508-090, SP, Brazil
| | | | | | | | | | | |
Collapse
|
34
|
Häussler U, von Wichert G, Schmid RM, Keller F, Schneider G. Epidermal growth factor activates nuclear factor-κB in human proximal tubule cells. Am J Physiol Renal Physiol 2005; 289:F808-15. [PMID: 15798085 DOI: 10.1152/ajprenal.00434.2003] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The promotion of cell survival and regeneration in acute renal failure (ARF) is important for the restitution of renal function. Epidermal growth factor (EGF) has been implicated in the regulation of cell proliferation. We provide evidence for a direct link between EGF, nuclear factor-κB (NF-κB), and cell cycle regulation (cyclin D1). EGF was found to stimulate NF-κB-dependent gene transcription and DNA binding. In addition, EGF stimulated cyclin D1 promoter activity as well as cyclin D1 expression. Moreover, inhibition of NF-κB caused a pronounced reduction of EGF-induced cyclin D1 promoter activity. Furthermore, both EGF-mediated NF-κB activation and cyclin D1 expression were inhibited by coexpression of super IκB. Taken together, these data identify NF-κB and cyclin D1 as downstream targets of EGF and establish a molecular link between stimulation of EGF via activation of NF-κB and cyclin D1 expression in human proximal tubular cells.
Collapse
Affiliation(s)
- Ulla Häussler
- Nephrology Div., Internal Medicine II, Univ. of Ulm, Robert-Koch-Strasse 8, D-89081 Ulm, Germany.
| | | | | | | | | |
Collapse
|
35
|
Abstract
Nuclear Factor-kappa B (NF-kappa B) is an inducible transcription factor that regulates the expression of many genes involved in the immune response. Recently, NF-kappa B activity has been shown to be upregulated in many cancers, including melanoma. Data indicate that the enhanced activation of NF-kappa B may be due to deregulations in upstream signaling pathways such as Ras/Raf, PI3K/Akt, and NIK. Multiple studies have shown that NF-kappa B is involved in the regulation of apoptosis, angiogenesis, and tumor cell invasion, all of which indicate the important role of NF-kappa B in tumorigenesis. Thus, understanding the molecular mechanism of melanoma progression will aid in designing new therapeutic approaches for melanoma. In this review, the association between NF-kappa B and melanoma tumorigenesis are discussed. Additionally, the potential of emerging selective NF-kappa B inhibitors for the treatment of melanoma is reviewed.
Collapse
|
36
|
Starczynowski DT, Reynolds JG, Gilmore TD. Mutations of tumor necrosis factor α-responsive serine residues within the C-terminal transactivation domain of human transcription factor REL enhance its in vitro transforming ability. Oncogene 2005; 24:7355-68. [PMID: 16027730 DOI: 10.1038/sj.onc.1208902] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The human c-rel gene (REL), encoding an NF-kappaB transcription factor, is amplified or mutated in several human B-cell lymphomas and can transform chicken lymphoid cells in vitro. We have previously shown that certain deletions of C-terminal transactivation sequences enhance REL's transforming ability in chicken spleen cells. In this report, we have analysed the effect of single amino-acid changes at select serine residues in the C-terminal transactivation domain on REL's transforming ability. Mutation of either of two TNFalpha-inducible serine residues (Ser460 and Ser471) to nonphosphorylatable residues (alanine, asparagine, phenylalanine) made REL more efficient at transforming chicken spleen cells in vitro. In contrast, mutation of Ser471 to a phosphorylation mimetic aspartate residue impaired REL's transforming ability, even though it increased REL's inherent transactivation ability as a GAL4-fusion protein. Alanine mutations of several other serine residues within the transactivation domain did not substantially affect REL's transforming ability. Transactivation by GAL4-REL fusion proteins containing either transformation enhancing or nonenhancing mutations at serine residues was generally similar to wild-type GAL4-REL. However, more transforming mutants with mutations at either Ser460 or Ser471 differed from wild-type REL in their ability to transactivate certain kappaB-site reporter genes. In particular, the SOD2 promoter, encoding manganese superoxide dismutase, was activated less strongly by the more transforming REL mutant REL-S471N in transient assays, but REL-S471N-transformed chicken spleen cells had increased levels of MnSOD protein as compared to wild-type REL-transformed cells. Taken together, our results show that mutations of certain serine residues can enhance REL's transforming ability in vitro and suggest that these mutations increase REL-mediated transformation by altering REL's ability to modulate the expression of select target genes. Furthermore, phosphorylation of Ser471 may be involved in REL-mediated modulation of transformation-specific target gene expression. Lastly, these results suggest that similar mutations in the REL transactivation domain contribute to the development of certain human B-cell lymphomas.
Collapse
|
37
|
Dolcet X, Llobet D, Pallares J, Matias-Guiu X. NF-kB in development and progression of human cancer. Virchows Arch 2005; 446:475-82. [PMID: 15856292 DOI: 10.1007/s00428-005-1264-9] [Citation(s) in RCA: 867] [Impact Index Per Article: 43.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2005] [Accepted: 03/30/2005] [Indexed: 02/07/2023]
Abstract
The nuclear factor kB (NF-kB) comprises a family of transcription factors involved in the regulation of a wide variety of biological responses. NF-kB plays a well-known function in the regulation of immune responses and inflammation, but growing evidences support a major role in oncogenesis. NF-kB regulates the expression of genes involved in many processes that play a key role in the development and progression of cancer such as proliferation, migration and apoptosis. Aberrant or constitutive NF-kB activation has been detected in many human malignancies. In recent years, numerous studies have focused on elucidating the functional consequences of NF-kB activation as well as its signaling mechanisms. NF-kB has turned out to be an interesting therapeutic target for treatment of cancer.
Collapse
Affiliation(s)
- Xavier Dolcet
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, Av Alcalde Rovira Roure 80, 25198, Lleida, Spain
| | | | | | | |
Collapse
|
38
|
Alexia C, Fallot G, Lasfer M, Schweizer-Groyer G, Groyer A. An evaluation of the role of insulin-like growth factors (IGF) and of type-I IGF receptor signalling in hepatocarcinogenesis and in the resistance of hepatocarcinoma cells against drug-induced apoptosis. Biochem Pharmacol 2004; 68:1003-15. [PMID: 15313394 DOI: 10.1016/j.bcp.2004.05.029] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2004] [Accepted: 05/10/2004] [Indexed: 01/18/2023]
Abstract
Strong evidence emphasizes the role of the insulin-like growth factor (IGF) system and of type-I IGF receptor (IGF-IR) signalling in tumourigenesis. In this connection: (i) changes in the expression pattern of components of the IGF system (autocrine/paracrine expression of IGF-I and -II, overexpression of IGF-IR, decreased expression of IGF-binding proteins (IGFBPs) and of type-II IGF receptor/cation-independent mannose-6-phosphate receptor (IGF-II/M6PR) and (ii) increased serum concentrations of proteases that cleave the IGFBPs (e.g., cathepsin D) were observed in patients with hepatocellular carcinomas (HCC), in human hepatoma cell lines and in their conditioned culture medium, as well as in rodent models of hepatocarcinogenesis. Accordingly, studies carried out with animal models do suggest that the IGF system and IGF-IR signalling may play a role in hepatocarcinogenesis and in deregulated proliferation and apoptosis of HCC cells. Finally the instrumental role of Raf/MEK/ERK, one of the signalling cascades stimulated by IGF-IR, in anthracycline-induced apoptosis of HepG2 and Huh-7 human hepatoma cell lines emphasizes that care must be taken when designing combinations of antitumoural molecules for antineoplastic treatment. This review addresses the putative roles of the IGF system in primary HCC, with a special focus on the underlying molecular mechanisms. In a second part it emphasizes the putative interference of IGF-IR signalling with chemotherapeutic drug-induced apoptosis in human hepatoma cells.
Collapse
Affiliation(s)
- Catherine Alexia
- Inserm U.481, Faculté de Médecine Xavier Bichat, 16 rue Henri Huchard, BP416, 75870 Paris Cédex 18, France
| | | | | | | | | |
Collapse
|
39
|
Chandler NM, Canete JJ, Callery MP. Increased expression of NF-kappa B subunits in human pancreatic cancer cells. J Surg Res 2004; 118:9-14. [PMID: 15093710 DOI: 10.1016/s0022-4804(03)00354-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2003] [Indexed: 01/03/2023]
Abstract
BACKGROUND Activation of NF-kappa B-dependent antiapoptotic genes may factor in the chemoresistance of pancreatic cancer. It is not known whether NF-kappa B subunit composition changes during oncogenesis and regulates overall NF-kappa B activation. We compared the relative expression of NF-kappa B subunits with nuclear activation of p65 between variably differentiated pancreatic cancer cells. MATERIALS AND METHODS Proliferating human pancreatic cancer cells (PANC-1, BxPC-3) and nonmalignant intestinal cells (FHS 74 Int) were harvested. Baseline expression of NF-kappa B subunits (p65, p52, p50, c-Rel) and its inhibitor I kappa B-alpha were determined by Western blot. Nuclear NF-kappa B p65 activity was measured by ELISA. Results were analyzed by ANOVA (P < 0.05) and Tukey's HSD for pairwise comparisons when appropriate (P < 0.05). RESULTS Constitutive expression of NF-kappa B subunits was detected in proliferating, intestinal cells (FHS 74 Int). Both cytoplasmic (I kappa B-alpha, p50, p52, p65) and nuclear (p50, p52, p65, c-Rel) NF-kappa B subunits were significantly increased in both PANC-1 and BxPC-3 cells compared to FHS 74 Int. While nuclear p65 subunit levels were similarly elevated, actual p65 activity was only significantly greater in PANC-1 cells compared to either BxPC-3 or FHS 74 Int (P < 0.05). CONCLUSIONS Compared to nonmalignant proliferating intestinal cells, these pancreatic cancer cell lines have increased levels of NF-kappa B subunits. Actual nuclear NF-kappa B activity, however, appears to correlate more with degree of tumor differentiation than with NF-kappa B subunit expression.
Collapse
Affiliation(s)
- Nicole M Chandler
- Department of Surgery, University of Massachusetts Medical School, Worcester, MA, USA.
| | | | | |
Collapse
|
40
|
Abstract
Senescence is now established as a genetically controlled phenomenon that alters different cell functions, including proliferation, apoptosis, resistance to stress, and energetic metabolism. Underlying changes in gene expression are governed by some transcription factors, whose expression or activity must change with senescence as well. Transcription factors of the Rel/NF-kappa B family are good candidates to participate in the establishment of senescence. Arguments range from correlation between cell functions controlled by these factors and cell functions altered during senescence, to phenotypes resulting from in vitro manipulations of Rel/NF-kappa B activity.
Collapse
Affiliation(s)
- Karo Gosselin
- UMR 8117 CNRS-Institut Pasteur de Lille-Université Lille 1, Institut de Biologie de Lille, 1 rue Calmette, BP 447, 59021 Lille Cedex, France
| | | |
Collapse
|
41
|
Panwalkar A, Verstovsek S, Giles F. Nuclear factor-kappaB modulation as a therapeutic approach in hematologic malignancies. Cancer 2004; 100:1578-89. [PMID: 15073843 DOI: 10.1002/cncr.20182] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nuclear factor-kappaB (NF-kappaB) is a collective term that refers to a small class of dimeric transcription factors for a number of genes, including growth factors, angiogenesis modulators, cell-adhesion molecules, and antiapoptotic factors. Although most NF-kappaB proteins promote transcription, some act as inactivating or repressive complexes. The most common p50-RelA (p65) dimer known "specifically" as NF-kappaB, is relatively abundant, controls the expression of numerous genes, and exists as an inactive cytoplasmic complex bound to inhibitory proteins of the NF-kappaB inhibitor (IkappaB) family. The inactive NF-kappaB-IkappaB complex is activated by a variety of stimuli, including proinflammatory cytokines, mitogens, growth factors, and stress-inducing agents. The release of NF-kappaB facilitates its translocation to the nucleus, where it promotes cell survival by initiating the transcription of genes encoding stress-response enzymes, cell-adhesion molecules, proinflammatory cytokines, and antiapoptotic proteins. Constitutive activation of NF-kappaB in the nucleus is observed in some hematologic disorders. With the recent approval of bortezomib for patients with advanced multiple myeloma, NF-kappaB modulation is likely to be a therapeutic endeavor of increasing interest in coming years.
Collapse
Affiliation(s)
- Amit Panwalkar
- Section of Developmental Therapeutics, Department of Leukemia, The University of Texas, M. D. Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | |
Collapse
|
42
|
Kucharczak J, Simmons MJ, Fan Y, Gélinas C. To be, or not to be: NF-kappaB is the answer--role of Rel/NF-kappaB in the regulation of apoptosis. Oncogene 2004; 22:8961-82. [PMID: 14663476 DOI: 10.1038/sj.onc.1207230] [Citation(s) in RCA: 592] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
During their lifetime, cells encounter many life or death situations that challenge their very own existence. Their survival depends on the interplay within a complex yet precisely orchestrated network of proteins. The Rel/NF-kappaB signaling pathway and the transcription factors that it activates have emerged as critical regulators of the apoptotic response. These proteins are best known for the key roles that they play in normal immune and inflammatory responses, but they are also implicated in the control of cell proliferation, differentiation, apoptosis and oncogenesis. In recent years, there has been remarkable progress in understanding the pathways that activate the Rel/NF-kappaB factors and their role in the cell's decision to either fight or surrender to apoptotic challenge. Whereas NF-kappaB is most commonly involved in suppressing apoptosis by transactivating the expression of antiapoptotic genes, it can promote programmed cell death in response to certain death-inducing signals and in certain cell types. This review surveys our current understanding of the role of NF-kappaB in the apoptotic response and focuses on many developments since this topic was last reviewed in Oncogene 4 years ago. These recent findings shed new light on the activity of NF-kappaB as a critical regulator of apoptosis in the immune, hepatic, epidermal and nervous systems, on the mechanisms through which it operates and on its role in tissue development, homoeostasis and cancer.
Collapse
Affiliation(s)
- Jérôme Kucharczak
- Center for Advanced Biotechnology and Medicine, 679 Hoes Lane, Piscataway, NJ, USA
| | | | | | | |
Collapse
|
43
|
Cheng S, Hsia CY, Leone G, Liou HC. Cyclin E and Bcl-xL cooperatively induce cell cycle progression in c-Rel−/− B cells. Oncogene 2003; 22:8472-86. [PMID: 14627988 DOI: 10.1038/sj.onc.1206917] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Aberrant overexpression of the c-rel protooncogene is associated with lymphoid malignancy, while c-rel deletion produces severe lymphoproliferative defects and immunodeficiency. To investigate the mechanism of c-rel-induced proliferation and cell cycle progression in B lymphocytes, we have compared signaling events elicited through the BCR in c-rel-/- and wild-type B cells. BCR stimulation of c-rel-/- B cells fails to induce proper cyclin expression, resulting in G1 phase arrest, but it is unclear whether these defects are in fact secondary events of decreased B-cell survival, since c-rel deletion also affects the expression of antiapoptotic genes such as bcl-xL. Here, we use the bcl-xL transgene to correct the viability of c-rel-deficient B cells, and show that the inhibition of apoptosis does not necessarily confer hyperproliferation of B cells activated through the BCR. c-rel-/- B cells still fail to enter the S phase despite improved survival by bcl-xL overexpression, suggesting that c-Rel-associated cell cycle progression is dependent on more than just enhanced cell viability. Overexpression of cyclin E protein, however, can cooperate with Bcl-xL to restore cell cycle progression to c-rel-/- B cells via induction of the cyclin-CDK/Rb-E2F pathway. Furthermore, we show that c-Rel can directly regulate transcription of the e2f3a promoter/enhancer, which is then likely to lead to transcriptional activation of the cyclin E promoter by E2F3a. Hence, these studies provide clear evidence that control of lymphocyte proliferation via c-Rel is linked to a cyclin-dependent process, and suggest that c-Rel not only activates antiapoptotic signaling but also the induction of cell cycle progression.
Collapse
Affiliation(s)
- Shuhua Cheng
- Division of Immunology, Department of Medicine, Weill Medical College of Cornell University, New York, NY 10021, USA
| | | | | | | |
Collapse
|
44
|
Gilmore TD, Jean-Jacques J, Richards R, Cormier C, Kim J, Kalaitzidis D. Stable expression of the avian retroviral oncoprotein v-Rel in avian, mouse, and dog cell lines. Virology 2003; 316:9-16. [PMID: 14599786 DOI: 10.1016/s0042-6822(03)00562-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Overexpression of the retroviral oncoprotein v-Rel can rapidly transform and immortalize a variety of avian cells in culture. However, mammalian models for v-Rel-mediated oncogenesis have been compromised by the fact that high-level expression of v-Rel has been reported to be toxic in many mammalian cell types, including mouse 3T3 cells, Rat-1 cells, and mouse bone marrow cells. In this article, we demonstrate that 3T3 cells can support expression of v-Rel for at least 24 days when infected with a mouse stem cell virus (MSCV) retroviral vector containing v-rel. In retrovirus-infected 3T3 cells, v-Rel is located in the nucleus and can bind to DNA, but does not transform the cells. On the other hand, 3T3 and Rat-2 cells do not express v-Rel after stable transfection with a pcDNA-based v-Rel expression vector. We also show that infection of the IL3-dependent mouse B cell line BaF3 with the MSCV-v-rel vector results in expression of v-Rel, but does not convert these cells to growth factor independence. In contrast to 3T3 cells, the dog osteosarcoma D17 cell line can support a high level of v-Rel expression, after either transfection or infection with a retroviral vector. That is, v-Rel can be stably expressed as a nuclear, DNA-binding protein in D17 cells to approximately the same level as in chicken embryo fibroblasts. These results suggest that the restriction to v-Rel expression in rodent fibroblasts is generally absent in D17 cells and that the type of v-rel expression vector determines whether 3T3 cells can support stable expression of v-Rel. The findings reported here are an essential first step in the development of mammalian systems to study Rel-mediated oncogenesis.
Collapse
Affiliation(s)
- Thomas D Gilmore
- Biology Department, Boston University, 5 Cummington Street, Boston, MA 02215, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Xia HHX, Wong BCY. Nitric oxide in Helicobacter pylori-induced apoptosis and its significance in gastric carcinogenesis. J Gastroenterol Hepatol 2003; 18:1227-1230. [PMID: 14535977 DOI: 10.1046/j.1440-1746.2003.03147.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
46
|
Algül H, Adler G, Schmid RM. NF-kappaB/Rel transcriptional pathway: implications in pancreatic cancer. INTERNATIONAL JOURNAL OF GASTROINTESTINAL CANCER 2003; 31:71-8. [PMID: 12622417 DOI: 10.1385/ijgc:31:1-3:71] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite considerable efforts in understanding the cellular mechanisms contributing to pancreatic cancer, the prognosis of this malignant disease is still extremely poor. Although pancreatic cancer is the fifth common cause of cancer death in Western countries, current options in treatment enable a 5-yr survival rate for all stages of less than 5%. In the face fo the fatal outcome, new approaches to the therapy have been established. Based on its role in malignant transformation, apoptosis, and cell proliferation, the transcription factor NF-kappaB/Rel has gained the attention of many laboratories. This review provides basic information for the understanding of the biology of NF-kappaB and aims at presenting experimental data illustrating the involvement of NF-kappaB/Rel in pancreatic cancer.
Collapse
Affiliation(s)
- Hana Algül
- Department of Internal Medicine I, University of Ulm, Germany
| | | | | |
Collapse
|
47
|
Hinata K, Gervin AM, Jennifer Zhang Y, Khavari PA. Divergent gene regulation and growth effects by NF-kappa B in epithelial and mesenchymal cells of human skin. Oncogene 2003; 22:1955-64. [PMID: 12673201 DOI: 10.1038/sj.onc.1206198] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
NF-kappa B regulates normal and pathological processes, including neoplasia, in a tissue-context-dependent manner. In skin, NF-kappa B is implicated in epidermal homeostasis as well as in the pathogenesis of squamous cell carcinoma; however, its function in the underlying mesenchymal dermis has been unclear. To gain insight into NF-kappa B roles in these two adjacent cutaneous tissue compartments, NF-kappa B effects on expression of 12 435 genes were determined in epidermal keratinocytes and dermal fibroblasts. Although NF-kappa B induced proinflammatory and antiapoptotic genes in both settings, it exhibited divergent effects on growth regulatory genes. In keratinocytes, but not in fibroblasts, NF-kappa B induced p21(CIP1), which was sufficient to inhibit growth of both cell types. Levels of growth inhibitory factor (GIF), in contrast, were increased by NF-kappa B in both settings but inhibited growth only in keratinocytes. These findings indicate that transcription factors such as NF-kappa B can program tissue-selective effects via both differential target gene induction as well as by inducing common targets that exert differing effects depending on cellular lineage.
Collapse
Affiliation(s)
- Kaede Hinata
- Program in Epithelial Biology, Stanford University School of Medicine, CA 94305, USA
| | | | | | | |
Collapse
|
48
|
Edelstein LC, Lagos L, Simmons M, Tirumalai H, Gélinas C. NF-kappa B-dependent assembly of an enhanceosome-like complex on the promoter region of apoptosis inhibitor Bfl-1/A1. Mol Cell Biol 2003; 23:2749-61. [PMID: 12665576 PMCID: PMC152543 DOI: 10.1128/mcb.23.8.2749-2761.2003] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Expression of the prosurvival Bcl-2 homologue Bfl-1/A1 is induced by NF-kappa B-activating stimuli, while B and T cells from c-rel knockout mice show an absolute defect in bfl-1/a1 gene activation. Here, we demonstrate NF-kappa B-dependent assembly of an enhanceosome-like complex on the promoter region of bfl-1. Binding of NF-kappa B subunit c-Rel to DNA nucleated the concerted binding of transcription factors AP-1 and C/EBP beta to the 5'-regulatory region of bfl-1. Optimal stability of the complex was dependent on proper orientation and phasing of the NF-kappa B site. Chromatin immunoprecipitation analyses demonstrated that T-cell activation triggers in vivo binding of endogenous c-Rel, c-Jun, C/EBP beta, and HMG-IC to the bfl-1 regulatory region, coincident with selective recruitment of coactivators TAFII250 and p300, SWI/SNF chromatin remodeling factor component BRG-1, and basal transcription factors TATA-binding protein (TBP) and TFIIB, as well as hyperacetylation of histones H3 and H4. These results highlight a critical role for NF-kappa B in bfl-1 transcription and point to the need for a complex and precise regulatory network to control bfl-1 expression. To our knowledge, this is the first demonstration of enhanceosome-mediated regulation of a cell death inhibitor.
Collapse
Affiliation(s)
- Leonard C Edelstein
- Center for Advanced Biotechnology and Medicine and Graduate Program in Biotechnology, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, New Jersey 08854, USA
| | | | | | | | | |
Collapse
|
49
|
van Hogerlinden M, Auer G, Toftgård R. Inhibition of Rel/Nuclear Factor-kappaB signaling in skin results in defective DNA damage-induced cell cycle arrest and Ha-ras- and p53-independent tumor development. Oncogene 2002; 21:4969-77. [PMID: 12118375 DOI: 10.1038/sj.onc.1205620] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2002] [Revised: 04/12/2002] [Accepted: 04/26/2002] [Indexed: 11/08/2022]
Abstract
In recent years a growth inhibitory role in skin for the Rel/NF-kappaB transcription factors has been established, and the block of Rel/NF-kappaB signaling results in rapid development of spontaneous skin cancer. The molecular mechanism underlying tumor development is however unknown. In the present study, we show that inhibition of NF-kappaB signaling in mouse skin by targeted expression of degradation resistant IkappaB-alpha generates transgenic keratinocytes unable to arrest the cell cycle in response to DNA damage induced by gamma-radiation. The results indicate that transgenic keratinocytes have a defect at the G1-S checkpoint whereas the G2-M checkpoint response was found to be intact. However, transgenic keratinocytes still respond by induction of the cyclin dependent kinase inhibitor p21(Cip1/Waf) after exposure to gamma-radiation. In the spontaneous skin tumors that develop in transgenic mice no mutations were found in the Ha-ras or p53 gene, suggesting that inhibition of NF-kappaB signaling in skin can induce cancer development independently of initiating mutations in the Ha-ras gene or additional mutations in the p53 gene. These findings demonstrate an involvement of NF-kappaB signaling in the DNA damage response and cell cycle checkpoint control in the skin.
Collapse
Affiliation(s)
- Max van Hogerlinden
- Department of Bioscience at Novum, Karolinska Institutet, NOVUM, SE-141 57 Huddinge, Sweden
| | | | | |
Collapse
|
50
|
Bernard D, Monte D, Vandenbunder B, Abbadie C. The c-Rel transcription factor can both induce and inhibit apoptosis in the same cells via the upregulation of MnSOD. Oncogene 2002; 21:4392-402. [PMID: 12080470 DOI: 10.1038/sj.onc.1205536] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2001] [Revised: 03/11/2002] [Accepted: 03/27/2002] [Indexed: 12/24/2022]
Abstract
Rel/NF-kappaB transcription factors are involved in several physiological processes, including the regulation of apoptosis. These factors were shown to exhibit pro- or anti-apoptotic activities in different cellular models, but at present, the mechanisms underlying these opposite effects are poorly understood. In this study, we show that the constitutive expression of a transcriptionally active member of the Rel/NF-kappaB family, c-Rel, first induces a resistance against TNFalpha-induced apoptosis and later increases the level of spontaneous apoptosis of HeLa cells. Both the anti- and pro-apoptotic effects increase with the level of c-Rel overexpression. The up-regulation by c-Rel of the manganese superoxide dismutase (MnSOD) could explain both the rapid anti-apoptotic effect and the delayed pro-apoptotic one. Indeed, the enzymatic activity of MnSOD is to transform the toxic O(2)(*)(-) in H(2)O(2). Hence, on one hand, its induction helps cells to resist against the apoptogenic burst of O(2)(*)(-) produced upon TNFalpha stimulation, but on the other hand, it leads to a progressive H(2)O(2) accumulation that ultimately results in apoptosis. These results indicate that the anti- and pro-apoptotic effects of Rel/NF-kappaB factors are not necessarily alternative but can occur successively in the same cell, via the up-regulation of the same target gene.
Collapse
Affiliation(s)
- David Bernard
- FRE 2353 CNRS/Institut Pasteur de Lille/Université Lille 2, Institut de Biologie de Lille, 1 rue Calmette, 59021 Lille Cedex, France
| | | | | | | |
Collapse
|