51
|
Koschmieder S, Chatain N. Role of inflammation in the biology of myeloproliferative neoplasms. Blood Rev 2020; 42:100711. [PMID: 32505517 DOI: 10.1016/j.blre.2020.100711] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/11/2019] [Accepted: 05/04/2020] [Indexed: 12/20/2022]
Abstract
What is the role of inflammation in Myeloproliferative Neoplasms? This is currently a topic of much debate. In this review, we will discuss experimental results and basic concepts of inflammatory processes in the pathogenesis of myeloproliferative neoplasms (MPN). So, which are the players involved? First, these are the clonal hematopoietic stem cells (HSC) and their normal stem cell counterparts in the bone marrow (BM), as well as their more mature progeny in the BM and the peripheral blood (PB), including neutrophils, macrophages, erythrocytes, and platelets, but also other cell lineages. Second, these cells produce a plethora of inflammatory cytokines, such as interleukin 6 (IL6), interleukin 8 (IL8), TNF-alpha (TNFa), interferon-alpha (IFNa), and others. Third, these cells and cytokines act in concert with non-hematopoietic cells, including endothelial cells and mesenchymal stromal cells (MSCs). The latter cells, in particular GLI1 positive or leptin receptor (LepR) positive stromal cells, may become activated by the hematopoietic clone to give rise to myofibroblasts, producing excessive fibrosis in the bone marrow (myelofibrosis). Ultimately, the inflammatory and fibrotic circuit involving these three key players may lead to progression of the disease, resulting in BM failure and transformation into acute leukemia, also termed blast crisis. Here, we review the role of these three effectors in the pathogenesis of MPN.
Collapse
Affiliation(s)
- Steffen Koschmieder
- Department of Hematology, Oncology, Hemostaselogy, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Nicolas Chatain
- Department of Hematology, Oncology, Hemostaselogy, and Stem Cell Transplantation, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
52
|
17-Aminogeldanamycin Inhibits Constitutive Nuclear Factor-Kappa B (NF-κB) Activity in Patient-Derived Melanoma Cell Lines. Int J Mol Sci 2020; 21:ijms21113749. [PMID: 32466509 PMCID: PMC7312877 DOI: 10.3390/ijms21113749] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022] Open
Abstract
Melanoma remains incurable skin cancer, and targeting heat shock protein 90 (HSP90) is a promising therapeutic approach. In this study, we investigate the effect of 17-aminogeldanamycin, a potent HSP90 inhibitor, on nuclear factor-kappa B (NF-κB) activity in BRAFV600E and NRASQ61R patient-derived melanoma cell lines. We performed time-lapse microscopy and flow cytometry to monitor changes in cell confluence and viability. The NF-κB activity was determined by immunodetection of phospho-p65 and assessment of expression of NF-κB-dependent genes by quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA). Constitutive activity of p65/NF-κB was evident in all melanoma cell lines. Differences in its level might be associated with genetic alterations in CHUK, IL1B, MAP3K14, NFKBIE, RIPK1, and TLR4, while differences in transcript levels of NF-κB-inducible genes revealed by PCR array might result from the contribution of other regulatory mechanisms. 17-Aminogeldanamycin markedly diminished the level of phospho-p65, but the total p65 protein level was unaltered, indicating that 17-aminogeldanamycin inhibited activation of p65/NF-κB. This conclusion was supported by significantly reduced expression of selected NF-κB-dependent genes: cyclin D1 (CCND1), C-X-C motif chemokine ligand 8 (CXCL8), and vascular endothelial growth factor (VEGF), as shown at transcript and protein levels, as well as secretion of IL-8 and VEGF. Our study indicates that 17-aminogeldanamycin can be used for efficient inhibition of NF-κB activity and the simultaneous diminution of IL-8 and VEGF levels in the extracellular milieu of melanoma.
Collapse
|
53
|
De Dominici M, Porazzi P, Xiao Y, Chao A, Tang HY, Kumar G, Fortina P, Spinelli O, Rambaldi A, Peterson LF, Petruk S, Barletta C, Mazo A, Cingolani G, Salvino JM, Calabretta B. Selective inhibition of Ph-positive ALL cell growth through kinase-dependent and -independent effects by CDK6-specific PROTACs. Blood 2020; 135:1560-1573. [PMID: 32040545 PMCID: PMC7193186 DOI: 10.1182/blood.2019003604] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/23/2020] [Indexed: 12/13/2022] Open
Abstract
Expression of the cell cycle regulatory gene CDK6 is required for Philadelphia-positive (Ph+) acute lymphoblastic leukemia (ALL) cell growth, whereas expression of the closely related CDK4 protein is dispensable. Moreover, CDK6 silencing is more effective than treatment with the dual CDK4/6 inhibitor palbociclib in suppressing Ph+ ALL in mice, suggesting that the growth-promoting effects of CDK6 are, in part, kinase-independent in Ph+ ALL. Accordingly, we developed CDK4/6-targeted proteolysis-targeting chimeras (PROTACs) that inhibit CDK6 enzymatic activity in vitro, promote the rapid and preferential degradation of CDK6 over CDK4 in Ph+ ALL cells, and markedly suppress S-phase cells concomitant with inhibition of CDK6-regulated phospho-RB and FOXM1 expression. No such effects were observed in CD34+ normal hematopoietic progenitors, although CDK6 was efficiently degraded. Treatment with the CDK6-degrading PROTAC YX-2-107 markedly suppressed leukemia burden in mice injected with de novo or tyrosine kinase inhibitor-resistant primary Ph+ ALL cells, and this effect was comparable or superior to that of the CDK4/6 enzymatic inhibitor palbociclib. These studies provide "proof of principle" that targeting CDK6 with PROTACs that inhibit its enzymatic activity and promote its degradation represents an effective strategy to exploit the "CDK6 dependence" of Ph+ ALL and, perhaps, of other hematologic malignancies. Moreover, they suggest that treatment of Ph+ ALL with CDK6-selective PROTACs would spare a high proportion of normal hematopoietic progenitors, preventing the neutropenia induced by treatment with dual CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Marco De Dominici
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Patrizia Porazzi
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | | | | | - Gaurav Kumar
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Paolo Fortina
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Orietta Spinelli
- Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo, Italy
| | - Alessandro Rambaldi
- Hematology and Bone Marrow Transplant Unit, Ospedale Papa Giovanni XXIII, Bergamo, Italy
- Department of Oncology and Hematology-Oncology, Università Statale Milano, Milan, Italy
| | - Luke F Peterson
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI; and
| | - Svetlana Petruk
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Camilla Barletta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Alexander Mazo
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Gino Cingolani
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | | | - Bruno Calabretta
- Department of Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
54
|
Uras IZ, Sexl V, Kollmann K. CDK6 Inhibition: A Novel Approach in AML Management. Int J Mol Sci 2020; 21:ijms21072528. [PMID: 32260549 PMCID: PMC7178035 DOI: 10.3390/ijms21072528] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 02/01/2023] Open
Abstract
Acute myeloid leukemia (AML) is a complex disease with an aggressive clinical course and high mortality rate. The standard of care for patients has only changed minimally over the past 40 years. However, potentially useful agents have moved from bench to bedside with the potential to revolutionize therapeutic strategies. As such, cell-cycle inhibitors have been discussed as alternative treatment options for AML. In this review, we focus on cyclin-dependent kinase 6 (CDK6) emerging as a key molecule with distinct functions in different subsets of AML. CDK6 exerts its effects in a kinase-dependent and -independent manner which is of clinical significance as current inhibitors only target the enzymatic activity.
Collapse
Affiliation(s)
- Iris Z. Uras
- Department of Pharmacology, Center of Physiology and Pharmacology & Comprehensive Cancer Center (CCC), Medical University of Vienna, 1090 Vienna, Austria;
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria;
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, 1210 Vienna, Austria;
- Correspondence: ; Tel.: + 43-1-25077-2917
| |
Collapse
|
55
|
Qi R, Li X, Zhang X, Huang Y, Fei Q, Han Y, Cai R, Gao Y, Qi Y. Ethanol extract of Elephantopus scaber Linn. Attenuates inflammatory response via the inhibition of NF-κB signaling by dampening p65-DNA binding activity in lipopolysaccharide-activated macrophages. JOURNAL OF ETHNOPHARMACOLOGY 2020; 250:112499. [PMID: 31877363 DOI: 10.1016/j.jep.2019.112499] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Elephantopus scaber Linn. (E.scaber) is a widely-used traditional herb whose use has been documented for various inflammatory diseases such as fever, sore throat, dysentery, carbuncle and so on. However, the effect and mechanism of E.scaber in LPS-activated macrophages remain unclear. AIM This study aims to investigate the anti-inflammatory mechanism of the ethanol extract of E.scaber (ESE) in lipopolysaccharide (LPS)-induced inflammatory models. MATERIALS AND METHODS Griess reagent was used to determine NO production, and the levels of TNF-α, IL-6, MCP-1 and IL-1β were determined by ELISA kits. The molecular mechanism research was performed by RT-PCR, Western blot, and electrophoretic mobility shift assay (EMSA). LPS-induced endotoxemia mouse model was used for evaluating the in vivo anti-inflammatory action of ESE. RESULTS ESE suppressed LPS-induced iNOS, TNF-α, IL-6, MCP-1 and IL-1β transcription as well as supernatant NO, TNF-α, IL-6, MCP-1 and IL-1β production in macrophages. Although ESE inhibited NF-κB activation, it did not affect the IκBα phosphorylation and degradation and the NF-κB p65 nuclear translocation. The result of EMSA revealed that ESE inhibited the NF-κB p65-DNA binding activity. Additionally, ESE also decreased the proinflammatory cytokines in serum and peritoneal lavage fluid of LPS-induced endotoxemic mice. CONCLUSION ESE has a potently anti-inflammatory effect through inhibiting the NF-κB p65-DNA binding activity in LPS-activated macrophages.
Collapse
Affiliation(s)
- Ruijuan Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Ximeng Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Xiaoyu Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yunfeng Huang
- Guangxi Institute of Chinese Medicine & Pharmaceutical Science, Nanning, 530022, Guangxi, China.
| | - Qiaoling Fei
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yixin Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Runlan Cai
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yuan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Yun Qi
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
56
|
Li X, Liu C, Zhao X, Wang R, Gu N, Shen H, Li X, Wang L, Li C. Effects of CDK6 regulated by miR-298 on proliferation and apoptosis of thyroid cancer cells. Oncol Lett 2020; 19:2909-2915. [PMID: 32218846 PMCID: PMC7068225 DOI: 10.3892/ol.2020.11398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/10/2020] [Indexed: 12/01/2022] Open
Abstract
Effects of CDK6 regulated by miR-298 on proliferation and apoptosis of thyroid cancer cells were explored. Seventy-five cases of thyroid carcinoma and adjacent tissues were collected. The expression levels of miR-298 and CDK6 mRNA in tissues and cells were detected by RT-PCR. In addition, thyroid cancer cells and human normal thyroid cells Nthy-ori3-1 were purchased, with the former transfected with miR-298-mimics, miR-298-inhibitor, miR-NC, si-CDK6, si-NC, Sh-CDK6, Sh-NC to build cell models. Then the expression levels of miR-298 and CDK6 in thyroid cancer tissues and cells were detected by qRT-PCR, and the expression of CDK6, Bax, Bcl-2 and caspase-3 by WB. CCK-8 and flow cytometry were employed to detect cell proliferation and apoptosis, and dual luciferase report was adopted to determine the relationship between miR-298 and CDK6. miR-298 was underexpressed in thyroid cancer, and CDK6 was highly expressed in thyroid cancer. Cell experiments revealed that overexpression of miR-298 or inhibition of CDK6 expression could suppress cell proliferation, promote apoptosis, and significantly increase the expression levels of Bax and caspase-3 proteins, decrease Bcl-2 protein expression, which was contrary to the biological phenotype of cells after inhibition of miR-298 or further overexpression of CDK6. Dual luciferase report confirmed that miR-298 was a targeting site of CDK6. miR-298 can inhibit the proliferation of thyroid cells and promote apoptosis of thyroid cancer cells by regulating the expression of CDK6, which is expected to be a potential target for clinical application.
Collapse
Affiliation(s)
- Xinyan Li
- Department of Pharmacology, Cangzhou Medical College, Cangzhou, Hebei 061000, P.R. China
| | - Cuicui Liu
- Department of Pharmacology, Cangzhou Medical College, Cangzhou, Hebei 061000, P.R. China
| | - Xiumei Zhao
- Centre for Research and Development of Anti-tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Rui Wang
- Department of Critical Care Medicine, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Na Gu
- Centre for Research and Development of Anti-tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Hongsheng Shen
- Centre for Research and Development of Anti-tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Xijing Li
- Centre for Research and Development of Anti-tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Luyao Wang
- Centre for Research and Development of Anti-tumor Drugs, Tianjin Institute of Medical and Pharmaceutical Sciences, Tianjin 300020, P.R. China
| | - Chao Li
- Department of Pharmacology, Cangzhou Medical College, Cangzhou, Hebei 061000, P.R. China
| |
Collapse
|
57
|
Li M, Yang J, Liu K, Yang J, Zhan X, Wang L, Shen X, Chen J, Mao Z. p16 promotes proliferation in cervical carcinoma cells through CDK6-HuR-IL1A axis. J Cancer 2020; 11:1457-1467. [PMID: 32047552 PMCID: PMC6995400 DOI: 10.7150/jca.35479] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 12/02/2019] [Indexed: 11/05/2022] Open
Abstract
The Cyclin-Dependent Kinase Inhibitor p16 (p16) acts as a tumor suppressor in most cells, but for HPV transformed cervical cancer, in which oncoprotein E7 expressed by human papillomavirus (HPV) mediates the degradation of retinoblastoma protein (Rb), p16 exhibits oncogenic activity. Our study was conducted to study the mechanism underling p16 mediated promoting effect of cell proliferation in cervical cancer cell lines. CCK8 assay and EdU incorporation were conducted to evaluate cell proliferation. Loss-of-function assay was used to silence p16 in Ca Ski and SiHa cells. Next, western blot, qPCR, RNA silencing, luciferase activity assay, run-on assay, mRNA stability assay, RNA immunoprecipitation, co-immunoprecipitation Immunofluorescence were performed to examine the interaction between CDK6, HuR, and IL1A mRNA in p16 mediated proliferation promoting effect. Our results showed that: (1) Silencing p16 inhibited the proliferation of cervical cancer cells by decreasing the half-life of IL1A mRNA in CDK6 dependent manner; (2) The stabilization of IL1A mRNA was regulated by HuR which could be inactivated by p16/CDK6 mediated phosphorylation at Ser202; (3) IL1A mediated the oncogenic activity of p16 in cervical carcinoma cell lines. In conclusion, p16 promotes proliferation in cervical carcinoma cells through CDK6-HuR-IL1A axis.
Collapse
Affiliation(s)
- Mingzhe Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jiong Yang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China
| | - Kaiyu Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jianming Yang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiangwen Zhan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Le Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaomeng Shen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jing Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Zebin Mao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
58
|
Weiterer S, Meier‐Soelch J, Georgomanolis T, Mizi A, Beyerlein A, Weiser H, Brant L, Mayr‐Buro C, Jurida L, Beuerlein K, Müller H, Weber A, Tenekeci U, Dittrich‐Breiholz O, Bartkuhn M, Nist A, Stiewe T, van IJcken WFJ, Riedlinger T, Schmitz ML, Papantonis A, Kracht M. Distinct IL-1α-responsive enhancers promote acute and coordinated changes in chromatin topology in a hierarchical manner. EMBO J 2020; 39:e101533. [PMID: 31701553 PMCID: PMC6939198 DOI: 10.15252/embj.2019101533] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 12/14/2022] Open
Abstract
How cytokine-driven changes in chromatin topology are converted into gene regulatory circuits during inflammation still remains unclear. Here, we show that interleukin (IL)-1α induces acute and widespread changes in chromatin accessibility via the TAK1 kinase and NF-κB at regions that are highly enriched for inflammatory disease-relevant SNPs. Two enhancers in the extended chemokine locus on human chromosome 4 regulate the IL-1α-inducible IL8 and CXCL1-3 genes. Both enhancers engage in dynamic spatial interactions with gene promoters in an IL-1α/TAK1-inducible manner. Microdeletions of p65-binding sites in either of the two enhancers impair NF-κB recruitment, suppress activation and biallelic transcription of the IL8/CXCL2 genes, and reshuffle higher-order chromatin interactions as judged by i4C interactome profiles. Notably, these findings support a dominant role of the IL8 "master" enhancer in the regulation of sustained IL-1α signaling, as well as for IL-8 and IL-6 secretion. CRISPR-guided transactivation of the IL8 locus or cross-TAD regulation by TNFα-responsive enhancers in a different model locus supports the existence of complex enhancer hierarchies in response to cytokine stimulation that prime and orchestrate proinflammatory chromatin responses downstream of NF-κB.
Collapse
Affiliation(s)
- Sinah‐Sophia Weiterer
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Johanna Meier‐Soelch
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | | | - Athanasia Mizi
- Center for Molecular Medicine CologneUniversity of CologneCologneGermany
- Department of PathologyUniversity Medical Center GöttingenGöttingenGermany
| | - Anna Beyerlein
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Hendrik Weiser
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Lilija Brant
- Department of PathologyUniversity Medical Center GöttingenGöttingenGermany
| | - Christin Mayr‐Buro
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Liane Jurida
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Knut Beuerlein
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Helmut Müller
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Axel Weber
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Ulas Tenekeci
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
| | - Oliver Dittrich‐Breiholz
- Research Core Unit GenomicsInstitute of Physiological ChemistryMedical School HannoverHannoverGermany
| | - Marek Bartkuhn
- Institute for GeneticsJustus Liebig University GiessenGiessenGermany
| | - Andrea Nist
- Genomics Core Facility and Institute of Molecular OncologyPhilipps University MarburgMarburgGermany
| | - Thorsten Stiewe
- Genomics Core Facility and Institute of Molecular OncologyPhilipps University MarburgMarburgGermany
- Member of the German Center for Lung Research (DZL)GiessenGermany
| | | | - Tabea Riedlinger
- Institute of BiochemistryJustus Liebig University GiessenGiessenGermany
| | - M Lienhard Schmitz
- Member of the German Center for Lung Research (DZL)GiessenGermany
- Institute of BiochemistryJustus Liebig University GiessenGiessenGermany
| | - Argyris Papantonis
- Center for Molecular Medicine CologneUniversity of CologneCologneGermany
- Department of PathologyUniversity Medical Center GöttingenGöttingenGermany
| | - Michael Kracht
- Rudolf Buchheim Institute of PharmacologyJustus Liebig University GiessenGiessenGermany
- Member of the German Center for Lung Research (DZL)GiessenGermany
| |
Collapse
|
59
|
Shen Z, Feng X, Fang Y, Li Y, Li Z, Zhan Y, Lin M, Li G, Ding Y, Deng H. POTEE drives colorectal cancer development via regulating SPHK1/p65 signaling. Cell Death Dis 2019; 10:863. [PMID: 31723122 PMCID: PMC6853991 DOI: 10.1038/s41419-019-2046-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 09/08/2019] [Accepted: 10/03/2019] [Indexed: 12/24/2022]
Abstract
Aberrant gene expression plays critical roles in the development of colorectal cancer (CRC). Here we show that POTEE, which was identified as a member E of POTE ankyrin domain family, was significantly upregulated in colorectal tumors and predicted poor overall survival of CRC patients. In CRC cells, POTEE could act as an oncogene and could promote cell growth, cell-cycle progression, inhibit apoptosis, and elevates xenograft tumor growth. Mechanically, we used microarray analysis and identified a POTEE/SPHK1/p65 signaling axis, which affected the biological functions of CRC cells. Further evaluation showed that overexpression of POTEE could increase the protein expression of SPHK1, followed by promoting the phosphorylation and activation of p65 protein. Altogether, our findings suggested a POTEE/SPHK1/p65 signaling axis could promote colorectal tumorigenesis and POTEE might potentially serve as a novel biomarker for the diagnosis and an intervention of colorectal cancer.
Collapse
Affiliation(s)
- Zhiyong Shen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Xiaochuang Feng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Yuan Fang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Yongsheng Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Zhenkang Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Yizhi Zhan
- Department of Pathology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Mingdao Lin
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China
| | - Yi Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China.
| | - Haijun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Ave., 510515, Guangzhou, Guangdong Province, China.
| |
Collapse
|
60
|
Naciri I, Laisné M, Ferry L, Bourmaud M, Gupta N, Di Carlo S, Huna A, Martin N, Peduto L, Bernard D, Kirsh O, Defossez PA. Genetic screens reveal mechanisms for the transcriptional regulation of tissue-specific genes in normal cells and tumors. Nucleic Acids Res 2019; 47:3407-3421. [PMID: 30753595 PMCID: PMC6468300 DOI: 10.1093/nar/gkz080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 12/14/2022] Open
Abstract
The proper tissue-specific regulation of gene expression is essential for development and homeostasis in metazoans. However, the illegitimate expression of normally tissue-restricted genes—like testis- or placenta-specific genes—is frequently observed in tumors; this promotes transformation, but also allows immunotherapy. Two important questions are: how is the expression of these genes controlled in healthy cells? And how is this altered in cancer? To address these questions, we used an unbiased approach to test the ability of 350 distinct genetic or epigenetic perturbations to induce the illegitimate expression of over 40 tissue-restricted genes in primary human cells. We find that almost all of these genes are remarkably resistant to reactivation by a single alteration in signaling pathways or chromatin regulation. However, a few genes differ and are more readily activated; one is the placenta-expressed gene ADAM12, which promotes invasion. Using cellular systems, an animal model, and bioinformatics, we find that a non-canonical but druggable TGF-β/KAT2A/TAK1 axis controls ADAM12 induction in normal and cancer cells. More broadly, our data show that illegitimate gene expression in cancer is an heterogeneous phenomenon, with a few genes activatable by simple events, and most genes likely requiring a combination of events to become reactivated.
Collapse
Affiliation(s)
- Ikrame Naciri
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Marthe Laisné
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Laure Ferry
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Morgane Bourmaud
- INSERM U1132 and USPC Paris-Diderot, Hôpital Lariboisière, Paris, France
| | - Nikhil Gupta
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Selene Di Carlo
- Unité Stroma, Inflammation & Tissue Repair, Institut Pasteur, 75724 Paris, France; INSERM U1224, 75724 Paris, France
| | - Anda Huna
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| | - Nadine Martin
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| | - Lucie Peduto
- Unité Stroma, Inflammation & Tissue Repair, Institut Pasteur, 75724 Paris, France; INSERM U1224, 75724 Paris, France
| | - David Bernard
- Centre de Recherche en Cancérologie de Lyon, Inserm U1052, CNRS UMR 5286, Université de Lyon, Centre Léon Bérard, 69008 Lyon, France
| | - Olivier Kirsh
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| | - Pierre-Antoine Defossez
- Univ. Paris Diderot, Sorbonne Paris Cité, Epigenetics and Cell Fate, UMR 7216 CNRS, 75013 Paris, France
| |
Collapse
|
61
|
Hou YB, Ji K, Sun YT, Zhang LN, Chen JJ. CDK4/6 inhibitor palbociclib suppresses IgE-mediated mast cell activation. J Transl Med 2019; 17:276. [PMID: 31429774 PMCID: PMC6702723 DOI: 10.1186/s12967-019-2026-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Mast cell activation causes degranulation and release of cytokines, thereby promoting inflammation. The aim of this study was to investigate the inhibitory effect of CDK4/6 inhibition on mast cell activation in vitro and in vivo. METHODS RBL-2H3 rat basophilic leukemia cells (BLCs) and mouse bone marrow-derived mast cells (BMMCs) were sensitized with anti-dinitrophenol (DNP) immunoglobulin (Ig)E antibodies, stimulated with DNP-human serum albumin (HSA) antigens, and treated with the CDK4/6 inhibitor palbociclib. Histological stains were applied to reveal cytomorphological changes. Murine IgE-mediated passive cutaneous anaphylaxis (PCA) and ovalbumin (OVA)-induced active systemic anaphylaxis (ASA) models were used to examine palbociclib effects on allergic reactions in vivo. Western blots were performed to detect the expression of cell signaling molecules associated with mast cell activation. RESULTS Activated BLCs and BMMCs released copious granule-related mediators (histamine and β-hexosaminidase), which was reduced by palbociclib in a concentration-dependent manner. Palbociclib inhibited expression of the mast cell activation marker CD63 in activated BLCs and inhibited granule release (visualized with toluidine blue staining) while preventing morphological changes, (elongated shape maintained) and filamentous actin (F-actin) reorganization. Palbociclib suppressed molecular Lyn and/or mitogen-activated protein kinase (MAPK) signaling associated with mast cell activation in stimulated BLCs and attenuated allergic reactions in PCA mice dose dependently. Palbociclib attenuated body temperature reduction and diminished serum histamine levels in ovalbumin OVA-challenged ASA mice. CONCLUSION Palbociclib suppresses IgE-mediated mast cell activation in vitro and in vivo, suggesting that it may be developed into a therapy for mast cell-mediated allergic diseases via inhibition of mast cell degranulation.
Collapse
Affiliation(s)
- Yi-Bo Hou
- Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Kunmei Ji
- Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Yue-Tong Sun
- Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Li-Na Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, People's Republic of China
| | - Jia-Jie Chen
- Department of Biochemistry and Molecular Biology, School of Medicine, Shenzhen University, Shenzhen, 518060, People's Republic of China.
| |
Collapse
|
62
|
Single-Cell Analysis of Multiple Steps of Dynamic NF-κB Regulation in Interleukin-1α-Triggered Tumor Cells Using Proximity Ligation Assays. Cancers (Basel) 2019; 11:cancers11081199. [PMID: 31426445 PMCID: PMC6721548 DOI: 10.3390/cancers11081199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/02/2019] [Accepted: 08/09/2019] [Indexed: 12/21/2022] Open
Abstract
The frequently occurring heterogeneity of cancer cells and their functional interaction with immune cells in the tumor microenvironment raises the need to study signaling pathways at the single cell level with high precision, sensitivity, and spatial resolution. As aberrant NF-κB activity has been implicated in almost all steps of cancer development, we analyzed the dynamic regulation and activation status of the canonical NF-κB pathway in control and IL-1α-stimulated individual cells using proximity ligation assays (PLAs). These systematic experiments allowed the visualization of the dynamic dissociation and re-formation of endogenous p65/IκBα complexes and the nuclear translocation of NF-κB p50/p65 dimers. PLA combined with immunostaining for p65 or with NFKBIA single molecule mRNA-FISH facilitated the analysis of (i) further levels of the NF-κB pathway, (i) its functionality for downstream gene expression, and (iii) the heterogeneity of the NF-κB response in individual cells. PLA also revealed the interaction between NF-κB p65 and the P-body component DCP1a, a new p65 interactor that contributes to efficient p65 NF-κB nuclear translocation. In summary, these data show that PLA technology faithfully mirrored all aspects of dynamic NF-κB regulation, thus allowing molecular diagnostics of this key pathway at the single cell level which will be required for future precision medicine.
Collapse
|
63
|
Kong T, Xue Y, Cencic R, Zhu X, Monast A, Fu Z, Pilon V, Sangwan V, Guiot MC, Foulkes WD, Porco JA, Park M, Pelletier J, Huang S. eIF4A Inhibitors Suppress Cell-Cycle Feedback Response and Acquired Resistance to CDK4/6 Inhibition in Cancer. Mol Cancer Ther 2019; 18:2158-2170. [PMID: 31395685 DOI: 10.1158/1535-7163.mct-19-0162] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 06/04/2019] [Accepted: 07/29/2019] [Indexed: 11/16/2022]
Abstract
CDK4/6 inhibitors are FDA-approved drugs for estrogen receptor-positive (ER+) breast cancer and are being evaluated to treat other tumor types, including KRAS-mutant non-small cell lung cancer (NSCLC). However, their clinical utility is often limited by drug resistance. Here, we sought to better understand the resistant mechanisms and help devise potential strategies to overcome this challenge. We show that treatment with CDK4/6 inhibitors in both ER+ breast cancer and KRAS-mutant NSCLC cells induces feedback upregulation of cyclin D1, CDK4, and cyclin E1, mediating drug resistance. We demonstrate that rocaglates, which preferentially target translation of key cell-cycle regulators, effectively suppress this feedback upregulation induced by CDK4/6 inhibition. Consequently, combination treatment of CDK4/6 inhibitor palbociclib with the eukaryotic initiation factor (eIF) 4A inhibitor, CR-1-31-B, is synergistic in suppressing the growth of these cancer cells in vitro and in vivo Furthermore, ER+ breast cancer and KRAS-mutant NSCLC cells that acquired resistance to palbociclib after chronic drug exposure are also highly sensitive to this combination treatment strategy. Our findings reveal a novel strategy using eIF4A inhibitors to suppress cell-cycle feedback response and to overcome resistance to CDK4/6 inhibition in cancer.
Collapse
Affiliation(s)
- Tim Kong
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Yibo Xue
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Regina Cencic
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Xianbing Zhu
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Anie Monast
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Zheng Fu
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Virginie Pilon
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Veena Sangwan
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Marie-Christine Guiot
- Department of Pathology, Montreal Neurological Hospital/Institute, McGill University Health Centre, Montreal, Quebec, Canada
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada.,Department of Medical Genetics, and Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, Quebec, Canada.,Department of Medical Genetics and Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - John A Porco
- Department of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, Boston, Massachusetts
| | - Morag Park
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Jerry Pelletier
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada
| | - Sidong Huang
- Department of Biochemistry, The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
64
|
Kaneshiro K, Sakai Y, Suzuki K, Uchida K, Tateishi K, Terashima Y, Kawasaki Y, Shibanuma N, Yoshida K, Hashiramoto A. Interleukin-6 and tumour necrosis factor-α cooperatively promote cell cycle regulators and proliferate rheumatoid arthritis fibroblast-like synovial cells. Scand J Rheumatol 2019; 48:353-361. [DOI: 10.1080/03009742.2019.1602164] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- K Kaneshiro
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Y Sakai
- Division of Rehabilitation Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - K Suzuki
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - K Uchida
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - K Tateishi
- Department of Orthopedics, Kohnan Kakogawa Hospital, Kakogawa, Japan
| | - Y Terashima
- Department of Orthopedics, Kohnan Kakogawa Hospital, Kakogawa, Japan
| | - Y Kawasaki
- Department of Rheumatology, Kobe Kaisei Hospital, Kobe, Japan
| | - N Shibanuma
- Department of Orthopedic Surgery, Kobe Kaisei Hospital, Kobe, Japan
| | - K Yoshida
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - A Hashiramoto
- Department of Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
65
|
CSCs in Breast Cancer-One Size Does Not Fit All: Therapeutic Advances in Targeting Heterogeneous Epithelial and Mesenchymal CSCs. Cancers (Basel) 2019; 11:cancers11081128. [PMID: 31394796 PMCID: PMC6721464 DOI: 10.3390/cancers11081128] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/02/2019] [Accepted: 08/04/2019] [Indexed: 02/06/2023] Open
Abstract
Unlike other breast cancer subtypes, triple-negative breast cancer (TNBC) has no specific targets and is characterized as one of the most aggressive subtypes of breast cancer that disproportionately accounts for the majority of breast cancer-related deaths. Current conventional chemotherapeutics target the bulk tumor population, but not the cancer stem cells (CSCs) that are capable of initiating new tumors to cause disease relapse. Recent studies have identified distinct epithelial-like (E) ALDH+ CSCs, mesenchymal-like (M) CD44+/CD24- CSCs, and hybrid E/M ALDH+/CD44+/CD24- CSCs. These subtypes of CSCs exhibit differential signal pathway regulations, possess plasticity, and respond differently to treatment. As such, co-inhibition of different subtypes of CSCs is key to viable therapy. This review serves to highlight different pathway regulations in E and M CSCs in TNBC, and to further describe their role in disease progression. Potential inhibitors targeting E and/or M CSCs based on clinical trials are summarized for further investigation. Since future research needs to adopt suitable tumor models and take into account the divergence of E and M CSCs for the development of effective treatments, TNBC models for clinically translatable studies are further discussed.
Collapse
|
66
|
Dai Y, Jin F, Wu W, Kumar SK. Cell cycle regulation and hematologic malignancies. BLOOD SCIENCE 2019; 1:34-43. [PMID: 35402801 PMCID: PMC8975093 DOI: 10.1097/bs9.0000000000000009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 07/01/2019] [Indexed: 02/05/2023] Open
Abstract
A complex network precisely regulates the cell cycle through the G1, S, G2, and M phases and is the basis for cell division under physiological and pathological conditions. On the one hand, the transition from one phase to another as well as the progression within each phase is driven by the specific cyclin-dependent kinases (CDKs; e.g., CDK1, CDK2, CDK4, CDK6, and CDK7), together with their exclusive partner cyclins (e.g., cyclin A1, B1, D1-3, and E1). On the other hand, these phases are negatively regulated by endogenous CDK inhibitors such as p16ink4a, p18ink4c, p19ink4d, p21cip1, and p27kip1. In addition, several checkpoints control the commitment of cells to replicate DNA and undergo mitosis, thereby avoiding the passage of genomic errors to daughter cells. CDKs are often constitutively activated in cancer, which is characterized by the uncontrolled proliferation of transformed cells, due to genetic and epigenetic abnormalities in the genes involved in the cell cycle. Moreover, several oncogenes and defective tumor suppressors promote malignant changes by stimulating cell cycle entry and progression or disrupting DNA damage responses, including the cell cycle checkpoints, DNA repair mechanisms, and apoptosis. Thus, genes or proteins related to cell cycle regulation remain the main targets of interest in the treatment of various cancer types, including hematologic malignancies. In this context, advances in the understanding of the cell cycle regulatory machinery provide a basis for the development of novel therapeutic approaches. The present article summarizes the pathways as well as their genetic and epigenetic alterations that regulate the cell cycle; moreover, it discusses the various approved or potential therapeutic targets associated with the cell cycle, focusing on hematologic malignancies.
Collapse
Affiliation(s)
- Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Fengyan Jin
- Department of Hematology, Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Wei Wu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | | |
Collapse
|
67
|
Zhang H, Zhao B, Wang X, Zhang F, Yu W. LINC00511 knockdown enhances paclitaxel cytotoxicity in breast cancer via regulating miR-29c/CDK6 axis. Life Sci 2019; 228:135-144. [DOI: 10.1016/j.lfs.2019.04.063] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 04/12/2019] [Accepted: 04/28/2019] [Indexed: 01/23/2023]
|
68
|
Jiang B, Wang ES, Donovan KA, Liang Y, Fischer ES, Zhang T, Gray NS. Development of Dual and Selective Degraders of Cyclin-Dependent Kinases 4 and 6. Angew Chem Int Ed Engl 2019; 58:6321-6326. [PMID: 30802347 PMCID: PMC7678623 DOI: 10.1002/anie.201901336] [Citation(s) in RCA: 190] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Indexed: 12/24/2022]
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) are key regulators of the cell cycle, and there are FDA-approved CDK4/6 inhibitors for treating patients with metastatic breast cancer. However, due to conservation of their ATP-binding sites, development of selective agents has remained elusive. Here, we report imide-based degrader molecules capable of degrading both CDK4/6, or selectively degrading either CDK4 or CDK6. We were also able to tune the activity of these molecules against Ikaros (IKZF1) and Aiolos (IKZF3), which are well-established targets of imide-based degraders. We found that in mantle cell lymphoma cell lines, combined IKZF1/3 degradation with dual CDK4/6 degradation produced enhanced anti-proliferative effects compared to CDK4/6 inhibition, CDK4/6 degradation, or IKZF1/3 degradation. In summary, we report here the first compounds capable of inducing selective degradation of CDK4 and CDK6 as tools to pharmacologically dissect their distinct biological functions.
Collapse
Affiliation(s)
| | | | | | - Yanke Liang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA (USA)
| | - Eric S. Fischer
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA (USA)
| | | | | |
Collapse
|
69
|
Pan Y, Jiang Z, Sun D, Li Z, Pu Y, Wang D, Huang A, He C, Cao L. Cyclin-dependent Kinase 18 Promotes Oligodendrocyte Precursor Cell Differentiation through Activating the Extracellular Signal-Regulated Kinase Signaling Pathway. Neurosci Bull 2019; 35:802-814. [PMID: 31028571 DOI: 10.1007/s12264-019-00376-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 01/09/2019] [Indexed: 12/21/2022] Open
Abstract
The correct differentiation of oligodendrocyte precursor cells (OPCs) is essential for the myelination and remyelination processes in the central nervous system. Determining the regulatory mechanism is fundamental to the treatment of demyelinating diseases. By analyzing the RNA sequencing data of different neural cells, we found that cyclin-dependent kinase 18 (CDK18) is exclusively expressed in oligodendrocytes. In vivo studies showed that the expression level of CDK18 gradually increased along with myelin formation during development and in the remyelination phase in a lysophosphatidylcholine-induced demyelination model, and was distinctively highly expressed in oligodendrocytes. In vitro overexpression and interference experiments revealed that CDK18 directly promotes the differentiation of OPCs, without affecting their proliferation or apoptosis. Mechanistically, CDK18 activated the RAS/mitogen-activated protein kinase kinase 1/extracellular signal-regulated kinase pathway, thus promoting OPC differentiation. The results of the present study suggest that CDK18 is a promising cell-type specific target to treat demyelinating disease.
Collapse
Affiliation(s)
- Yuchen Pan
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
- Department of Internal Medicine, Jiangsu Provincial Corps Hospital, Chinese People's Armed Police Force, Yangzhou, 225003, China
| | - Zeping Jiang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Dingya Sun
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Zhenghao Li
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Yingyan Pu
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Dan Wang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Aijun Huang
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China
| | - Cheng He
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China.
| | - Li Cao
- Institute of Neuroscience, Key Laboratory of Molecular Neurobiology of the Ministry of Education and the Collaborative Innovation Center for Brain Science, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
70
|
Jiang B, Wang ES, Donovan KA, Liang Y, Fischer ES, Zhang T, Gray NS. Development of Dual and Selective Degraders of Cyclin‐Dependent Kinases 4 and 6. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201901336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Baishan Jiang
- Department of Cancer BiologyDana-Farber Cancer Institute Boston MA USA
| | - Eric S. Wang
- Department of Cancer BiologyDana-Farber Cancer Institute Boston MA USA
| | | | - Yanke Liang
- Department of Cancer BiologyDana-Farber Cancer Institute Boston MA USA
| | - Eric S. Fischer
- Department of Cancer BiologyDana-Farber Cancer Institute Boston MA USA
| | - Tinghu Zhang
- Department of Cancer BiologyDana-Farber Cancer Institute Boston MA USA
| | - Nathanael S. Gray
- Department of Cancer BiologyDana-Farber Cancer Institute Boston MA USA
| |
Collapse
|
71
|
Hu JZ, Rong ZJ, Li M, Li P, Jiang LY, Luo ZX, Duan CY, Cao Y, Lu HB. Silencing of lncRNA PKIA-AS1 Attenuates Spinal Nerve Ligation-Induced Neuropathic Pain Through Epigenetic Downregulation of CDK6 Expression. Front Cell Neurosci 2019; 13:50. [PMID: 30873006 PMCID: PMC6401634 DOI: 10.3389/fncel.2019.00050] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 02/01/2019] [Indexed: 01/07/2023] Open
Abstract
Neuropathic pain (NP) is among the most intractable comorbidities of spinal cord injury. Dysregulation of non-coding RNAs has also been implicated in the development of neuropathic pain. Here, we identified a novel lncRNA, PKIA-AS1, by using lncRNA array analysis in spinal cord tissue of spinal nerve ligation (SNL) model rats, and investigated the role of PKIA-AS1 in SNL-mediated neuropathic pain. We observed that PKIA-AS1 was significantly upregulated in SNL model rats and that PKIA-AS1 knockdown attenuated neuropathic pain progression. Alternatively, overexpression of PKIA-AS1 was sufficient to induce neuropathic pain-like symptoms in uninjured rats. We also found that PKIA-AS1 mediated SNL-induced neuropathic pain by directly regulating the expression and function of CDK6, which is essential for the initiation and maintenance of neuroinflammation and neuropathic pain. Therefore, our study identifies PKIA-AS1 as a novel therapeutic target for neuroinflammation related neuropathic pain.
Collapse
Affiliation(s)
- Jian-Zhong Hu
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Zi-Jie Rong
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Miao Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Ping Li
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.,Department of Obstetrics, Xiangya Hospital, Central South University, Changsha, China
| | - Li-Yuan Jiang
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Zi-Xiang Luo
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Chun-Yue Duan
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Yong Cao
- Department of Spine Surgery, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China
| | - Hong-Bin Lu
- Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Xiangya Hospital, Central South University, Changsha, China.,Department of Sports Medicine, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
72
|
Kollmann K, Briand C, Bellutti F, Schicher N, Blunder S, Zojer M, Hoeller C. The interplay of CDK4 and CDK6 in melanoma. Oncotarget 2019; 10:1346-1359. [PMID: 30858922 PMCID: PMC6402717 DOI: 10.18632/oncotarget.26515] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 11/25/2022] Open
Abstract
The cyclin-dependent kinases CDK4 and CDK6 promote progression through the cell cycle, where their functions are considered to be redundant. Recent studies have identified an additional role for CDK6 in the transcriptional regulation of cancer-relevant genes such as VEGF-A and EGR1 in hematopoietic malignancies. We show that the CDK4/6 inhibitor PD0332991 causes a significant decrease in tumor growth in a xenotransplantation mouse model of human melanoma. shRNA knockdown of either CDK4 or CDK6 significantly reduces cell proliferation and impedes their migratory capacity in vitro, which translates into a strong inhibition of tumor growth in xenotransplantation experiments. CDK4/6 inhibition results not only in the pronounced reduction of cell proliferation but also in an impaired tumor angiogenesis. CDK6 knockdown in melanoma cell lines impairs VEGF-A expression and reduces the potential stimulation of endothelial cell growth. The knockdown of CDK4 ends in similar results. The effect is caused by changes of CDK6 localization, less CDK6 is detected on the VEGF-A promoter. Bioinformatic analysis of human melanoma patient data verifies the key role of CDK6 in tumor angiogenesis in melanoma. The results highlight the importance of the delicate balance between CDK4 and CDK6 in regulating the cell cycle and transcription.
Collapse
Affiliation(s)
- Karoline Kollmann
- Institute of Pharmacology and Toxicology, Veterinary University of Vienna, Vienna, Austria
| | - Coralie Briand
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| | - Florian Bellutti
- Institute of Pharmacology and Toxicology, Veterinary University of Vienna, Vienna, Austria
| | - Nikolaus Schicher
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| | - Stefan Blunder
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, Veterinary University of Vienna, Vienna, Austria
| | - Christoph Hoeller
- Department of Dermatology, Division of General Dermatology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
73
|
CDK6 coordinates JAK2 V617F mutant MPN via NF-κB and apoptotic networks. Blood 2019; 133:1677-1690. [PMID: 30635286 DOI: 10.1182/blood-2018-08-872648] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/07/2019] [Indexed: 01/27/2023] Open
Abstract
Over 80% of patients with myeloproliferative neoplasms (MPNs) harbor the acquired somatic JAK2 V617F mutation. JAK inhibition is not curative and fails to induce a persistent response in most patients, illustrating the need for the development of novel therapeutic approaches. We describe a critical role for CDK6 in MPN evolution. The absence of Cdk6 ameliorates clinical symptoms and prolongs survival. The CDK6 protein interferes with 3 hallmarks of disease: besides regulating malignant stem cell quiescence, it promotes nuclear factor κB (NF-κB) signaling and contributes to cytokine production while inhibiting apoptosis. The effects are not mirrored by palbociclib, showing that the functions of CDK6 in MPN pathogenesis are largely kinase independent. Our findings thus provide a rationale for targeting CDK6 in MPN.
Collapse
|
74
|
Liu J, Duan Z, Guo W, Zeng L, Wu Y, Chen Y, Tai F, Wang Y, Lin Y, Zhang Q, He Y, Deng J, Stewart RL, Wang C, Lin PC, Ghaffari S, Evers BM, Liu S, Zhou MM, Zhou BP, Shi J. Targeting the BRD4/FOXO3a/CDK6 axis sensitizes AKT inhibition in luminal breast cancer. Nat Commun 2018; 9:5200. [PMID: 30518851 PMCID: PMC6281582 DOI: 10.1038/s41467-018-07258-y] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 10/22/2018] [Indexed: 01/06/2023] Open
Abstract
BRD4 assembles transcriptional machinery at gene super-enhancer regions and governs the expression of genes that are critical for cancer progression. However, it remains unclear whether BRD4-mediated gene transcription is required for tumor cells to develop drug resistance. Our data show that prolonged treatment of luminal breast cancer cells with AKT inhibitors induces FOXO3a dephosphorylation, nuclear translocation, and disrupts its association with SirT6, eventually leading to FOXO3a acetylation as well as BRD4 recognition. Acetylated FOXO3a recognizes the BD2 domain of BRD4, recruits the BRD4/RNAPII complex to the CDK6 gene promoter, and induces its transcription. Pharmacological inhibition of either BRD4/FOXO3a association or CDK6 significantly overcomes the resistance of luminal breast cancer cells to AKT inhibitors in vitro and in vivo. Our study reports the involvement of BRD4/FOXO3a/CDK6 axis in AKTi resistance and provides potential therapeutic strategies for treating resistant breast cancer.
Collapse
Affiliation(s)
- Jingyi Liu
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, 21702, USA
| | - Zhibing Duan
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Weijie Guo
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Lei Zeng
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Bethune Institute of Epigenetic Medicine, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Yadi Wu
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Yule Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Fang Tai
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Yifan Wang
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Yiwei Lin
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Qiang Zhang
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Bethune Institute of Epigenetic Medicine, The First Hospital, Jilin University, Changchun, Jilin, 130021, China
| | - Yanling He
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jiong Deng
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rachel L Stewart
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Chi Wang
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Pengnian Charles Lin
- Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD, 21702, USA
| | - Saghi Ghaffari
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - B Mark Evers
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
- Department of Surgery, University of Kentucky College of Medicine, Lexington, KY, 40506, USA
| | - Suling Liu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Ming-Ming Zhou
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Binhua P Zhou
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
| | - Jian Shi
- Department of Molecular and Cellular Biochemistry, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
- Markey Cancer Center, University of Kentucky College of Medicine, Lexington, KY, 40506, USA.
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
75
|
Vuokila N, Lukasiuk K, Bot AM, van Vliet EA, Aronica E, Pitkänen A, Puhakka N. miR-124-3p is a chronic regulator of gene expression after brain injury. Cell Mol Life Sci 2018; 75:4557-4581. [PMID: 30155647 PMCID: PMC11105702 DOI: 10.1007/s00018-018-2911-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/02/2018] [Accepted: 08/22/2018] [Indexed: 02/06/2023]
Abstract
Traumatic brain injury (TBI) initiates molecular and cellular pathologies that underlie post-injury morbidities, including hippocampus-related memory decline and epileptogenesis. Non-coding small RNAs are master regulators of gene expression with the potential to affect multiple molecular pathways. To evaluate whether hippocampal gene expression networks are chronically regulated by microRNAs after TBI, we sampled the dentate gyrus of rats with severe TBI induced by lateral fluid-percussion injury 3 months earlier. Ingenuity pathway analysis revealed 30 upregulated miR-124-3p targets, suggesting that miR-124-3p is downregulated post-TBI (z-score = - 5.146, p < 0.05). Droplet digital polymerase chain reaction (ddPCR) and in situ hybridization confirmed the chronic downregulation of miR-124-3p (p < 0.05). Quantitative PCR analysis of two targets, Plp2 and Stat3, indicated that their upregulation correlated with the miR-124-3p downregulation (r = - 0.647, p < 0.05; r = - 0.629, p < 0.05, respectively). Immunohistochemical staining of STAT3 confirmed the increased protein expression. STRING analysis showed that 9 of the 30 miR-124-3p targets belonged to a STAT3 network. Reactome analysis and data mining connected the targets especially to inflammation and signal transduction. L1000CDS2 software revealed drugs (e.g., importazole, trichostatin A, and IKK-16) that could reverse the observed molecular changes. The translational value of our data was emphasized by in situ hybridization showing chronic post-traumatic downregulation of miR-124-3p in the dentate gyrus of TBI patients. Analysis of another brain injury model, status epilepticus, highlighted the fact that chronic downregulation of miR-124 is a common phenomenon after brain injury. Together, our findings indicate that miR-124-3p is a chronic modulator of molecular networks relevant to post-injury hippocampal pathologies in experimental models and in humans.
Collapse
Affiliation(s)
- Niina Vuokila
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland
| | - Katarzyna Lukasiuk
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str, 02-093, Warsaw, Poland
| | - Anna Maria Bot
- The Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str, 02-093, Warsaw, Poland
| | - Erwin A van Vliet
- Department of (Neuro)pathology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of (Neuro)pathology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
- Stichting Epilepsie Instellingen Nederland (SEIN), Amsterdam, The Netherlands
| | - Asla Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland.
| | - Noora Puhakka
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
76
|
Abstract
The transcription factor NF-κB is a critical regulator of immune and inflammatory responses. In mammals, the NF-κB/Rel family comprises five members: p50, p52, p65 (Rel-A), c-Rel, and Rel-B proteins, which form homo- or heterodimers and remain as an inactive complex with the inhibitory molecules called IκB proteins in resting cells. Two distinct NF-κB signaling pathways have been described: 1) the canonical pathway primarily activated by pathogens and inflammatory mediators, and 2) the noncanonical pathway mostly activated by developmental cues. The most abundant form of NF-κB activated by pathologic stimuli via the canonical pathway is the p65:p50 heterodimer. Disproportionate increase in activated p65 and subsequent transactivation of effector molecules is integral to the pathogenesis of many chronic diseases such as the rheumatoid arthritis, inflammatory bowel disease, multiple sclerosis, and even neurodegenerative pathologies. Hence, the NF-κB p65 signaling pathway has been a pivotal point for intense drug discovery and development. This review begins with an overview of p65-mediated signaling followed by discussion of strategies that directly target NF-κB p65 in the context of chronic inflammation.
Collapse
Affiliation(s)
- Sivagami Giridharan
- Department of Oral Medicine, Madha Dental College, Kundrathur, Chennai, TN, India
| | - Mythily Srinivasan
- Department of Oral Pathology, Medicine and Radiology, Indiana University School of Dentistry, Indiana University Purdue University at Indianapolis, Indianapolis, IN, USA,
- Provaidya LLC, Indianapolis, IN, USA,
| |
Collapse
|
77
|
Cheng J, Wu LM, Deng XS, Wu J, Lv Z, Zhao HF, Yang Z, Ni Y. MicroRNA-449a suppresses hepatocellular carcinoma cell growth via G1 phase arrest and the HGF/MET c-Met pathway. Hepatobiliary Pancreat Dis Int 2018; 17:336-344. [PMID: 30108016 DOI: 10.1016/j.hbpd.2018.07.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 07/16/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Accumulating evidence demonstrates that microRNAs (miRNAs) play essential roles in tumorigenesis and cancer progression of hepatocellular carcinoma (HCC). Average targets of a miRNA were more than 100. And one miRNA may act in tumor via regulating several targets. The present study aimed to explore more potential targets of miR-449a by proteomics technology and further uncover the role of miR-449a in HCC tumorigenesis. METHODS Technologies such as iTRAQ-based quantitative proteomic were used to investigate the effect of miR-449a on HCC. The expression of c-Met and miR-449a was detected by qRT-PCR in HCC samples. Gain- and loss-of-function experiments were performed to identify the function and potential target of miR-449a in HCC cells. RESULTS In HCC, miR-449a was significantly downregulated, while c-Met was upregulated concurrently. Quantitative proteomics and luciferase reporter assay identified c-Met as a direct target of miR-449a. Moreover, miR-449a inhibited HCC growth not only by targeting CDK6 but also by suppressing c-Met/Ras/Raf/ERK signaling pathway. Furthermore, the inhibition of c-Met expression with a specific siRNA significantly inhibited cells growth and deregulated the ERK pathway in HCC. CONCLUSION The tumor suppressor miR-449a suppresses HCC tumorigenesis by repressing the c-Met/ERK pathway.
Collapse
Affiliation(s)
- Jun Cheng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Li-Ming Wu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou 310003, China
| | - Xue-Song Deng
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Jian Wu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou 310003, China
| | - Zhen Lv
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, Hangzhou 310003, China
| | - Hang-Fen Zhao
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Zhang Yang
- Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China; School of Life Science, Fudan University, Shanghai 200433, China
| | - Yong Ni
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China.
| |
Collapse
|
78
|
Bellutti F, Tigan AS, Nebenfuehr S, Dolezal M, Zojer M, Grausenburger R, Hartenberger S, Kollmann S, Doma E, Prchal-Murphy M, Uras IZ, Höllein A, Neuberg DS, Ebert BL, Ringler A, Mueller AC, Loizou JI, Hinds PW, Vogl C, Heller G, Kubicek S, Zuber J, Malumbres M, Farlik M, Villunger A, Kollmann K, Sexl V. CDK6 Antagonizes p53-Induced Responses during Tumorigenesis. Cancer Discov 2018; 8:884-897. [PMID: 29899063 DOI: 10.1158/2159-8290.cd-17-0912] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 04/05/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
Abstract
Tumor formation is a multistep process during which cells acquire genetic and epigenetic changes until they reach a fully transformed state. We show that CDK6 contributes to tumor formation by regulating transcriptional responses in a stage-specific manner. In early stages, the CDK6 kinase induces a complex transcriptional program to block p53 in hematopoietic cells. Cells lacking CDK6 kinase function are required to mutate TP53 (encoding p53) to achieve a fully transformed immortalized state. CDK6 binds to the promoters of genes including the p53 antagonists Prmt5, Ppm1d, and Mdm4 The findings are relevant to human patients: Tumors with low levels of CDK6 have mutations in TP53 significantly more often than expected.Significance: CDK6 acts at the interface of p53 and RB by driving cell-cycle progression and antagonizing stress responses. While sensitizing cells to p53-induced cell death, specific inhibition of CDK6 kinase activity may provoke the outgrowth of p53-mutant clones from premalignant cells. Cancer Discov; 8(7); 884-97. ©2018 AACR.This article is highlighted in the In This Issue feature, p. 781.
Collapse
Affiliation(s)
- Florian Bellutti
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Anca-Sarmiza Tigan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sofie Nebenfuehr
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Marlies Dolezal
- Platform Bioinformatics and Biostatistics, University of Veterinary Medicine, Vienna, Austria
| | - Markus Zojer
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Reinhard Grausenburger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Svenja Hartenberger
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sebastian Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Eszter Doma
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | | | - Donna S Neuberg
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Benjamin L Ebert
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.,Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Anna Ringler
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Andre C Mueller
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Joanna I Loizou
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philip W Hinds
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Tufts Cancer Center, Boston, Massachusetts
| | - Claus Vogl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine, Vienna, Austria
| | | | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | | | - Matthias Farlik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | | | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria.
| |
Collapse
|
79
|
Meier-Soelch J, Jurida L, Weber A, Newel D, Kim J, Braun T, Schmitz ML, Kracht M. RNAi-Based Identification of Gene-Specific Nuclear Cofactor Networks Regulating Interleukin-1 Target Genes. Front Immunol 2018; 9:775. [PMID: 29755455 PMCID: PMC5934416 DOI: 10.3389/fimmu.2018.00775] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/28/2018] [Indexed: 12/22/2022] Open
Abstract
The potent proinflammatory cytokine interleukin (IL)-1 triggers gene expression through the NF-κB signaling pathway. Here, we investigated the cofactor requirements of strongly regulated IL-1 target genes whose expression is impaired in p65 NF-κB-deficient murine embryonic fibroblasts. By two independent small-hairpin (sh)RNA screens, we examined 170 genes annotated to encode nuclear cofactors for their role in Cxcl2 mRNA expression and identified 22 factors that modulated basal or IL-1-inducible Cxcl2 levels. The functions of 16 of these factors were validated for Cxcl2 and further analyzed for their role in regulation of 10 additional IL-1 target genes by RT-qPCR. These data reveal that each inducible gene has its own (quantitative) requirement of cofactors to maintain basal levels and to respond to IL-1. Twelve factors (Epc1, H2afz, Kdm2b, Kdm6a, Mbd3, Mta2, Phf21a, Ruvbl1, Sin3b, Suv420h1, Taf1, and Ube3a) have not been previously implicated in inflammatory cytokine functions. Bioinformatics analysis indicates that they are components of complex nuclear protein networks that regulate chromatin functions and gene transcription. Collectively, these data suggest that downstream from the essential NF-κB signal each cytokine-inducible target gene has further subtle requirements for individual sets of nuclear cofactors that shape its transcriptional activation profile.
Collapse
Affiliation(s)
- Johanna Meier-Soelch
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Giessen, Germany
| | - Liane Jurida
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Giessen, Germany
| | - Axel Weber
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Giessen, Germany
| | - Doris Newel
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Giessen, Germany
| | - Johnny Kim
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Thomas Braun
- Max-Planck-Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Michael Kracht
- Rudolf Buchheim Institute of Pharmacology, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
80
|
Wu J, Wang Q, Dai W, Wang W, Yue M, Wang J. Massive GGAAs in genomic repetitive sequences serve as a nuclear reservoir of NF-κB. J Genet Genomics 2018; 45:193-203. [PMID: 29748061 DOI: 10.1016/j.jgg.2018.04.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/01/2022]
Abstract
Nuclear factor κB (NF-κB) is a DNA-binding transcription factor. Characterizing its genomic binding sites is crucial for understanding its gene regulatory function and mechanism in cells. This study characterized the binding sites of NF-κB RelA/p65 in the tumor neurosis factor-α (TNFα) stimulated HeLa cells by a precise chromatin immunoprecipitation-sequencing (ChIP-seq). The results revealed that NF-κB binds nontraditional motifs (nt-motifs) containing conserved GGAA quadruplet. Moreover, nt-motifs mainly distribute in the peaks nearby centromeres that contain a larger number of repetitive elements such as satellite, simple repeats and short interspersed nuclear elements (SINEs). This intracellular binding pattern was then confirmed by the in vitro detection, indicating that NF-κB dimers can bind the nontraditional κB (nt-κB) sites with low affinity. However, this binding hardly activates transcription. This study thus deduced that NF-κB binding nt-motifs may realize functions other than gene regulation as NF-κB binding traditional motifs (t-motifs). To testify the deduction, many ChIP-seq data of other cell lines were then analyzed. The results indicate that NF-κB binding nt-motifs is also widely present in other cells. The ChIP-seq data analysis also revealed that nt-motifs more widely distribute in the peaks with low-fold enrichment. Importantly, it was also found that NF-κB binding nt-motifs is mainly present in the resting cells, whereas NF-κB binding t-motifs is mainly present in the stimulated cells. Astonishingly, no known function was enriched by the gene annotation of nt-motif peaks. Based on these results, this study proposed that the nt-κB sites that extensively distribute in larger numbers of repeat elements function as a nuclear reservoir of NF-κB. The nuclear NF-κB proteins stored at nt-κB sites in the resting cells may be recruited to the t-κB sites for regulating its target genes upon stimulation.
Collapse
Affiliation(s)
- Jian Wu
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Qiao Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Dai
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Wei Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China
| | - Ming Yue
- Department of Infectious Diseases, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210002, China
| | - Jinke Wang
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing 210096, China.
| |
Collapse
|
81
|
Nakajima M, Kawaguchi M, Matsuyama M, Ota K, Fujita J, Matsukura S, Huang SK, Morishima Y, Ishii Y, Satoh H, Sakamoto T, Hizawa N. Transcription Elongation Factor P-TEFb Is Involved in IL-17F Signaling in Airway Smooth Muscle Cells. Int Arch Allergy Immunol 2018; 176:83-90. [PMID: 29649811 DOI: 10.1159/000488154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/06/2018] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND IL-17F is involved in the pathogenesis of several inflammatory diseases, including asthma and COPD. However, the effects of steroids on the function of IL-17F signaling mechanisms are largely unknown. One of the transcription elongation factors, positive transcription elongation factor b (P-TEFb) composed of cyclin T1 and cyclin-dependent kinase 9 (CDK9), is known as a novel checkpoint regulator of gene expression via bromodomain-containing protein 4 (Brd4). METHODS Human airway smooth muscle cells were stimulated with IL-17F and the expression of IL-8 was evaluated by real-time PCR and ELISA. Next, the phosphorylation of CDK9 was determined by Western blotting. The CDK9 inhibitor and short interfering RNAs (siRNAs) targeting Brd4, cyclin T1, and CDK9 were used to identify the effect on IL-17F-induced IL-8 expression. Finally, the effect of steroids and its signaling were evaluated. RESULTS IL-17F markedly induced the transcription of the IL-8 gene and the expression of the protein. Pretreatment of CDK9 inhibitor and transfection of siRNAs targeting CDK9 markedly abrogated IL-17F-induced IL-8 production. Transfection of siRNAs targeting Brd4 and cyclin T1 diminished IL-17F-induced phosphorylation of CDK9 and IL-8 production. Moreover, budesonide decreased CDK9 phosphorylation and markedly inhibited IL-17F-induced IL-8 production. CONCLUSIONS This is the first report that P-TEFb is involved in IL-17F-induced IL-8 expression and that steroids diminish it via the inhibition of CDK9 phosphorylation. IL-17F and P-TEFb might be novel therapeutic targets for airway inflammatory diseases.
Collapse
Affiliation(s)
- Masayuki Nakajima
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Mio Kawaguchi
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Masashi Matsuyama
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Kyoko Ota
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Junichi Fujita
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Matsukura
- Respiratory Disease Center, Showa University Northern Yokohama Hospital, Kanagawa, Japan
| | - Shau-Ku Huang
- Johns Hopkins University, Asthma and Allergy Center, Baltimore, Maryland, USA.,National Health Research Institutes, Taiwan, Taiwan
| | - Yuko Morishima
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Yukio Ishii
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Hiroaki Satoh
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tohru Sakamoto
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Nobuyuki Hizawa
- Department of Pulmonary Medicine, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
82
|
Sooreshjani MA, Gursoy UK, Aryal UK, Sintim HO. Proteomic analysis of RAW macrophages treated with cGAMP or c-di-GMP reveals differentially activated cellular pathways. RSC Adv 2018; 8:36840-36851. [PMID: 35558957 PMCID: PMC9089301 DOI: 10.1039/c8ra04603d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/23/2018] [Indexed: 12/28/2022] Open
Abstract
Global and quantitative analysis of the proteome help to reveal how host cells sense invading bacteria and respond to bacterial signaling molecules. Here, we performed label free quantitative proteomic analysis of RAW macrophages treated with host-derived cGAMP and bacterial-derived c-di-GMP, in an attempt to identify cellular pathways impacted by these dinucleotides and determine if the host responds differentially to these two cyclic dinucleotides. We identified a total of 3811 proteins of which abundances of 404 proteins in cGAMP and 236 proteins in c-di-GMP treated cells were significantly different compared to the control. Many of the proteins that were strongly and commonly upregulated, such as interferon-induced proteins 47, 202 and 204 (Ifi47, Ifi202, Ifi204), ubiquitin-activating enzyme E7 (Uba7), interferon-induced protein with tetratricopeptide repeats 1, 2 or 3 (Ifit1, Ifit2, Ifit3), ubiquitin-like protein ISG15 (ISG15), might be due to the fact that both dinucleotides promote the production of interferons, which induce the expression of many proteins. However, there were also other proteins that were differentially affected by cGAMP or c-di-GMP treatment, including probable ATP-dependent RNA helicase DHX58 (Dhx58), nuclear autoantigen Sp-100 (Sp100), MARCKS-related protein (Marcksl1) and antigen peptide transporter 2 (Tap2). This is probably due to the differential levels of IFNs produced by the dinucleotides or may indicate that non-STING activation might also contribute to the host's response to c-di-GMP and cGAMP. Interestingly Trex1, a nuclease that degrades DNA (an activator of cGAS to produce cGAMP), was upregulated (3.22 fold) upon cGAMP treatment, hinting at a possible feedback loop to regulate cGAMP synthesis. These results lay a foundation for future studies to better characterize and understand the complex c-di-GMP and cGAMP signaling network. cGAMP modulates proteins involved in antigen presentation and inflammation.![]()
Collapse
Affiliation(s)
| | - Ulvi K. Gursoy
- Department of Periodontology
- Institute of Dentistry
- University of Turku
- Turku
- Finland
| | - Uma K. Aryal
- Purdue Proteomics Facility
- Bindley Bioscience Center
- Purdue University
- West Lafayette
- USA
| | - Herman O. Sintim
- Department of Chemistry
- Purdue University
- West Lafayette
- USA
- Department of Periodontology
| |
Collapse
|
83
|
Abstract
In this issue of Cancer Cell, Gong et al. have analyzed the sensitivity of 560 cell lines to the selective CDK4/6 inhibitor abemaciclib and have defined cancers with specific genomic "D-cyclin activating features (DCAF)" as particularly vulnerable. These findings will facilitate patient selection as development of this drug class continues.
Collapse
Affiliation(s)
- Geoffrey I Shapiro
- Early Drug Development Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
84
|
Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, Chhabra S, Huang W, Liu H, Aref AR, Ivanova E, Paweletz CP, Bowden M, Zhou CW, Herter-Sprie GS, Sorrentino JA, Bisi JE, Lizotte PH, Merlino AA, Quinn MM, Bufe LE, Yang A, Zhang Y, Zhang H, Gao P, Chen T, Cavanaugh ME, Rode AJ, Haines E, Roberts PJ, Strum JC, Richards WG, Lorch JH, Parangi S, Gunda V, Boland GM, Bueno R, Palakurthi S, Freeman GJ, Ritz J, Haining WN, Sharpless NE, Arthanari H, Shapiro GI, Barbie DA, Gray NS, Wong KK. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov 2017; 8:216-233. [PMID: 29101163 DOI: 10.1158/2159-8290.cd-17-0915] [Citation(s) in RCA: 519] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/24/2017] [Accepted: 10/31/2017] [Indexed: 12/26/2022]
Abstract
Immune checkpoint blockade, exemplified by antibodies targeting the PD-1 receptor, can induce durable tumor regressions in some patients. To enhance the efficacy of existing immunotherapies, we screened for small molecules capable of increasing the activity of T cells suppressed by PD-1. Here, we show that short-term exposure to small-molecule inhibitors of cyclin-dependent kinases 4 and 6 (CDK4/6) significantly enhances T-cell activation, contributing to antitumor effects in vivo, due in part to the derepression of NFAT family proteins and their target genes, critical regulators of T-cell function. Although CDK4/6 inhibitors decrease T-cell proliferation, they increase tumor infiltration and activation of effector T cells. Moreover, CDK4/6 inhibition augments the response to PD-1 blockade in a novel ex vivo organotypic tumor spheroid culture system and in multiple in vivo murine syngeneic models, thereby providing a rationale for combining CDK4/6 inhibitors and immunotherapies.Significance: Our results define previously unrecognized immunomodulatory functions of CDK4/6 and suggest that combining CDK4/6 inhibitors with immune checkpoint blockade may increase treatment efficacy in patients. Furthermore, our study highlights the critical importance of identifying complementary strategies to improve the efficacy of immunotherapy for patients with cancer. Cancer Discov; 8(2); 216-33. ©2017 AACR.See related commentary by Balko and Sosman, p. 143See related article by Jenkins et al., p. 196This article is highlighted in the In This Issue feature, p. 127.
Collapse
Affiliation(s)
- Jiehui Deng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - Eric S Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Russell W Jenkins
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Division of Medical Oncology, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, Massachusetts
| | - Shuai Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ruben Dries
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kathleen Yates
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sandeep Chhabra
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Wei Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hongye Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amir R Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Elena Ivanova
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Cloud P Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Chensheng W Zhou
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Grit S Herter-Sprie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - John E Bisi
- G1 Therapeutics, Research Triangle Park, North Carolina
| | - Patrick H Lizotte
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ashley A Merlino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Max M Quinn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Lauren E Bufe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Yanxi Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Hua Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Peng Gao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ting Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Megan E Cavanaugh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amanda J Rode
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eric Haines
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Jay C Strum
- G1 Therapeutics, Research Triangle Park, North Carolina
| | - William G Richards
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Jochen H Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Viswanath Gunda
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raphael Bueno
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sangeetha Palakurthi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jerome Ritz
- Division of Hematologic Malignancies, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - W Nicholas Haining
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Norman E Sharpless
- The Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Haribabu Arthanari
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - David A Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts.
| | - Kwok-Kin Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. .,Division of Hematology & Medical Oncology, Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York.,Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
85
|
Amulic B, Knackstedt SL, Abu Abed U, Deigendesch N, Harbort CJ, Caffrey BE, Brinkmann V, Heppner FL, Hinds PW, Zychlinsky A. Cell-Cycle Proteins Control Production of Neutrophil Extracellular Traps. Dev Cell 2017; 43:449-462.e5. [DOI: 10.1016/j.devcel.2017.10.013] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 07/12/2017] [Accepted: 10/09/2017] [Indexed: 01/09/2023]
|
86
|
Yuan W, Wang D, Liu Y, Tian D, Wang Y, Zhang R, Yin L, Deng Z. miR‑494 inhibits cell proliferation and metastasis via targeting of CDK6 in osteosarcoma. Mol Med Rep 2017; 16:8627-8634. [PMID: 28990071 PMCID: PMC5779916 DOI: 10.3892/mmr.2017.7709] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 06/01/2017] [Indexed: 12/25/2022] Open
Abstract
Tumorigenesis is a multistep process involving various cell growth-associated factors. Accumulated evidence indicates that the disordered regulation of microRNAs (miRNAs) contributes to tumorigenesis. However, the detailed mechanism underlying the involvement of miRNAs in oncogenesis remains to be fully elucidated. In the present study, the repressed expression of microRNA (miR)-494 was identified in 18 patients with osteosarcoma (OS) and OS cell lines, compared with corresponding controls. To determine whether deregulated miR-494 exerts tumor-suppressive effects in the development of OS, the effects of miR-494 on cell proliferation and metastasis were evaluated. It was found that the restoration of miR-494 in MG-63 and U2OS cells led to inhibited cell proliferation and attenuated migratory propensity in vitro, determined through analysis using MTT, colony formation and Transwell assays. In addition, overexpression of miR-494 markedly suppressed the tumor volume and weight in vivo. In accordance, the ectopic expression of miR-494 induced cell cycle arrest at the G1/S phase in OS cells. Bioinformatics analysis and luciferase reporter assays were performed to investigate the potential regulatory role of miR-494, the results of which indicated that miR-494 directly targeted cyclin-dependent kinase 6 (CDK6). Of note, the data obtained through reverse transcription-quantitative polymerase chain reaction and western blot analyses suggested that the elevated expression of miR-494 resulted in reduced mRNA and protein expression levels of CDK6. Taken together, these findings indicated that the miR-494/CDK6 axis has a significant tumor-suppressive effect on OS, and maybe a diagnostic and therapeutic target for the treatment of OS.
Collapse
Affiliation(s)
- Wei Yuan
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Du Wang
- Department of Orthopedics, Wuhan Hospital No. 3 and Tongren Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yang Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Dongdong Tian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yang Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Ranxi Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Liangjun Yin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| | - Zhongliang Deng
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
87
|
Chirivella L, Kirstein M, Ferrón SR, Domingo-Muelas A, Durupt FC, Acosta-Umanzor C, Cano-Jaimez M, Pérez-Sánchez F, Barbacid M, Ortega S, Burks DJ, Fariñas I. Cyclin-Dependent Kinase 4 Regulates Adult Neural Stem Cell Proliferation and Differentiation in Response to Insulin. Stem Cells 2017; 35:2403-2416. [DOI: 10.1002/stem.2694] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/25/2017] [Accepted: 07/11/2017] [Indexed: 12/21/2022]
Affiliation(s)
- Laura Chirivella
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| | - Martina Kirstein
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| | - Sacri R. Ferrón
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| | - Ana Domingo-Muelas
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| | - Fabrice C. Durupt
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| | - Carlos Acosta-Umanzor
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Centro de Investigación Príncipe Felipe; Valencia Spain
| | - Marifé Cano-Jaimez
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Centro de Investigación Príncipe Felipe; Valencia Spain
| | - Francisco Pérez-Sánchez
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| | - Mariano Barbacid
- Centro Nacional de Investigaciones Oncológicas (CNIO); Madrid Spain
| | - Sagrario Ortega
- Centro Nacional de Investigaciones Oncológicas (CNIO); Madrid Spain
| | - Deborah J. Burks
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), Centro de Investigación Príncipe Felipe; Valencia Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED); Spain
- Departamento de Biología Celular; Biología Funcional y Antropología Física and Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Universidad de Valencia; Burjassot Spain
| |
Collapse
|
88
|
CBX3 promotes colon cancer cell proliferation by CDK6 kinase-independent function during cell cycle. Oncotarget 2017; 8:19934-19946. [PMID: 28193906 PMCID: PMC5386735 DOI: 10.18632/oncotarget.15253] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 12/12/2016] [Indexed: 12/29/2022] Open
Abstract
Heterochromatin protein 1γ (CBX3) links histone methylation marks to transcriptional silence, DNA repair and RNA splicing, but a role for CBX3 in cancer remains largely unknown. In this study, we show that CBX3 in colon cancer cells promotes the progression of the cell cycle and proliferation in vitro and in vivo. Cell cycle (G1 phase to S phase) related gene CDK6 and p21 were further identified as targets of CBX3. In addition, we found that enhancing CDK6 suppresses cell proliferation by upregulating inhibitor p21 in the absence of CBX3, and this function is independent of the kinase activity of CDK6. Our results demonstrate a key role of CBX3 in colon carcinogenesis via suppressing the expression of CDK6/p21, which may disrupt the role of CDK6 in transcriptionally regulating p21, as part of a negative feedback loop to limit CDK6 excessive activation.
Collapse
|
89
|
Tenekeci U, Poppe M, Beuerlein K, Buro C, Müller H, Weiser H, Kettner-Buhrow D, Porada K, Newel D, Xu M, Chen ZJ, Busch J, Schmitz ML, Kracht M. K63-Ubiquitylation and TRAF6 Pathways Regulate Mammalian P-Body Formation and mRNA Decapping. Mol Cell 2017; 62:943-957. [PMID: 27315556 DOI: 10.1016/j.molcel.2016.05.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 01/22/2016] [Accepted: 05/13/2016] [Indexed: 01/09/2023]
Abstract
Signals and posttranslational modifications regulating the decapping step in mRNA degradation pathways are poorly defined. In this study we reveal the importance of K63-linked ubiquitylation for the assembly of decapping factors, P-body formation, and constitutive decay of instable mRNAs encoding mediators of inflammation by various experimental approaches. K63-branched ubiquitin chains also regulate IL-1-inducible phosphorylation of the P-body component DCP1a. The E3 ligase TRAF6 binds to DCP1a and indirectly regulates DCP1a phosphorylation, expression of decapping factors, and gene-specific mRNA decay. Mutation of six C-terminal lysines of DCP1a suppresses decapping activity and impairs the interaction with the mRNA decay factors DCP2, EDC4, and XRN1, but not EDC3, thus remodeling P-body architecture. The usage of ubiquitin chains for the proper assembly and function of the decay-competent mammalian decapping complex suggests an additional layer of control to allow a coordinated function of decapping activities and mRNA metabolism in higher eukaryotes.
Collapse
Affiliation(s)
- Ulas Tenekeci
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Michael Poppe
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Knut Beuerlein
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Christin Buro
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Helmut Müller
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Hendrik Weiser
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Daniela Kettner-Buhrow
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Katharina Porada
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Doris Newel
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Ming Xu
- Department of Molecular Biology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Zhijian J Chen
- Department of Molecular Biology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Julia Busch
- Institute of Biochemistry, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - M Lienhard Schmitz
- Institute of Biochemistry, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Michael Kracht
- Rudolf-Buchheim-Institute of Pharmacology, Justus-Liebig-University Giessen, 35392 Giessen, Germany.
| |
Collapse
|
90
|
Genes directly regulated by NF-κB in human hepatocellular carcinoma HepG2. Int J Biochem Cell Biol 2017; 89:157-170. [PMID: 28579529 DOI: 10.1016/j.biocel.2017.05.031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/25/2017] [Accepted: 05/29/2017] [Indexed: 12/14/2022]
Abstract
It has been well-known that over activation of NF-κB has close relationship with hepatitis and hepatocellular carcinoma (HCC). However, the complete and exact underlying molecular pathways and mechanisms still remain not fully understood. By manipulating NF-κB activity with its recognized activator TNFα and using ChIP-seq and RNA-seq techniques, this study identified 699 NF-κB direct target genes (DTGs) in a widely used HCC cell line, HepG2, including 399 activated and 300 repressed genes. In these NF-κB DTGs, 216 genes (126 activated and 90 repressed genes) are among the current HCC gene signature. In comparison with NF-κB target genes identified in LPS-induced THP-1 and TNFα-induced HeLa cells, only limited numbers (24-46) of genes were shared by the two cell lines, indicating the HCC specificity of identified genes. Functional annotation revealed that NF-κB DTGs in HepG2 cell are mainly related with many typical NF-κB-related biological processes including immune system process, response to stress, response to stimulus, defense response, and cell death, and signaling pathways of MAPK, TNF, TGF-beta, Chemokine, NF-kappa B, and Toll-like receptor. Some NF-κB DTGs are also involved in Hepatitis C and B pathways. It was found that 82 NF-κB DTGs code secretory proteins, which include CCL2 and DKK1 that have already been used as HCC markers. Finally, the NF-κB DTGs were further confirmed by detecting the NF-κB binding and expression of 14 genes with ChIP-PCR and RT-PCR. This study thus provides a useful NF-κB DTG list for future studies of NF-κB-related molecular mechanisms and theranostic biomarkers of HCC.
Collapse
|
91
|
Zhang J, Zhou L, Zhao S, Dicker DT, El-Deiry WS. The CDK4/6 inhibitor palbociclib synergizes with irinotecan to promote colorectal cancer cell death under hypoxia. Cell Cycle 2017; 16:1193-1200. [PMID: 28486050 DOI: 10.1080/15384101.2017.1320005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Hypoxia is an inherent impediment to cancer therapy. Palbociclib, a highly selective inhibitor for CDK4/6, has been tested in numerous clinical trials and has been approved by the FDA. We previously reported that CDK inhibitors can destabilize HIF1α regardless of the presence of hypoxia and can sensitize tumor cells to TRAIL through dual blockade of CDK1 and GSK-3β. To translate this knowledge into a cancer therapeutic strategy, we investigated the therapeutic effects and molecular mechanisms of CDK inhibition against colon cancer cells under normoxia and hypoxia. We found that palbociclib sensitizes colon cancer cells to hypoxia-induced apoptotic resistance via deregulation of HIF-1α accumulation. In addition to inhibition of cell proliferation, we observed that palbociclib promotes colon cancer cell death regardless of the presence of hypoxia at a comparatively high concentration via regulating ERK/GSK-3β signaling and GSK-3β expression. Furthermore, palbociclib synergized with irinotecan in a variety of colon cancer cell lines with various molecular subtypes via deregulating irinotecan-induced Rb phosphorylation and reducing HIF-1α accumulation under normoxia or hypoxia. Collectively, our findings provide a novel combination therapy strategy against hypoxic colon cancer cells that may be further translated in the clinic.
Collapse
Affiliation(s)
- Jun Zhang
- a Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program , Fox Chase Cancer Center , Philadelphia , PA , USA
| | - Lanlan Zhou
- a Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program , Fox Chase Cancer Center , Philadelphia , PA , USA
| | - Shuai Zhao
- a Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program , Fox Chase Cancer Center , Philadelphia , PA , USA
| | - David T Dicker
- a Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program , Fox Chase Cancer Center , Philadelphia , PA , USA
| | - Wafik S El-Deiry
- a Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Department of Medical Oncology and Molecular Therapeutics Program , Fox Chase Cancer Center , Philadelphia , PA , USA
| |
Collapse
|
92
|
Lin J, Kato M, Nagata K, Okuwaki M. Efficient DNA binding of NF-κB requires the chaperone-like function of NPM1. Nucleic Acids Res 2017; 45:3707-3723. [PMID: 28003476 PMCID: PMC5397172 DOI: 10.1093/nar/gkw1285] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/21/2016] [Accepted: 12/13/2016] [Indexed: 12/11/2022] Open
Abstract
NPM1/nucleophosmin is frequently overexpressed in various tumors, although the oncogenic role of NPM1 remains unclear. Here we revealed the link between NPM1 and nuclear factor-κB (NF-κB), a master regulator of inflammation. We found that NPM1 knockdown decreased NF-κB-mediated transcription of selected target genes by decreasing the recruitment of NF-κB p65 to the gene promoters. NPM1 is directly associated with the DNA binding domain of p65 to enhance its DNA binding activity without being a part of the DNA-NF-κB complex. This result suggests that NF-κB requires the chaperone-like function of NPM1 for DNA binding. Furthermore, we demonstrated that NPM1 was required for efficient inflammatory gene expression induced by tumor necrosis factor alpha (TNF-α) and lipopolysaccharide in fibroblasts and macrophages. The NF-κB-mediated invasion of breast cancer cells was significantly decreased by NPM1 knockdown. Our study suggests a novel mechanistic insight into the NF-κB-mediated transcription and an oncogenic role of NPM1 in both tumor cells and the tumor micro-environment through the regulation of NF-κB.
Collapse
Affiliation(s)
- Jianhuang Lin
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| | - Mitsuyasu Kato
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
- Department of Experimental Pathology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| | - Kyosuke Nagata
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| | - Mitsuru Okuwaki
- PhD Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
- Department of Infection Biology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba 305-8575 Japan
| |
Collapse
|
93
|
The NF-κB-dependent and -independent transcriptome and chromatin landscapes of human coronavirus 229E-infected cells. PLoS Pathog 2017; 13:e1006286. [PMID: 28355270 PMCID: PMC5386326 DOI: 10.1371/journal.ppat.1006286] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/10/2017] [Accepted: 03/09/2017] [Indexed: 12/20/2022] Open
Abstract
Coronavirus replication takes place in the host cell cytoplasm and triggers inflammatory gene expression by poorly characterized mechanisms. To obtain more insight into the signals and molecular events that coordinate global host responses in the nucleus of coronavirus-infected cells, first, transcriptome dynamics was studied in human coronavirus 229E (HCoV-229E)-infected A549 and HuH7 cells, respectively, revealing a core signature of upregulated genes in these cells. Compared to treatment with the prototypical inflammatory cytokine interleukin(IL)-1, HCoV-229E replication was found to attenuate the inducible activity of the transcription factor (TF) NF-κB and to restrict the nuclear concentration of NF-κB subunits by (i) an unusual mechanism involving partial degradation of IKKβ, NEMO and IκBα and (ii) upregulation of TNFAIP3 (A20), although constitutive IKK activity and basal TNFAIP3 expression levels were shown to be required for efficient virus replication. Second, we characterized actively transcribed genomic regions and enhancers in HCoV-229E-infected cells and systematically correlated the genome-wide gene expression changes with the recruitment of Ser5-phosphorylated RNA polymerase II and prototypical histone modifications (H3K9ac, H3K36ac, H4K5ac, H3K27ac, H3K4me1). The data revealed that, in HCoV-infected (but not IL-1-treated) cells, an extensive set of genes was activated without inducible p65 NF-κB being recruited. Furthermore, both HCoV-229E replication and IL-1 were shown to upregulate a small set of genes encoding immunomodulatory factors that bind p65 at promoters and require IKKβ activity and p65 for expression. Also, HCoV-229E and IL-1 activated a common set of 440 p65-bound enhancers that differed from another 992 HCoV-229E-specific enhancer regions by distinct TF-binding motif combinations. Taken together, the study shows that cytoplasmic RNA viruses fine-tune NF-κB signaling at multiple levels and profoundly reprogram the host cellular chromatin landscape, thereby orchestrating the timely coordinated expression of genes involved in multiple signaling, immunoregulatory and metabolic processes. Coronaviruses are major human and animal pathogens. They belong to a family of plus-strand RNA viruses that have extremely large genomes and encode a variety of proteins involved in virus-host interactions. The four common coronaviruses (HCoV-229E, NL63, OC43, HKU1) cause mainly upper respiratory tract infections, while zoonotic coronaviruses (SARS-CoV and MERS-CoV) cause severe lung disease, including acute respiratory distress syndrome (ARDS). The molecular basis for this fundamentally different pathology is incompletely understood. Our study provides a genome-wide investigation of epigenetic changes occurring in response to HCoV-229E. We identify at high resolution a large number of regulatory regions in the genome of infected cells that coordinate de novo gene transcription. Many of these genes have immunomodulatory functions and, most likely, contribute to limiting viral replication, while other factors may promote viral replication. The study provides an intriguing example of a virus that completes its entire life cycle in the cytoplasm while sending multiple signals to the nuclear chromatin compartment to adjust the host cell repertoire of transcribed genes. The approach taken in this study is expected to provide a suitable framework for future studies aimed at dissecting and comparing host responses to representative coronaviruses with different pathogenic potential in humans.
Collapse
|
94
|
Di Giovanni C, Novellino E, Chilin A, Lavecchia A, Marzaro G. Investigational drugs targeting cyclin-dependent kinases for the treatment of cancer: an update on recent findings (2013-2016). Expert Opin Investig Drugs 2017; 25:1215-30. [PMID: 27606939 DOI: 10.1080/13543784.2016.1234603] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Cell cycle and gene transcription are under the control of cyclin-dependent kinases (CDKs), whose activity depends on the binding with cyclins. Deregulated CDK activities have been reported in a majority of human cancers, representing potential therapeutic targets. AREAS COVERED This review provides preclinical and clinical (phase I/II) updates of promising therapeutic compounds targeting CDKs published between 2013 and 2016 EXPERT OPINION: First generation pan-CDK inhibitors showed marked toxicity in clinical trials and most compounds were discontinued. Despite their failure was ascribed also to inadequate patient selection rules, novel pan-CDK inhibitors have entered clinical trials with still poorly defined selection strategies. The most interesting results have been obtained with dual CDK4/6 inhibitors and through a more accurate evaluation of predictive biomarkers, suggesting the usefulness of CDK inhibitors for personalized treatment. The increased knowledge on the roles of CDKs in cell cycle and gene transcription suggests to review also the anticancer potential of first generation CDK inhibitors by defining more appropriate rules for patients engagement. Recent findings has highlighted CDK8 as a novel target for cancer treatment. Indeed some biomarkers for CDK8 inhibition sensitivity have already been proposed. CDK8 inhibition is also supposed to prevent cancer metastasis.
Collapse
Affiliation(s)
- Carmen Di Giovanni
- a Department of Pharmacy , University of Naples Federico II , Naples , Italy
| | - Ettore Novellino
- a Department of Pharmacy , University of Naples Federico II , Naples , Italy
| | - Adriana Chilin
- b Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Padova , Italy
| | - Antonio Lavecchia
- a Department of Pharmacy , University of Naples Federico II , Naples , Italy
| | - Giovanni Marzaro
- b Department of Pharmaceutical and Pharmacological Sciences , University of Padova , Padova , Italy
| |
Collapse
|
95
|
Uras IZ, Scheicher RM, Kollmann K, Glösmann M, Prchal-Murphy M, Tigan AS, Fux DA, Altamura S, Neves J, Muckenthaler MU, Bennett KL, Kubicek S, Hinds PW, von Lindern M, Sexl V. Cdk6 contributes to cytoskeletal stability in erythroid cells. Haematologica 2017; 102:995-1005. [PMID: 28255017 PMCID: PMC5451331 DOI: 10.3324/haematol.2016.159947] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 02/22/2017] [Indexed: 12/03/2022] Open
Abstract
Mice lacking Cdk6 kinase activity suffer from mild anemia accompanied by elevated numbers of Ter119+ cells in the bone marrow. The animals show hardly any alterations in erythroid development, indicating that Cdk6 is not required for proliferation and maturation of erythroid cells. There is also no difference in stress erythropoiesis following hemolysis in vivo. However, Cdk6−/− erythrocytes have a shortened lifespan and are more sensitive to mechanical stress in vitro, suggesting differences in cytoskeletal architecture. Erythroblasts contain both Cdk4 and Cdk6, while mature erythrocytes apparently lack Cdk4 and their Cdk6 is partly associated with the cytoskeleton. We used mass spectrometry to show that Cdk6 interacts with a number of proteins involved in cytoskeleton organization. Cdk6−/− erythroblasts show impaired F-actin formation and lower levels of gelsolin, which interacts with Cdk6. We also found that Cdk6 regulates the transcription of a panel of genes involved in actin (de-)polymerization. Cdk6-deficient cells are sensitive to drugs that interfere with the cytoskeleton, suggesting that our findings are relevant to the treatment of patients with anemia – and may be relevant to cancer patients treated with the new generation of CDK6 inhibitors.
Collapse
Affiliation(s)
- Iris Z Uras
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Ruth M Scheicher
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Karoline Kollmann
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | | | - Michaela Prchal-Murphy
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Anca S Tigan
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Daniela A Fux
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| | - Sandro Altamura
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Germany
| | - Joana Neves
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Germany
| | - Martina U Muckenthaler
- Department of Pediatric Hematology, Oncology, and Immunology, University of Heidelberg, Germany.,Molecular Medicine Partnership Unit, University of Heidelberg, Germany
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Stefan Kubicek
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Philip W Hinds
- Department of Developmental, Molecular and Chemical Biology, Tufts University School of Medicine, and Tufts Cancer Center, Boston, MA, USA
| | | | - Veronika Sexl
- Institute of Pharmacology and Toxicology, University of Veterinary Medicine, Vienna, Austria
| |
Collapse
|
96
|
Rocca A, Schirone A, Maltoni R, Bravaccini S, Cecconetto L, Farolfi A, Bronte G, Andreis D. Progress with palbociclib in breast cancer: latest evidence and clinical considerations. Ther Adv Med Oncol 2017; 9:83-105. [PMID: 28203301 PMCID: PMC5298405 DOI: 10.1177/1758834016677961] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Deregulation of the cell cycle is a hallmark of cancer, and research on cell cycle control has allowed identification of potential targets for anticancer treatment. Palbociclib is a selective inhibitor of the cyclin-dependent kinases 4 and 6 (CDK4/6), which are involved, with their coregulatory partners cyclin D, in the G1-S transition. Inhibition of this step halts cell cycle progression in cells in which the involved pathway, including the retinoblastoma protein (Rb) and the E2F family of transcription factors, is functioning, although having been deregulated. Among breast cancers, those with functioning cyclin D-CDK4/6-Rb-E2F are mainly hormone-receptor (HR) positive, with some HER2-positive and rare triple-negative cases. Deregulation results from genetic or otherwise occurring hyperactivation of molecules subtending cell cycle progression, or inactivation of cell cycle inhibitors. Based on results of randomized clinical trials, palbociclib was granted accelerated approval by the US Food and Drug Administration (FDA) for use in combination with letrozole as initial endocrine-based therapy for metastatic disease in postmenopausal women with HR-positive, HER2-negative breast cancer, and was approved for use in combination with fulvestrant in women with HR-positive, HER2-negative advanced breast cancer with disease progression following endocrine therapy. This review provides an update of the available knowledge on the cell cycle and its regulation, on the alterations in cyclin D-CDK4/6-Rb-E2F axis in breast cancer and their roles in endocrine resistance, on the preclinical activity of CDK4/6 inhibitors in breast cancer, both as monotherapy and as partners of combinatorial synergic treatments, and on the clinical development of palbociclib in breast cancer.
Collapse
Affiliation(s)
- Andrea Rocca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Via Maroncelli 40, Meldola, FC 47014, Italy
| | - Alessio Schirone
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Roberta Maltoni
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Sara Bravaccini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Lorenzo Cecconetto
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Alberto Farolfi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Giuseppe Bronte
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| | - Daniele Andreis
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, FC, Italy
| |
Collapse
|
97
|
Zhang M, Wang Y, Qian F, Li P, Xu X. Hypericin inhibits oligomeric amyloid β42-induced inflammation response in microglia and ameliorates cognitive deficits in an amyloid β injection mouse model of Alzheimer's disease by suppressing MKL1. Biochem Biophys Res Commun 2016; 481:71-76. [DOI: 10.1016/j.bbrc.2016.11.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2016] [Accepted: 11/05/2016] [Indexed: 10/20/2022]
|
98
|
de Andrés MC, Takahashi A, Oreffo ROC. Demethylation of an NF-κB enhancer element orchestrates iNOS induction in osteoarthritis and is associated with altered chondrocyte cell cycle. Osteoarthritis Cartilage 2016; 24:1951-1960. [PMID: 27307355 DOI: 10.1016/j.joca.2016.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/18/2016] [Accepted: 06/06/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the methylation profile of the nuclear factor (NF)-κB enhancer region at -5.8 kb of inducible nitric oxide synthase (iNOS) and the subsequent role in the induction of osteoarthritis (OA) via cell cycle regulation. METHODS Percentage methylation was determined by pyrosequencing, gene expression by qRT-PCR and cell proliferation was determined using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Transient transfections were induced to determine the effect of the NF-κB enhancer region on cell proliferation and the influence of DNA methylation. RESULTS In vitro de-methylation with 5-aza-dC showed decreased levels of DNA methylation at CpG sites localised at -5.8 kb, which correlated with higher levels of iNOS expression. In vitro methylation of the NF-κB enhancer region at -5.8 kb increased the percentage of cells at G0/G1 cell cycle phase. Loss of methylation within this region correlated with, enhanced proliferation and increased number of cells at G2/M phase. OA chondrocytes demonstrated up-regulation of the G0/G1 cell cycle progression markers Cyclin D1 and CDK6 in contrast to control cells. We demonstrate the loss of methylation that occurs at specific CpG sites localised at the -5.8 kb NF-κB enhancer region of the iNOS gene in OA chondrocytes permits the binding of this transcription factor activating the expression of iNOS. This results in subsequent altered cell cycle regulation, altered proliferative phenotype and transmission of the pathogenic phenotype to daughter cells. CONCLUSIONS This study indicates that inhibition of cell cycle progression by iNOS enhancer hypermethylation is capable of reducing pro-inflammatory responses via down-regulation of NF-κB with important therapeutic implications in OA.
Collapse
Affiliation(s)
- M C de Andrés
- Bone and Joint Research Group, Centre for Human Development Stem Cells and Regeneration, Institute of Developmental Science, University of Southampton Medical School, Southampton, UK
| | - A Takahashi
- Bone and Joint Research Group, Centre for Human Development Stem Cells and Regeneration, Institute of Developmental Science, University of Southampton Medical School, Southampton, UK; Department of Orthopaedic Surgery, Tohoku University Hospital, Sendai, Japan
| | - R O C Oreffo
- Bone and Joint Research Group, Centre for Human Development Stem Cells and Regeneration, Institute of Developmental Science, University of Southampton Medical School, Southampton, UK.
| |
Collapse
|
99
|
Tadesse S, Yu M, Kumarasiri M, Le BT, Wang S. Targeting CDK6 in cancer: State of the art and new insights. Cell Cycle 2016; 14:3220-30. [PMID: 26315616 DOI: 10.1080/15384101.2015.1084445] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cyclin-dependent kinase 6 (CDK6) plays a vital role in regulating the progression of the cell cycle. More recently, CDK6 has also been shown to have a transcriptional role in tumor angiogenesis. Up-regulated CDK6 activity is associated with the development of several types of cancers. While CDK6 is over-expressed in cancer cells, it has a low detectable level in non-cancerous cells and CDK6-null mice develop normally, suggesting a specific oncogenic role of CDK6, and that its inhibition may represent an ideal mechanism-based and low toxic therapeutic strategy in cancer treatment. Identification of selective small molecule inhibitors of CDK6 is thus needed for drug development. Herein, we review the latest understandings of the biological regulation and oncogenic roles of CDK6. The potential clinical relevance of CDK6 inhibition, the progress in the development of small-molecule CDK6 inhibitors and the rational design of potential selective CDK6 inhibitors are also discussed.
Collapse
Affiliation(s)
- Solomon Tadesse
- a Center for Drug Discovery and Development, Sansom Institute for Health Research, Center for Cancer Biology; and School of Pharmacy and Medical Sciences, University of South Australia ; Adelaide , Australia
| | - Mingfeng Yu
- a Center for Drug Discovery and Development, Sansom Institute for Health Research, Center for Cancer Biology; and School of Pharmacy and Medical Sciences, University of South Australia ; Adelaide , Australia
| | - Malika Kumarasiri
- a Center for Drug Discovery and Development, Sansom Institute for Health Research, Center for Cancer Biology; and School of Pharmacy and Medical Sciences, University of South Australia ; Adelaide , Australia
| | - Bich Thuy Le
- a Center for Drug Discovery and Development, Sansom Institute for Health Research, Center for Cancer Biology; and School of Pharmacy and Medical Sciences, University of South Australia ; Adelaide , Australia
| | - Shudong Wang
- a Center for Drug Discovery and Development, Sansom Institute for Health Research, Center for Cancer Biology; and School of Pharmacy and Medical Sciences, University of South Australia ; Adelaide , Australia
| |
Collapse
|
100
|
The Influenza A Virus Genotype Determines the Antiviral Function of NF-κB. J Virol 2016; 90:7980-90. [PMID: 27356900 DOI: 10.1128/jvi.00946-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 06/05/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED The role of NF-κB in influenza A virus (IAV) infection does not reveal a coherent picture, as pro- and also antiviral functions of this transcription factor have been described. To address this issue, we used clustered regularly interspaced short palindromic repeat with Cas9 (CRISPR-Cas9)-mediated genome engineering to generate murine MLE-15 cells lacking two essential components of the NF-κB pathway. Cells devoid of either the central NF-κB essential modulator (NEMO) scaffold protein and thus defective in IκB kinase (IKK) activation or cells not expressing the NF-κB DNA-binding and transactivation subunit p65 were tested for propagation of the SC35 virus, which has an avian host range, and its mouse-adapted variant, SC35M. While NF-κB was not relevant for replication of SC35M, the absence of NF-κB activity increased replication of the nonadapted SC35 virus. This antiviral effect of NF-κB was most prominent upon infection of cells with low virus titers as they usually occur during the initiation phase of IAV infection. The defect in NF-κB signaling resulted in diminished IAV-triggered phosphorylation of interferon regulatory factor 3 (IRF3) and expression of the antiviral beta interferon (IFN-β) gene. To identify the viral proteins responsible for NF-κB dependency, reassortant viruses were generated by reverse genetics. SC35 viruses containing the SC35M segment encoding neuraminidase (NA) were completely inert to the inhibitory effect of NF-κB, emphasizing the importance of the viral genotype for susceptibility to the antiviral functions of NF-κB. IMPORTANCE This study addresses two different issues. First, we investigated the role of the host cell transcription factor NF-κB in IAV replication by genetic manipulation of IAVs by reverse genetics combined with targeted genome engineering of host cells using CRISPR-Cas9. The analysis of these two highly defined genetic systems indicated that the IAV genotype can influence whether NF-κB displays an antiviral function and thus might in part explain incoherent results from the literature. Second, we found that perturbation of NF-κB function greatly improved the growth of a nonadapted IAV, suggesting that NF-κB may contribute to the maintenance of the host species barrier.
Collapse
|