1
|
Phuagkhaopong S, Janpattanapichai J, Sirirak N, Khemawoot P, Vivithanaporn P, Suknuntha K. Transcriptome analysis reveals a role of FOXO3 in antileukemia/lymphoma properties of panduratin A. Sci Rep 2024; 14:24795. [PMID: 39433897 PMCID: PMC11494127 DOI: 10.1038/s41598-024-75630-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
Boesenbergia rotunda, commonly known as fingerroot, is a medicinal and culinary plant native to the Indochina Peninsula. We found that panduratin A (Pan-A), one of the compounds present in the rhizome extract of fingerroot, inhibited cell proliferation, induced apoptosis, and promoted cell cycle arrest at G0/G1 phase in multiple hematologic malignant cell lines including leukemia and lymphoma lines. Pan-A inhibited these activities in leukemia and lymphoma cells in a concentration-dependent manner. High-throughput transcriptome analysis indicated that Pan-A is involved in the cell cycle, cellular senescence, apoptosis, and multiple canonical signaling pathways in lymphoma cells. The Forkhead box O (FOXO) transcription factor family was identified as a potential target of Pan-A. Western blot showed elevated caspase 7 and cPARP/PARP in the B-cell lymphoma cells after treatment with Pan-A. The inhibitory effects were accompanied by stimulation of Akt signaling and phosphorylation of FOXO3. Immunohistochemistry of tissues from patients with B-cell lymphoma revealed detectable levels of FOXO3 protein specifically in neoplastic B cells. Overall, our results highlight FOXO3 as a player underlying antileukemia/lymphoma effects of Pan-A.
Collapse
Affiliation(s)
- Suttinee Phuagkhaopong
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jiranan Janpattanapichai
- Interdisciplinary Program in Pharmacology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Noppavut Sirirak
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Phisit Khemawoot
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Pornpun Vivithanaporn
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Kran Suknuntha
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
2
|
Ghasemi N, Azizi H. Exploring Myc puzzle: Insights into cancer, stem cell biology, and PPI networks. Gene 2024; 916:148447. [PMID: 38583818 DOI: 10.1016/j.gene.2024.148447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/13/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
"The grand orchestrator," "Universal Amplifier," "double-edged sword," and "Undruggable" are just some of the Myc oncogene so-called names. It has been around 40 years since the discovery of the Myc, and it remains in the mainstream of cancer treatment drugs. Myc is part of basic helix-loop-helix leucine zipper (bHLH-LZ) superfamily proteins, and its dysregulation can be seen in many malignant human tumors. It dysregulates critical pathways in cells that are connected to each other, such as proliferation, growth, cell cycle, and cell adhesion, impacts miRNAs action, intercellular metabolism, DNA replication, differentiation, microenvironment regulation, angiogenesis, and metastasis. Myc, surprisingly, is used in stem cell research too. Its family includes three members, MYC, MYCN, and MYCL, and each dysfunction was observed in different cancer types. This review aims to introduce Myc and its function in the body. Besides, Myc deregulatory mechanisms in cancer cells, their intricate aspects will be discussed. We will look at promising drugs and Myc-based therapies. Finally, Myc and its role in stemness, Myc pathways based on PPI network analysis, and future insights will be explained.
Collapse
Affiliation(s)
- Nima Ghasemi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran.
| |
Collapse
|
3
|
Maharati A, Moghbeli M. Forkhead box proteins as the critical regulators of cisplatin response in tumor cells. Eur J Pharmacol 2023; 956:175937. [PMID: 37541368 DOI: 10.1016/j.ejphar.2023.175937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/11/2023] [Accepted: 07/31/2023] [Indexed: 08/06/2023]
Abstract
Cisplatin (CDDP) is one of the most common chemotherapy drugs used in a wide range of cancer patients; however, there is a high rate of CDDP resistance among cancer patients. Considering the side effects of cisplatin in normal tissues, it is necessary to predict the CDDP response in cancer patients. Therefore, identifying the molecular mechanisms involved in CDDP resistance can help to introduce the prognostic markers. Several molecular mechanisms such as apoptosis inhibition, drug efflux, drug detoxification, and increased DNA repair are involved in CDDP resistance. Regarding the key role of transcription factors in regulation of many cellular processes related to drug resistance, in the present review, we discussed the role of Forkhead box (FOX) protein family in CDDP response. It has been reported that FOX proteins mainly promote CDDP resistance through the regulation of DNA repair, autophagy, epithelial-mesenchymal transition (EMT), and signaling pathways. Therefore, FOX proteins can be introduced as the prognostic markers to predict CDDP response in cancer patients. In addition, considering that oncogenic role of FOX proteins, the CDDP treatment along with FOX inhibition can be used as a therapeutic strategy in cancer patients.
Collapse
Affiliation(s)
- Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
La T, Chen S, Zhao XH, Zhou S, Xu R, Teng L, Zhang YY, Ye K, Xu L, Guo T, Jamaluddin MF, Feng YC, Tang HJ, Wang Y, Xu Q, Gu Y, Cao H, Liu T, Thorne RF, Shao F, Zhang XD, Jin L. LncRNA LIMp27 Regulates the DNA Damage Response through p27 in p53-Defective Cancer Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204599. [PMID: 36638271 PMCID: PMC9982580 DOI: 10.1002/advs.202204599] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/06/2022] [Indexed: 06/17/2023]
Abstract
P53 inactivation occurs in about 50% of human cancers, where p53-driven p21 activity is devoid and p27 becomes essential for the establishment of the G1/S checkpoint upon DNA damage. Here, this work shows that the E2F1-responsive lncRNA LIMp27 selectively represses p27 expression and contributes to proliferation, tumorigenicity, and treatment resistance in p53-defective colon adenocarcinoma (COAD) cells. LIMp27 competes with p27 mRNA for binding to cytoplasmically localized hnRNA0, which otherwise stabilizes p27 mRNA leading to cell cycle arrest at the G0/G1 phase. In response to DNA damage, LIMp27 is upregulated in both wild-type and p53-mutant COAD cells, whereas cytoplasmic hnRNPA0 is only increased in p53-mutant COAD cells due to translocation from the nucleus. Moreover, high LIMp27 expression is associated with poor survival of p53-mutant but not wild-type p53 COAD patients. These results uncover an lncRNA mechanism that promotes p53-defective cancer pathogenesis and suggest that LIMp27 may constitute a target for the treatment of such cancers.
Collapse
Affiliation(s)
- Ting La
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
- National‐Local Joint Engineering Research Center of Biodiagnosis & BiotherapyThe Second Affiliated HospitalXi'an Jiaotong UniversityXi'anShaanxi710004China
| | - Song Chen
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Institute of Medicinal BiotechnologyJiangsu College of NursingHuai'anJiangsu223300China
| | - Xiao Hong Zhao
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Shuai Zhou
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Ran Xu
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Liu Teng
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Yuan Yuan Zhang
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Kaihong Ye
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
| | - Liang Xu
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Tao Guo
- Institute of Future AgricultureNorthwest A&F UniversityYanglingShaanxi712100China
| | - Muhammad Fairuz Jamaluddin
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Yu Chen Feng
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Medicine and Public HealthThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Hai Jie Tang
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Yanliang Wang
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Qin Xu
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Yue Gu
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Huixia Cao
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Tao Liu
- Children's Cancer Institute Australia for Medical ResearchUniversity of New South WalesSydneyNew South Wales2750Australia
| | - Rick F. Thorne
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Feng‐Min Shao
- Department of NephrologyHenan Provincial Key Laboratory of Kidney Disease and ImmunologyHenan Provincial Clinical Research Center for Kidney DiseaseHenan Provincial People's HospitalZhengzhouHenan450053China
| | - Xu Dong Zhang
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South Wales2308Australia
| | - Lei Jin
- Translational Research InstituteHenan Provincial and Zhengzhou City Key laboratory of Non‐coding RNA and Cancer MetabolismHenan International Join Laboratory of Non‐coding RNA and Metabolism in CancerHenan Provincial People's HospitalAcademy of Medical SciencesZhengzhou UniversityZhengzhouHenan450053China
- Noncoding Cancer Biomarkers and Therapeutics GroupCancer Detection & Therapy Research ProgramHunter Medical Research InstituteCallaghanNew South Wales2305Australia
- School of Medicine and Public HealthThe University of NewcastleCallaghanNew South Wales2308Australia
| |
Collapse
|
5
|
Carrillo P, Bernal M, Téllez-Quijorna C, Marrero AD, Vidal I, Castilla L, Caro C, Domínguez A, García-Martín ML, Quesada AR, Medina MA, Martínez-Poveda B. The synthetic molecule stauprimide impairs cell growth and migration in triple-negative breast cancer. Biomed Pharmacother 2023; 158:114070. [PMID: 36526536 DOI: 10.1016/j.biopha.2022.114070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
Stauprimide, a semi-synthetic derivative of staurosporine, is known mainly for its potent differentiation-enhancing properties in embryonic stem cells. Here, we studied the effects of stauprimide in cell growth and migration of triple-negative breast cancer cells in vitro, evaluating its potential antitumoral activity in an orthotopic mouse model of breast cancer in vivo. Our results from survival curves, EdU incorporation, cell cycle analysis and annexin-V detection in MDA-MB-231 cells indicated that stauprimide inhibited cell proliferation, arresting cell cycle in G2/M without induction of apoptosis. A decrease in the migratory capability of MDA-MB-231 was also assessed in response to stauprimide. In this work we pointed to a mechanism of action of stauprimide involving the modulation of ERK1/2, Akt and p38 MAPK signalling pathways, and the downregulation of MYC in MDA-MB-231 cells. In addition, orthotopic MDA-MB-231 xenograft and 4T1 syngeneic models suggested an effect of stauprimide in vivo, increasing the necrotic core of tumors and reducing metastasis in lung and liver of mice. Together, our results point to the promising role of stauprimide as a putative therapeutic agent in triple-negative breast cancer.
Collapse
Affiliation(s)
- P Carrillo
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - M Bernal
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - C Téllez-Quijorna
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain
| | - A D Marrero
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - I Vidal
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - L Castilla
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - C Caro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - A Domínguez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain
| | - M L García-Martín
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain; Biomedical Research Networking Center in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Spain
| | - A R Quesada
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain; CIBER de Enfermedades Raras (CIBERER, Instituto de Salud Carlos III), Spain
| | - M A Medina
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain; CIBER de Enfermedades Raras (CIBERER, Instituto de Salud Carlos III), Spain
| | - B Martínez-Poveda
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, Andalucía Tech, E-29071 Málaga, Spain; Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), C/Severo Ochoa, 35, 29590, Málaga, Spain; CIBER de Enfermedades Cardiovasculares (CIBERCV, Instituto de Salud Carlos III, Madrid), Spain.
| |
Collapse
|
6
|
Scalia P, Williams SJ, Fujita-Yamaguchi Y, Giordano A. Cell cycle control by the insulin-like growth factor signal: at the crossroad between cell growth and mitotic regulation. Cell Cycle 2023; 22:1-37. [PMID: 36005738 PMCID: PMC9769454 DOI: 10.1080/15384101.2022.2108117] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In proliferating cells and tissues a number of checkpoints (G1/S and G2/M) preceding cell division (M-phase) require the signal provided by growth factors present in serum. IGFs (I and II) have been demonstrated to constitute key intrinsic components of the peptidic active fraction of mammalian serum. In vivo genetic ablation studies have shown that the cellular signal triggered by the IGFs through their cellular receptors represents a non-replaceable requirement for cell growth and cell cycle progression. Retroactive and current evaluation of published literature sheds light on the intracellular circuitry activated by these factors providing us with a better picture of the pleiotropic mechanistic actions by which IGFs regulate both cell size and mitogenesis under developmental growth as well as in malignant proliferation. The present work aims to summarize the cumulative knowledge learned from the IGF ligands/receptors and their intracellular signaling transducers towards control of cell size and cell-cycle with particular focus to their actionable circuits in human cancer. Furthermore, we bring novel perspectives on key functional discriminants of the IGF growth-mitogenic pathway allowing re-evaluation on some of its signal components based upon established evidences.
Collapse
Affiliation(s)
- Pierluigi Scalia
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,CST, Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United states,CONTACT Pierluigi Scalia ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA9102, USA
| | - Stephen J Williams
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,CST, Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United states
| | - Yoko Fujita-Yamaguchi
- Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Antonio Giordano
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,School of Medical Biotechnology, University of Siena, Italy
| |
Collapse
|
7
|
Zhang C, Zhao J, Zhao J, Liu B, Tang W, Liu Y, Huang W, Weinman SA, Li Z. CYP2E1-dependent upregulation of SIRT7 is response to alcohol mediated metastasis in hepatocellular carcinoma. Cancer Gene Ther 2022; 29:1961-1974. [PMID: 35902730 PMCID: PMC10832389 DOI: 10.1038/s41417-022-00512-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 02/02/2023]
Abstract
Long-term alcohol use is a confirmed risk factor of liver cancer tumorigenesis and metastasis. Multiple mechanisms responsible for alcohol related tumorigenesis have been proposed, including toxic reactive metabolite production, oxidative stress and fat accumulation. However, mechanisms underlying alcohol-mediated liver cancer metastasis remain largely unknown. We have previously demonstrated that SIRT7 regulates chemosensitivity by altering a p53-dependent pathway in human HCC. In the current study, we further revealed that SIRT7 is a critical factor in promoting liver cancer metastasis. SIRT7 expression is associated with disease stage and high SIRT7 predicts worse overall and disease-free survival. Overexpression of SIRT7 promotes HCC cell migration and EMT while knockdown of SIRT7 showed opposite effects. Mechanistically, we found that SIRT7 suppresses E-Cadherin expression through FOXO3-dependent promoter binding and H3K18 deacetylation. Knockdown of FOXO3 abolished the suppressive effect of SIRT7 on E-cadherin transcription. More importantly, we identified that alcohol treatment upregulates SIRT7 and suppresses E-cadherin expression via a CYP2E/ROS axis in hepatocytes both in vitro and in vivo. Antioxidant treatment in primary hepatocyte or CYP2E1-/- mice fed with alcohol impaired those effects. Reducing SIRT7 activity completely abolished alcohol-mediated promotion of liver cancer metastasis in vivo. Taken together, our data reveal that SIRT7 is a pivotal regulator of alcohol-mediated HCC metastasis.
Collapse
Affiliation(s)
- Chen Zhang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and Hunan Normal University School of Medicine, Changsha, Hunan, China
- Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Jinqiu Zhao
- Department of Infectious Disease, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Zhao
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Bohao Liu
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and Hunan Normal University School of Medicine, Changsha, Hunan, China
- Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Wenbin Tang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and Hunan Normal University School of Medicine, Changsha, Hunan, China
- Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, Hunan, China
| | - Yi Liu
- Department of General Surgery, People's Hospital of Hunan Province and Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Wenxiang Huang
- Department of Infectious Disease, First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Steven A Weinman
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- Liver Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - Zhuan Li
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Hunan Normal University School of Medicine, Changsha, Hunan, China.
- The Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, and Hunan Normal University School of Medicine, Changsha, Hunan, China.
- Department of Pharmacy, Hunan Normal University School of Medicine, Changsha, Hunan, China.
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
8
|
Stimulation of ROS Generation by Extract of Warburgia ugandensis Leading to G 0/G 1 Cell Cycle Arrest and Antiproliferation in A549 Cells. Antioxidants (Basel) 2021; 10:antiox10101559. [PMID: 34679694 PMCID: PMC8533466 DOI: 10.3390/antiox10101559] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/24/2022] Open
Abstract
Warburgia ugandensis Sprague (WU) is a traditional medicinal plant used for the treatment of various diseases, including cancer, in Africa. This study aimed to evaluate the anti-non-small cell lung cancer (NSCLC) activities of WU against A549 cells and to reveal potential molecular mechanisms. The cytotoxicity of various WU extracts was evaluated with HeLa (cervical cancer), HepG2 (liver cancer), HT-29 (colorectal cancer), and A549 (non-small cell lung cancer) cells by means of Sulforhodamine B (SRB) assay. Therein, the dimethyl carbonate extract of WU (WUD) was tested with the most potent anti-proliferative activity against the four cancer cell lines, and its effects on cell viability, cell cycle progression, DNA damage, intracellular reactive oxygen species (ROS), and expression levels of G0/G1-related proteins in A549 cells were further examined. First, it was found that WUD inhibited the proliferation of A549 cells in a time- and dose-dependent manner. In addition, WUD induced G0/G1 phase arrest and modulated the expression of G0/G1 phase-associated proteins Cyclin D1, Cyclin E1, and P27 in A549 cells. Furthermore, WUD increased the protein abundance of P27 by inhibiting FOXO3A/SKP2 axis-mediated protein degradation and also significantly induced the γH2AX expression and intracellular ROS generation of A549 cells. It was also found that the inhibitory effect of WUD on the proliferation and G0/G1 cell cycle progression of A549 cells could be attenuated by NAC, a ROS scavenger. On the other hand, phytochemical analysis of WUD with UPLC-QTOF-MS/MS indicated 10 sesquiterpenoid compounds. In conclusion, WUD exhibited remarkable anti-proliferative effects on A549 cells by improving the intracellular ROS level and by subsequently modulating the cell proliferation and G0/G1 cell cycle progression of A549 cells. These findings proved the good therapeutic potential of WU for the treatment of NSCLC.
Collapse
|
9
|
Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: a multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. J Hematol Oncol 2021; 14:121. [PMID: 34372899 PMCID: PMC8351444 DOI: 10.1186/s13045-021-01111-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022] Open
Abstract
MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and hematopoiesis. Due to the multi-functionality of MYC, its expression is regulated at multiple levels. Deregulation of this oncogene can give rise to a variety of cancers. In this review, MYC regulation and the mechanisms by which MYC adjusts cellular functions and its implication in hematologic malignancies are summarized. Further, we also discuss potential inhibitors of MYC that could be beneficial for treating hematologic malignancies.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Xu S, Lian Z, Zhang S, Xu Y, Zhang H. CircGNG4 Promotes the Progression of Prostate Cancer by Sponging miR-223 to Enhance EYA3/c-myc Expression. Front Cell Dev Biol 2021; 9:684125. [PMID: 34395419 PMCID: PMC8356047 DOI: 10.3389/fcell.2021.684125] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 06/30/2021] [Indexed: 12/30/2022] Open
Abstract
Patients diagnosed with prostate cancer often have a poor prognosis and limited treatment options, as the specific pathogenesis remains to be elucidated. Circular RNA (circRNA) is a type of non-coding RNA that interacts with microRNA (miRNA/miR) and transcription factors to regulate gene expression. However, little is known about specific circRNAs that serve roles in the pathogenesis of prostate cancer. Findings of the present study confirmed that circRNA G protein subunit γ 4 (circGNG4) was upregulated in prostate cancer tissues and cell lines. Knockdown of circGNG4 inhibited the malignant behavior of prostate cancer cells. Furthermore, bioinformatics were used to predict targeting interactions between circGNG4 or miR-223 and EYA transcriptional coactivator and phosphatase 3 (EYA3)/c-Myc mRNA. miR-223 inhibited the malignant behavior of prostate cancer cells, while EYA3/c-Myc had the opposite effect. circGNG4 enhanced the expression of EYA3/c-Myc by sponging miR-223 to promote the growth of prostate cancer tumors in vivo. In conclusion, the circGNG4/miR-223/EYA3/c-Myc regulatory pathway promoted the malignant progression of prostate cancer. The results of the present study may provide potential new targets for the diagnosis or treatment of prostate cancer.
Collapse
Affiliation(s)
- Shengxian Xu
- Department of Urology, Tianjin Key Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Zhenpeng Lian
- Department of Urology, Tianjin Key Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Siyang Zhang
- Department of Urology, Tianjin Key Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Yong Xu
- Department of Urology, Tianjin Key Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| | - Hongtuan Zhang
- Department of Urology, Tianjin Key Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin, China
| |
Collapse
|
11
|
Hafler D, Sumida T, Dulberg S, Schupp J, Stillwell H, Axisa PP, Comi M, Lincoln M, Unterman A, Kaminski N, Madi A, Kuchroo V. Type I Interferon Transcriptional Network Regulates Expression of Coinhibitory Receptors in Human T cells. RESEARCH SQUARE 2021:rs.3.rs-133494. [PMID: 34127967 PMCID: PMC8202434 DOI: 10.21203/rs.3.rs-133494/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
While inhibition of T cell co-inhibitory receptors has revolutionized cancer therapy, the mechanisms governing their expression on human T cells have not been elucidated. Type 1 interferon (IFN-I) modulates T cell immunity in viral infection, autoimmunity, and cancer, and may facilitate induction of T cell exhaustion in chronic viral infection. Here we show that IFN-I regulates co-inhibitory receptor expression on human T cells, inducing PD-1/TIM-3/LAG-3 while surprisingly inhibiting TIGIT expression. High-temporal-resolution mRNA profiling of IFN-I responses enabled the construction of dynamic transcriptional regulatory networks uncovering three temporal transcriptional waves. Perturbation of key transcription factors on human primary T cells revealed unique regulators that control expression of co-inhibitory receptors. We found that the dynamic IFN-I response in vitro closely mirrored T cell features with IFN-I linked acute SARS-CoV-2 infection in human, with high LAG3 and decreased TIGIT expression. Finally, our gene regulatory network identified SP140 as a key regulator for differential LAG3 and TIGIT expression, which were validated at the level of protein expression. The construction of IFN-I regulatory networks with identification of unique transcription factors controlling co-inhibitory receptor expression may provide targets for enhancement of immunotherapy in cancer, infectious diseases, and autoimmunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michela Comi
- Department of Immunobiology, Yale University School of Medicine; Department of Neurology, Yale University School of Medicine
| | | | - Avraham Unterman
- Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine
| | | | | | | |
Collapse
|
12
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
13
|
Akbari Dilmaghani N, Safaroghli-Azar A, Pourbagheri-Sigaroodi A, Bashash D. The PI3K/Akt/mTORC signaling axis in head and neck squamous cell carcinoma: Possibilities for therapeutic interventions either as single agents or in combination with conventional therapies. IUBMB Life 2021; 73:618-642. [PMID: 33476088 DOI: 10.1002/iub.2446] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/24/2020] [Indexed: 01/08/2023]
Abstract
The latest advances in the sequencing methods in head and neck squamous cell carcinoma (HNSCC) tissues have revolutionized our understanding of the disease by taking off the veil from the most frequent genetic alterations in the components of the oncogenic pathways. Among all the identified alterations, aberrancies in the genes attributed to the phosphoinositide 3-kinases (PI3K) axis have attracted special attention as they were altered in more than 90% of the tissues isolated from HNSCC patients. In fact, the association between these aberrancies and the increased risk of cancer metastasis suggested this axis as an "Achilles Heel" of HNSCC, which may be therapeutically targeted. The results of the clinical trials investigating the therapeutic potential of the inhibitors targeting the components of the PI3K axis in the treatment of HNSCC patients, either alone or in a combined-modal strategy, opened a new chapter in the treatment strategy of this malignancy. The present study aimed to review the importance of the PI3K axis in the pathogenesis of HNSCC and also provide a piece of information about the breakthroughs and challenges of PI3K inhibitors in the therapeutic strategies of the disease.
Collapse
Affiliation(s)
- Nader Akbari Dilmaghani
- Hearing Disorders Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Otolaryngology, Head and Neck Surgery, Loghman Hakim Educational Hospital, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ava Safaroghli-Azar
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Chen B, Dong C, Wang F, Wu J. Knockdown of NIR Suppresses Breast Cancer Cell Proliferation via Promoting FOXO3. Onco Targets Ther 2021; 14:637-651. [PMID: 33519211 PMCID: PMC7837597 DOI: 10.2147/ott.s287464] [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: 10/20/2020] [Accepted: 12/24/2020] [Indexed: 12/09/2022] Open
Abstract
Background Novel inhibitor of histone acetyltransferase repressor (NIR), a corepressor with a novel inhibitor of histone acetyltransferase (INHAT) activity, has been reported to be a negative modulator of p53 and a regulator of the cell cycle in cancer cells. However, the role of NIR in the progression of breast cancer remains elusive. Materials and Methods Oncomine database was used to analyze the mRNA levels and prognosis value of NIR in breast cancer. We performed loss-of-function and gain-of-function studies using lentivirus expressing shRNA targeting NIR, enhancer of zeste homolog 2 (EZH2) and forkhead box O3 (FOXO3) or lentivirus expressing NIR or FOXO3, respectively. Cell proliferation and colony formation assays were performed. Co-immunoprecipitation (Co-IP) and immunoprecipitation (IP) were performed to identify the interaction between NIR and polycomb repressive complex 2 (PRC2) subunits. ChIP assay was used to identify the enrichment of NIR, EZH2, H3K27ac and H3K27me3 at the FOXO3 promoter region and the regulation of H3K27 modification at the FOXO3 promoter by NIR. Results High levels of NIR expression were correlated with poor prognosis in breast cancer patients. Knockdown of NIR suppressed the proliferation of breast cancer cells. Mechanically, NIR was recruited by EZH2 to the promoter vicinity of FOXO3 via direct protein–protein interaction. Silencing NIR increased H3K27ac and decreased H3K27me3 levels at the FOXO3 promoter, resulting in enhancing FOXO3 expression. In accordance with this, growth inhibition of breast cancer cells caused by silencing of NIR could be reversed by FOXO3 knockdown. Conclusion NIR knockdown inhibited proliferation by switching the H3K27me3 and H3K27ac marks at the FOXO3 promoter to promote FOXO3 transcription, and this effect depends on the physical interaction between NIR and PRC2 in breast cancer cells. Our results suggest that NIR might be a potential target for breast cancer treatment.
Collapse
Affiliation(s)
- Bolin Chen
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, People's Republic of China
| | - Chengcheng Dong
- School of Biotechnology, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Fang Wang
- School of Biotechnology, Guilin Medical University, Guilin 541199, People's Republic of China
| | - Jiacai Wu
- School of Biotechnology, Guilin Medical University, Guilin 541199, People's Republic of China.,School of Pharmacy, Guilin Medical University, Guilin 541199, People's Republic of China
| |
Collapse
|
15
|
Datta N, Chakraborty S, Basu M, Ghosh MK. Tumor Suppressors Having Oncogenic Functions: The Double Agents. Cells 2020; 10:cells10010046. [PMID: 33396222 PMCID: PMC7824251 DOI: 10.3390/cells10010046] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/23/2020] [Accepted: 12/25/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer progression involves multiple genetic and epigenetic events, which involve gain-of-functions of oncogenes and loss-of-functions of tumor suppressor genes. Classical tumor suppressor genes are recessive in nature, anti-proliferative, and frequently found inactivated or mutated in cancers. However, extensive research over the last few years have elucidated that certain tumor suppressor genes do not conform to these standard definitions and might act as “double agents”, playing contrasting roles in vivo in cells, where either due to haploinsufficiency, epigenetic hypermethylation, or due to involvement with multiple genetic and oncogenic events, they play an enhanced proliferative role and facilitate the pathogenesis of cancer. This review discusses and highlights some of these exceptions; the genetic events, cellular contexts, and mechanisms by which four important tumor suppressors—pRb, PTEN, FOXO, and PML display their oncogenic potentials and pro-survival traits in cancer.
Collapse
Affiliation(s)
- Neerajana Datta
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India; (N.D.); (S.C.)
| | - Shrabastee Chakraborty
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India; (N.D.); (S.C.)
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Paraganas, West Bengal PIN-743372, India;
| | - Mrinal K. Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector–V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata-700032, India; (N.D.); (S.C.)
- Correspondence:
| |
Collapse
|
16
|
Sumida TS, Dulberg S, Schupp J, Stillwell HA, Axisa PP, Comi M, Lincoln M, Unterman A, Kaminski N, Madi A, Kuchroo VK, Hafler DA. Type I Interferon Transcriptional Network Regulates Expression of Coinhibitory Receptors in Human T cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.10.30.362947. [PMID: 33140047 PMCID: PMC7605554 DOI: 10.1101/2020.10.30.362947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
While inhibition of T cell co-inhibitory receptors has revolutionized cancer therapy, the mechanisms governing their expression on human T cells have not been elucidated. Type 1 interferon (IFN-I) modulates T cell immunity in viral infection, autoimmunity, and cancer, and may facilitate induction of T cell exhaustion in chronic viral infection 1,2 . Here we show that IFN-I regulates co-inhibitory receptors expression on human T cells, inducing PD-1/TIM-3/LAG-3 while surprisingly inhibiting TIGIT expression. High-temporal-resolution mRNA profiling of IFN-I responses enabled the construction of dynamic transcriptional regulatory networks uncovering three temporal transcriptional waves. Perturbation of key transcription factors on human primary T cells revealed both canonical and non-canonical IFN-I transcriptional regulators, and identified unique regulators that control expression of co-inhibitory receptors. To provide direct in vivo evidence for the role of IFN-I on co-inhibitory receptors, we then performed single cell RNA-sequencing in subjects infected with SARS-CoV-2, where viral load was strongly associated with T cell IFN-I signatures. We found that the dynamic IFN-I response in vitro closely mirrored T cell features with acute IFN-I linked viral infection, with high LAG3 and decreased TIGIT expression. Finally, our gene regulatory network identified SP140 as a key regulator for differential LAG3 and TIGIT expression. The construction of co-inhibitory regulatory networks induced by IFN-I with identification of unique transcription factors controlling their expression may provide targets for enhancement of immunotherapy in cancer, infectious diseases, and autoimmunity.
Collapse
Affiliation(s)
- Tomokazu S. Sumida
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Shai Dulberg
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jonas Schupp
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Helen A. Stillwell
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Pierre-Paul Axisa
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Michela Comi
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Matthew Lincoln
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Avraham Unterman
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Naftali Kaminski
- Section of Pulmonary, Critical Care and Sleep Medicine Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Asaf Madi
- Department of Pathology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Vijay K. Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David A. Hafler
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
17
|
Strassheim D, Karoor V, Nijmeh H, Weston P, Lapel M, Schaack J, Sullivan T, Dempsey EC, Stenmark KR, Gerasimovskaya E. c-Jun, Foxo3a, and c-Myc Transcription Factors are Key Regulators of ATP-Mediated Angiogenic Responses in Pulmonary Artery Vasa Vasorum Endothelial Cells. Cells 2020; 9:cells9020416. [PMID: 32054096 PMCID: PMC7072142 DOI: 10.3390/cells9020416] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/06/2020] [Accepted: 02/07/2020] [Indexed: 12/14/2022] Open
Abstract
Angiogenic vasa vasorum (VV) expansion plays an essential role in the pathogenesis of hypoxia-induced pulmonary hypertension (PH), a cardiovascular disease. We previously showed that extracellular ATP released under hypoxic conditions is an autocrine/paracrine, the angiogenic factor for pulmonary artery (PA) VV endothelial cells (VVECs), acting via P2Y purinergic receptors (P2YR) and the Phosphoinositide 3-kinase (PI3K)-Akt-Mammalian Target of Rapamycin (mTOR) signaling. To further elucidate the molecular mechanisms of ATP-mediated VV angiogenesis, we determined the profile of ATP-inducible transcription factors (TFs) in VVECs using a TranSignal protein/DNA array. C-Jun, c-Myc, and Foxo3 were found to be upregulated in most VVEC populations and formed nodes connecting several signaling networks. siRNA-mediated knockdown (KD) of these TFs revealed their critical role in ATP-induced VVEC angiogenic responses and the regulation of downstream targets involved in tissue remodeling, cell cycle control, expression of endothelial markers, cell adhesion, and junction proteins. Our results showed that c-Jun was required for the expression of ATP-stimulated angiogenic genes, c-Myc was repressive to anti-angiogenic genes, and Foxo3a predominantly controlled the expression of anti-apoptotic and junctional proteins. The findings from our study suggest that pharmacological targeting of the components of P2YR-PI3K-Akt-mTOR axis and specific TFs reduced ATP-mediated VVEC angiogenic response and may have a potential translational significance in attenuating pathological vascular remodeling.
Collapse
Affiliation(s)
- Derek Strassheim
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (V.K.); (T.S.); (E.C.D.); (K.R.S.)
| | - Vijaya Karoor
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (V.K.); (T.S.); (E.C.D.); (K.R.S.)
| | - Hala Nijmeh
- Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (H.N.); (P.W.); (M.L.)
| | - Philip Weston
- Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (H.N.); (P.W.); (M.L.)
| | - Martin Lapel
- Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (H.N.); (P.W.); (M.L.)
| | - Jerome Schaack
- Department of Microbiology, University of Colorado Denver, Aurora, CO 80045, USA;
| | - Timothy Sullivan
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (V.K.); (T.S.); (E.C.D.); (K.R.S.)
| | - Edward C. Dempsey
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (V.K.); (T.S.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Kurt R. Stenmark
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (V.K.); (T.S.); (E.C.D.); (K.R.S.)
- Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (H.N.); (P.W.); (M.L.)
| | - Evgenia Gerasimovskaya
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Aurora, CO 80045, USA; (D.S.); (V.K.); (T.S.); (E.C.D.); (K.R.S.)
- Department of Pediatrics, Division of Critical Care Medicine, University of Colorado Denver, Aurora, CO 80045, USA; (H.N.); (P.W.); (M.L.)
- Correspondence: ; Tel.: +1-303-724-5614
| |
Collapse
|
18
|
Ning JF, Stanciu M, Humphrey MR, Gorham J, Wakimoto H, Nishihara R, Lees J, Zou L, Martuza RL, Wakimoto H, Rabkin SD. Myc targeted CDK18 promotes ATR and homologous recombination to mediate PARP inhibitor resistance in glioblastoma. Nat Commun 2019; 10:2910. [PMID: 31266951 PMCID: PMC6606647 DOI: 10.1038/s41467-019-10993-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 06/13/2019] [Indexed: 12/19/2022] Open
Abstract
PARP inhibitors (PARPis) have clinical efficacy in BRCA-deficient cancers, but not BRCA-intact tumors, including glioblastoma (GBM). We show that MYC or MYCN amplification in patient-derived glioblastoma stem-like cells (GSCs) generates sensitivity to PARPi via Myc-mediated transcriptional repression of CDK18, while most tumors without amplification are not sensitive. In response to PARPi, CDK18 facilitates ATR activation by interacting with ATR and regulating ATR-Rad9/ATR-ETAA1 interactions; thereby promoting homologous recombination (HR) and PARPi resistance. CDK18 knockdown or ATR inhibition in GSCs suppressed HR and conferred PARPi sensitivity, with ATR inhibitors synergizing with PARPis or sensitizing GSCs. ATR inhibitor VE822 combined with PARPi extended survival of mice bearing GSC-derived orthotopic tumors, irrespective of PARPi-sensitivity. These studies identify a role of CDK18 in ATR-regulated HR. We propose that combined blockade of ATR and PARP is an effective strategy for GBM, even for low-Myc GSCs that do not respond to PARPi alone, and potentially other PARPi-refractory tumors. PARP inhibitors are mainly used to treat BRCA1/2 mutated cancers. Here, the authors show that MYC amplified glioblastomas are sensitive to PARP inhibition due to CDK18 repression, which impairs ATR regulated homologous recombination repair, and that ATR inhibition sensitises glioblastomas to PARP inhibition.
Collapse
Affiliation(s)
- Jian-Fang Ning
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA. .,Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, 55455, MN, USA.
| | - Monica Stanciu
- The David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Melissa R Humphrey
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, 02115, MA, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, 02115, MA, USA
| | - Reiko Nishihara
- Department of Pathology, Brigham's and Women's Hospital and Harvard Medical School, Boston, 02115, MA, USA
| | - Jacqueline Lees
- The David H. Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, 02139, MA, USA
| | - Lee Zou
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.,Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, 02129, MA, USA
| | - Robert L Martuza
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA
| | - Hiroaki Wakimoto
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA. .,Brain Tumor Stem Cell Laboratory, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.
| | - Samuel D Rabkin
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center and Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, MA, USA.
| |
Collapse
|
19
|
García-Gutiérrez L, Delgado MD, León J. MYC Oncogene Contributions to Release of Cell Cycle Brakes. Genes (Basel) 2019; 10:E244. [PMID: 30909496 PMCID: PMC6470592 DOI: 10.3390/genes10030244] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/16/2019] [Accepted: 03/18/2019] [Indexed: 12/12/2022] Open
Abstract
Promotion of the cell cycle is a major oncogenic mechanism of the oncogene c-MYC (MYC). MYC promotes the cell cycle by not only activating or inducing cyclins and CDKs but also through the downregulation or the impairment of the activity of a set of proteins that act as cell-cycle brakes. This review is focused on the role of MYC as a cell-cycle brake releaser i.e., how MYC stimulates the cell cycle mainly through the functional inactivation of cell cycle inhibitors. MYC antagonizes the activities and/or the expression levels of p15, ARF, p21, and p27. The mechanism involved differs for each protein. p15 (encoded by CDKN2B) and p21 (CDKN1A) are repressed by MYC at the transcriptional level. In contrast, MYC activates ARF, which contributes to the apoptosis induced by high MYC levels. At least in some cells types, MYC inhibits the transcription of the p27 gene (CDKN1B) but also enhances p27's degradation through the upregulation of components of ubiquitin ligases complexes. The effect of MYC on cell-cycle brakes also opens the possibility of antitumoral therapies based on synthetic lethal interactions involving MYC and CDKs, for which a series of inhibitors are being developed and tested in clinical trials.
Collapse
Affiliation(s)
- Lucía García-Gutiérrez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
- Current address: Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
| | - María Dolores Delgado
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC) CSIC-Universidad de Cantabria and Department of Biología Molecular, Universidad de Cantabria, 39011 Santander, Spain.
| |
Collapse
|
20
|
Small molecule inhibitor of c-Myc 10058-F4 inhibits proliferation and induces apoptosis in acute leukemia cells, irrespective of PTEN status. Int J Biochem Cell Biol 2019; 108:7-16. [DOI: 10.1016/j.biocel.2019.01.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/15/2018] [Accepted: 01/08/2019] [Indexed: 11/18/2022]
|
21
|
Liu Y, Ao X, Ding W, Ponnusamy M, Wu W, Hao X, Yu W, Wang Y, Li P, Wang J. Critical role of FOXO3a in carcinogenesis. Mol Cancer 2018; 17:104. [PMID: 30045773 PMCID: PMC6060507 DOI: 10.1186/s12943-018-0856-3] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
FOXO3a is a member of the FOXO subfamily of forkhead transcription factors that mediate a variety of cellular processes including apoptosis, proliferation, cell cycle progression, DNA damage and tumorigenesis. It also responds to several cellular stresses such as UV irradiation and oxidative stress. The function of FOXO3a is regulated by a complex network of processes, including post-transcriptional suppression by microRNAs (miRNAs), post-translational modifications (PTMs) and protein–protein interactions. FOXO3a is widely implicated in a variety of diseases, particularly in malignancy of breast, liver, colon, prostate, bladder, and nasopharyngeal cancers. Emerging evidences indicate that FOXO3a acts as a tumor suppressor in cancer. FOXO3a is frequently inactivated in cancer cell lines by mutation of the FOXO3a gene or cytoplasmic sequestration of FOXO3a protein. And its inactivation is associated with the initiation and progression of cancer. In experimental studies, overexpression of FOXO3a inhibits the proliferation, tumorigenic potential, and invasiveness of cancer cells, while silencing of FOXO3a results in marked attenuation in protection against tumorigenesis. The role of FOXO3a in both normal physiology as well as in cancer development have presented a great challenge to formulating an effective therapeutic strategy for cancer. In this review, we summarize the recent findings and overview of the current understanding of the influence of FOXO3a in cancer development and progression.
Collapse
Affiliation(s)
- Ying Liu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xiang Ao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Wei Ding
- Department of comprehensive internal medicine, Affiliated Hospital, Qingdao University, Qingdao, 266003, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Wei Wu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Xiaodan Hao
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Wanpeng Yu
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Yifei Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China.
| | - Jianxun Wang
- Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
22
|
Alternative NF-κB signaling promotes colorectal tumorigenesis through transcriptionally upregulating Bcl-3. Oncogene 2018; 37:5887-5900. [PMID: 29973688 DOI: 10.1038/s41388-018-0363-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 03/25/2018] [Accepted: 05/21/2018] [Indexed: 01/20/2023]
Abstract
Multiple studies have shown that chronic inflammation is closely related to the occurrence and development of colorectal cancer (CRC). Classical NF-κB signaling, the key factor in controlling inflammation, has been found to be of great importance to CRC development. However, the role of alternative NF-κB signaling in CRC is still elusive. Here, we found aberrant constitutive activation of alternative NF-κB signaling both in CRC tissue and CRC cells. Knockdown of RelB downregulates c-Myc and upregulates p27Kip1 protein level, which inhibits CRC cell proliferation and retards CRC xenograft growth. Conversely, overexpression of RelB increases proliferation of CRC cells. In addition, we revealed a significant correlation between Bcl-3 and RelB in CRC tissues. The expression of RelB was consistent with the expression of Bcl-3 and the phosphorylation of Bcl-3 downstream proteins p-Akt (S473) and p-GSK3β (S9). Bcl-3 overexpression can restore the phenotype changes caused by RelB knockdown. Importantly, we demonstrated that alternative NF-κB transcriptional factor (p52:RelB) can directly bind to the promoter region of Bcl-3 gene and upregulate its transcription. Moreover, the expression of RelB, NF-κB2 p52, and Bcl-3 was associated with poor survival of CRC patients. Taken together, these results represent that alternative NF-κB signaling may function as an oncogenic driver in CRC, and also provide new ideas and research directions for the pathogenesis, prevention, and treatment of other inflammatory-related diseases.
Collapse
|
23
|
Zhao X, Xie T, Dai T, Zhao W, Li J, Xu R, Jiang C, Li P, Deng J, Su X, Ma N. CHP2 Promotes Cell Proliferation in Breast Cancer via Suppression of FOXO3a. Mol Cancer Res 2018; 16:1512-1522. [PMID: 29967111 DOI: 10.1158/1541-7786.mcr-18-0157] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 05/10/2018] [Accepted: 06/13/2018] [Indexed: 11/16/2022]
Affiliation(s)
- Xiaohui Zhao
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Tian Xie
- Key Laboratory of Protein Modification and Degradation School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Ting Dai
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Wenhui Zhao
- Juancheng People's Hospital, Juancheng, China
| | - Jing Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Rui Xu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Chao Jiang
- Department of Cancer Center, People's Hospital of Baoan District, Shenzhen, China
| | - Peiqiong Li
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China
| | - Junyao Deng
- Department of Histology and Embryology, Southern Medical University, Guangzhou, China
| | - Xiaobo Su
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, China.
| | - Ningfang Ma
- Key Laboratory of Protein Modification and Degradation School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
24
|
Bachs O, Gallastegui E, Orlando S, Bigas A, Morante-Redolat JM, Serratosa J, Fariñas I, Aligué R, Pujol MJ. Role of p27 Kip1 as a transcriptional regulator. Oncotarget 2018; 9:26259-26278. [PMID: 29899857 PMCID: PMC5995243 DOI: 10.18632/oncotarget.25447] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 05/01/2018] [Indexed: 12/16/2022] Open
Abstract
The protein p27Kip1 is a member of the Cip/Kip family of cyclin-dependent kinase (Cdk) inhibitors. It interacts with both the catalytic and the regulatory subunit (cyclin) and introduces a region into the catalytic cleave of the Cdk inducing its inactivation. Its inhibitory capacity can be modulated by specific tyrosine phosphorylations. p27Kip1 also behaves as a transcriptional regulator. It associates with specific chromatin domains through different transcription factors. ChIP on chip, ChIP-seq and expression microarray analysis allowed the identification of the transcriptional programs regulated by p27Kip1. Thus, important cellular functions as cell division cycle, respiration, RNA processing, translation and cell adhesion, are under p27Kip1 regulation. Moreover, genes involved in pathologies as cancer and neurodegeneration are also regulated by p27Kip1, suggesting its implication in these pathologies. The carboxyl moiety of p27Kip1 can associate with different proteins, including transcriptional regulators. In contrast, its NH2-terminal region specifically interacts with cyclin-Cdk complexes. The general mechanistic model of how p27Kip1 regulates transcription is that it associates by its COOH region to the transcriptional regulators on the chromatin and by the NH2-domain to cyclin-Cdk complexes. After Cdk activation it would phosphorylate the specific targets on the chromatin leading to gene expression. This model has been demonstrated to apply in the transcriptional regulation of p130/E2F4 repressed genes involved in cell cycle progression. We summarize in this review our current knowledge on the role of p27Kip1 in the regulation of transcription, on the transcriptional programs under its regulation and on its relevance in pathologies as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Oriol Bachs
- Department of Biomedical Sciences, Faculty of Medicine, University of Barcelona, IDIBAPS, CIBERONC, Barcelona, Spain
| | - Edurne Gallastegui
- Department of Biomedical Sciences, Faculty of Medicine, University of Barcelona, IDIBAPS, CIBERONC, Barcelona, Spain
| | - Serena Orlando
- Department of Biomedical Sciences, Faculty of Medicine, University of Barcelona, IDIBAPS, CIBERONC, Barcelona, Spain
| | - Anna Bigas
- Program in Cancer Research, Institut Hospital Del Mar d'Investigacions Mèdiques (IMIM), CIBERONC, Barcelona, Spain
| | - José Manuel Morante-Redolat
- Departamento de Biología Celular, Biología Funcional y Antropología Física and ERI de Biotecnología y Biomedicina, CIBERNED, Universidad de Valencia, Valencia, Spain
| | - Joan Serratosa
- Department of Cerebral Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), IDIBAPS, Barcelona, Spain
| | - Isabel Fariñas
- Departamento de Biología Celular, Biología Funcional y Antropología Física and ERI de Biotecnología y Biomedicina, CIBERNED, Universidad de Valencia, Valencia, Spain
| | - Rosa Aligué
- Department of Biomedical Sciences, Faculty of Medicine, University of Barcelona, IDIBAPS, CIBERONC, Barcelona, Spain
| | - Maria Jesús Pujol
- Department of Biomedical Sciences, Faculty of Medicine, University of Barcelona, IDIBAPS, CIBERONC, Barcelona, Spain
| |
Collapse
|
25
|
Oak C, Khalifa AO, Isali I, Bhaskaran N, Walker E, Shukla S. Diosmetin suppresses human prostate cancer cell proliferation through the induction of apoptosis and cell cycle arrest. Int J Oncol 2018; 53:835-843. [PMID: 29767250 PMCID: PMC6017185 DOI: 10.3892/ijo.2018.4407] [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: 12/08/2017] [Accepted: 03/30/2018] [Indexed: 12/15/2022] Open
Abstract
Diosmetin, a plant flavonoid, has been shown to exert promising effects on prostate cancer cells as an anti‑proliferative and anticancer agent. In this study, using western blot analysis for protein expression and flow cytometry for cell cycle analysis, we determined that the treatment of the LNCaP and PC‑3 prostate cancer cells with diosmetin resulted in a marked decrease in cyclin D1, Cdk2 and Cdk4 expression levels (these proteins remain active in the G0‑G1 phases of the cell cycle). These changes were accompanied by a decrease in c-Myc and Bcl-2 expression, and by an increase in Bax, p27Kip1 and FOXO3a protein expression, which suggests the potential modulatory effects of diosmetin on protein transcription. The treatment of prostate cancer cells with diosmetin set in motion an apoptotic machinery by inhibiting X-linked inhibitor of apoptosis (XIAP) and increasing cleaved PARP and cleaved caspase-3 expression levels. On the whole, the findings of this study provide an in-depth analysis of the molecular mechanisms responsible for the regulatory effects of diosmetin on key molecules that perturb the cell cycle to inhibit cell growth, and suggest that diosmetin may prove to be an effective anticancer agent for use in the treatment of prostate cancer in the future.
Collapse
Affiliation(s)
- Christine Oak
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ahmad O Khalifa
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ilaha Isali
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Natarajan Bhaskaran
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Ethan Walker
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sanjeev Shukla
- Department of Urology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
26
|
van Doeselaar S, Burgering BMT. FOXOs Maintaining the Equilibrium for Better or for Worse. Curr Top Dev Biol 2018; 127:49-103. [PMID: 29433740 DOI: 10.1016/bs.ctdb.2017.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A paradigm shift is emerging within the FOXO field and accumulating evidence indicates that we need to reappreciate the role of FOXOs, at least in cancer development. Here, we discuss the possibility that FOXOs are both tumor suppressors as well as promoters of tumor progression. This is mostly dependent on the biological context. Critical to this dichotomous role is the notion that FOXOs are central in preserving cellular homeostasis in redox control, genomic stability, and protein turnover. From this perspective, a paradoxical role in both suppressing and enhancing tumor progression can be reconciled. As many small molecules targeting the PI3K pathway are developed by big pharmaceutical companies and/or are in clinical trial, we will discuss what the consequences may be for the context-dependent role of FOXOs in tumor development in treatment options based on active PI3K signaling in tumors.
Collapse
Affiliation(s)
- Sabina van Doeselaar
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
27
|
TRIP6 promotes cell proliferation in hepatocellular carcinoma via suppression of FOXO3a. Biochem Biophys Res Commun 2017; 494:594-601. [DOI: 10.1016/j.bbrc.2017.10.117] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 10/23/2017] [Indexed: 12/16/2022]
|
28
|
Decitabine-Induced Changes in Human Myelodysplastic Syndrome Cell Line SKM-1 Are Mediated by FOXO3A Activation. J Immunol Res 2017; 2017:4302320. [PMID: 29124072 PMCID: PMC5662805 DOI: 10.1155/2017/4302320] [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: 04/09/2017] [Revised: 07/12/2017] [Accepted: 07/20/2017] [Indexed: 01/03/2023] Open
Abstract
The epigenetic silencing of tumor suppressor genes in myelodysplastic syndromes (MDS) can potentially confer a growth advantage to individual cellular clones. Currently, the recommended treatment for patients with high-risk MDS is the methylation agent decitabine (DAC), a drug that can induce the reexpression of silenced tumor suppressor genes. We investigated the effects of DAC treatment on the myeloid MDS cell line SKM-1 and investigated the role of FOXO3A, a potentially tumor-suppressive transcription factor, by silencing its expression prior to DAC treatment. We found that FOXO3A exists in an inactive, hyperphosphorylated form in SKM-1 cells, but that DAC both induces FOXO3A expression and reactivates the protein by reducing its phosphorylation level. Furthermore, we show that this FOXO3A activation is responsible for the DAC-induced differentiation of SKM-1 cells into monocytes, as well as for SKM-1 cell cycle arrest, apoptosis, and autophagy. Collectively, these results suggest that FOXO3A reactivation may contribute to the therapeutic effects of DAC in MDS.
Collapse
|
29
|
Bencivenga D, Caldarelli I, Stampone E, Mancini FP, Balestrieri ML, Della Ragione F, Borriello A. p27 Kip1 and human cancers: A reappraisal of a still enigmatic protein. Cancer Lett 2017; 403:354-365. [DOI: 10.1016/j.canlet.2017.06.031] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/21/2022]
|
30
|
Breast cancer suppression by aplysin is associated with inhibition of PI3K/AKT/FOXO3a pathway. Oncotarget 2017; 8:63923-63934. [PMID: 28969041 PMCID: PMC5609973 DOI: 10.18632/oncotarget.19209] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 06/04/2017] [Indexed: 11/28/2022] Open
Abstract
Aplysin, a bromosesquiterpene isolated from Aplysia kurodai, was explored as a potential anti-breast cancer agent by us. However, the mechanisms underlying the anticarcinogenic effect of aplysin remain unclear. Here, aplysin was found to remarkably suppress tumor growth in vivo, inhibit cell proliferation and promote apoptosis in vitro. Additionally, we demonstrated that aplysin attained these effects in part by down-regulating PI3K/AKT/FOXO3a signaling pathway. Aplysin treatment inhibited the phosphorylation levels of AKT (Ser-473) and AKT-dependent phosphorylation of FOXO3a (Ser-253) in breast cancer cell lines and breast cancer tissues. The expression levels of FOXO3a-targeted genes were also destabilized by aplysin, cyclin D1 and Bcl-XL were declined; however, p21CIP1, p27KIP1, Bim, TRAIL and FasL were increased both in vivo and in vitro. Furthermore, activation of the PI3K/AKT signaling pathway by an activator and silencing of FOXO3a by shRNA protected the cells from aplysin mediated growth suppression and apoptosis. In summary, our findings revealed that aplysin could suppress breast cancer progression by inhibiting PI3K/AKT/FOXO3a pathway, thereby suggesting a potential role of aplysin as a chemoprevention drug for patients with breast cancer.
Collapse
|
31
|
MYC Modulation around the CDK2/p27/SKP2 Axis. Genes (Basel) 2017; 8:genes8070174. [PMID: 28665315 PMCID: PMC5541307 DOI: 10.3390/genes8070174] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 06/23/2017] [Accepted: 06/24/2017] [Indexed: 12/20/2022] Open
Abstract
MYC is a pleiotropic transcription factor that controls a number of fundamental cellular processes required for the proliferation and survival of normal and malignant cells, including the cell cycle. MYC interacts with several central cell cycle regulators that control the balance between cell cycle progression and temporary or permanent cell cycle arrest (cellular senescence). Among these are the cyclin E/A/cyclin-dependent kinase 2 (CDK2) complexes, the CDK inhibitor p27KIP1 (p27) and the E3 ubiquitin ligase component S-phase kinase-associated protein 2 (SKP2), which control each other by forming a triangular network. MYC is engaged in bidirectional crosstalk with each of these players; while MYC regulates their expression and/or activity, these factors in turn modulate MYC through protein interactions and post-translational modifications including phosphorylation and ubiquitylation, impacting on MYC's transcriptional output on genes involved in cell cycle progression and senescence. Here we elaborate on these network interactions with MYC and their impact on transcription, cell cycle, replication and stress signaling, and on the role of other players interconnected to this network, such as CDK1, the retinoblastoma protein (pRB), protein phosphatase 2A (PP2A), the F-box proteins FBXW7 and FBXO28, the RAS oncoprotein and the ubiquitin/proteasome system. Finally, we describe how the MYC/CDK2/p27/SKP2 axis impacts on tumor development and discuss possible ways to interfere therapeutically with this system to improve cancer treatment.
Collapse
|
32
|
miR-1307 promotes the proliferation of prostate cancer by targeting FOXO3A. Biomed Pharmacother 2017; 88:430-435. [DOI: 10.1016/j.biopha.2016.11.120] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/15/2016] [Accepted: 11/27/2016] [Indexed: 11/21/2022] Open
|
33
|
Zhou L, Li R, Liu C, Sun T, Htet Aung LH, Chen C, Gao J, Zhao Y, Wang K. Foxo3a inhibits mitochondrial fission and protects against doxorubicin-induced cardiotoxicity by suppressing MIEF2. Free Radic Biol Med 2017; 104:360-370. [PMID: 28137654 DOI: 10.1016/j.freeradbiomed.2017.01.037] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 01/24/2017] [Accepted: 01/26/2017] [Indexed: 10/20/2022]
Abstract
Doxorubicin (DOX) as a chemotherapeutic drug is widely used to treat a variety of human tumors. However, a major factor limiting its clinical use is its cardiotoxicity. The molecular components and detailed mechanisms regulating DOX-induced cardiotoxicity remain largely unidentified. Here we report that Foxo3a is downregulated in the cardiomyocyte and mouse heart in response to DOX treatment. Foxo3a attenuates DOX-induced mitochondrial fission and apoptosis in cardiomyocytes. Cardiac specific Foxo3a transgenic mice show reduced mitochondrial fission, apoptosis and cardiotoxicity upon DOX administration. Furthermore, Foxo3a directly targets mitochondrial dynamics protein of 49kDa (MIEF2) and suppresses its expression at transcriptional level. Knockdown of MIEF2 reduces DOX-induced mitochondrial fission and apoptosis in cardiomyocytes and in vivo. Also, knockdown of MIEF2 protects heart from DOX-induced cardiotoxicity. Our study identifies a novel pathway composed of Foxo3a and MIEF2 that mediates DOX cardiotoxicity. This discovery provides a promising therapeutic strategy for the treatment of cancer therapy and cardioprotection.
Collapse
Affiliation(s)
- Luyu Zhou
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Ruibei Li
- School of Professional Studies, Northwestern University, Chicago, IL 60611, USA
| | - Cuiyun Liu
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Teng Sun
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Lynn Htet Htet Aung
- College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Chao Chen
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Jinning Gao
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Yanfang Zhao
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China
| | - Kun Wang
- Center for Developmental Cardiology, Institute for Translational Medicine, College of Medicine, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
34
|
Chen YC, Chien LH, Huang BM, Chia YC, Chiu HF. Aqueous Extracts ofToona sinensisLeaves Inhibit Renal Carcinoma Cell Growth and Migration Through JAK2/stat3, Akt, MEK/ERK, and mTOR/HIF-2α Pathways. Nutr Cancer 2016; 68:654-66. [DOI: 10.1080/01635581.2016.1158292] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
35
|
Hnit SST, Xie C, Yao M, Holst J, Bensoussan A, De Souza P, Li Z, Dong Q. p27(Kip1) signaling: Transcriptional and post-translational regulation. Int J Biochem Cell Biol 2015; 68:9-14. [PMID: 26279144 DOI: 10.1016/j.biocel.2015.08.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 08/08/2015] [Accepted: 08/10/2015] [Indexed: 11/30/2022]
Abstract
p27(Kip1) is an inhibitor of a broad spectrum of cyclin-dependent kinases (CDKs), and the loss of a single p27(Kip1) allele is thereby sufficient to increase tumor incidence via CDK-mediated cell cycle entry. As such, down-regulation of p27(Kip1) protein levels, in particular nuclear expressed p27(Kip1), is implicated in both disease progression and poor prognosis in a variety of cancers. p27(Kip1) expression is positively regulated by the transcription factor MENIN, and inhibited by oncogenic transcription factors MYC and PIM. However, regulation of p27(Kip1) protein expression and function is predominantly through post-translational modifications that alter both the cellular localization and the extent of E3 ubiquitin ligase-mediated degradation. Phosphorylation of p27(Kip1) at Thr(187) and Ser(10) is a prerequisite for its degradation via the E3 ubiquitin ligases SKP2 (nuclear) and KPC (cytoplasmic), respectively. Additionally, Ser(10) phosphorylated p27(Kip1) is predominantly localized in the cytoplasm due to the nuclear export protein CRM1. Another E3 ubiquitin ligase, PIRH2, degrades p27(Kip1) in both the cytoplasm and nucleus independent of phosphorylation state. As such, inhibition of cell cycle entry and progression in a variety of cancers may be achieved with therapies designed to correct p27(Kip1) localization and/or block its degradation.
Collapse
Affiliation(s)
- Su Su Thae Hnit
- School of Science and Health, University of Western Sydney, Australia
| | - Chanlu Xie
- School of Science and Health, University of Western Sydney, Australia
| | - Mu Yao
- Central Clinical School and Charles Perkins Centre, The University of Sydney and Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Jeff Holst
- Origins of Cancer Program, Centenary Institute, Camperdown, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alan Bensoussan
- National Institute of Complementary Medicine, University of Western Sydney, Australia
| | - Paul De Souza
- School of Medicine, University of Western Sydney, Australia
| | - Zhong Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| | - Qihan Dong
- School of Science and Health, University of Western Sydney, Australia; Central Clinical School and Charles Perkins Centre, The University of Sydney and Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, Australia; School of Medicine, University of Western Sydney, Australia.
| |
Collapse
|
36
|
Huang J, Schriefer AE, Yang W, Cliften PF, Rudnick DA. Identification of an epigenetic signature of early mouse liver regeneration that is disrupted by Zn-HDAC inhibition. Epigenetics 2015; 9:1521-31. [PMID: 25482284 DOI: 10.4161/15592294.2014.983371] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Liver regeneration has been well studied with hope of discovering strategies to improve liver disease outcomes. Nevertheless, the signals that initiate such regeneration remain incompletely defined, and translation of mechanism-based pro-regenerative interventions into new treatments for hepatic diseases has not yet been achieved. We previously reported the isoform-specific regulation and essential function of zinc-dependent histone deacetylases (Zn-HDACs) during mouse liver regeneration. Those data suggest that epigenetically regulated anti-proliferative genes are deacetylated and transcriptionally suppressed by Zn-HDAC activity or that pro-regenerative factors are acetylated and induced by such activity in response to partial hepatectomy (PH). To investigate these possibilities, we conducted genome-wide interrogation of the liver histone acetylome during early PH-induced liver regeneration in mice using acetyL-histone chromatin immunoprecipitation and next generation DNA sequencing. We also compared the findings of that study to those seen during the impaired regenerative response that occurs with Zn-HDAC inhibition. The results reveal an epigenetic signature of early liver regeneration that includes both hyperacetylation of pro-regenerative factors and deacetylation of anti-proliferative and pro-apoptotic genes. Our data also show that administration of an anti-regenerative regimen of the Zn-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) not only disrupts gene-specific pro-regenerative changes in liver histone deacetylation but also reverses PH-induced effects on histone hyperacetylation. Taken together, these studies offer new insight into and suggest novel hypotheses about the epigenetic mechanisms that regulate liver regeneration.
Collapse
Affiliation(s)
- Jiansheng Huang
- a Department of Pediatrics ; Washington University School of Medicine ; St. Louis , MO USA
| | | | | | | | | |
Collapse
|
37
|
Xie L, Ritz O, Leithäuser F, Guan H, Färbinger J, Weitzer CD, Gehringer F, Bruederlein S, Holzmann K, Vogel MJ, Möller P, Wirth T, Ushmorov A. FOXO1 downregulation contributes to the oncogenic program of primary mediastinal B-cell lymphoma. Oncotarget 2015; 5:5392-402. [PMID: 24977668 PMCID: PMC4170625 DOI: 10.18632/oncotarget.2107] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Recently we have shown that the transcription factor FOXO1, highly expressed in B cells, is downregulated in classical Hodgkin lymphoma (cHL). As primary mediastinal B cell lymphoma (PMBL) has similarities with the cHL transcription program we investigated FOXO1 expression in this entity. By using immunohistochemistry we found that FOXO1 was absent or expressed at low levels in 19 of 20 primary PMBL cases. PMBL cell lines reproduce the low FOXO1 expression observed in primary cases. By analyzing gene expression profiling data we found that FOXO1 expression inversely correlated with JAK2 in PMBL cases. Targeting JAK2 activity by the small molecular weight inhibitor TG101348 resulted in upregulation of FOXO1 mRNA and protein expression in MedB-1 and U2940 cell lines, and the MYC inhibitor 10058-F4 increased FOXO1 mRNA in MedB-1 cells. Moreover, in MedB-1 cells FOXO1 expression was strongly upregulated by the inhibitor of DNA methylation 5-aza-2-deoxycytidine and by the histone deacetylase inhibitor trichostatin A. Since FOXO1 promoter was unmethylated, this effect is most likely indirect. FOXO1 activation in the FOXO1-negative MedB-1 cell line led to growth arrest and apoptosis, which was accompanied by repression of MYC and BCL2L1/BCLxL. Thus, FOXO1 repression might contribute to the oncogenic program and phenotype of PMBL.
Collapse
Affiliation(s)
- Linka Xie
- Cancer Center of Union Hospital, Tongji Medical College, HuaZhong University of Science and Technology, Wuhan, China. Institute of Physiological Chemistry, University of Ulm, Germany
| | - Olga Ritz
- Institute of Pathology, University of Ulm, Germany
| | | | - Hanfeng Guan
- Institute of Physiological Chemistry, University of Ulm, Germany. Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Hua Zhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | - Marion J Vogel
- Institute of Physiological Chemistry, University of Ulm, Germany
| | - Peter Möller
- Institute of Pathology, University of Ulm, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, University of Ulm, Germany
| | - Alexey Ushmorov
- Institute of Physiological Chemistry, University of Ulm, Germany
| |
Collapse
|
38
|
Jezkova J, Williams JS, Jones-Hutchins F, Sammut SJ, Gollins S, Cree I, Coupland S, McFarlane RJ, Wakeman JA. Brachyury regulates proliferation of cancer cells via a p27Kip1-dependent pathway. Oncotarget 2015; 5:3813-22. [PMID: 25003467 PMCID: PMC4116522 DOI: 10.18632/oncotarget.1999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The T-box transcription factor Brachyury is expressed in a number of tumour types and has been demonstrated to have cancer inducing properties. To date, it has been linked to cancer associated induction of epithelial to mesenchymal transition, tumour metastasis and expression of markers for cancer stem-like cells. Taken together, these findings indicate that Brachyury plays an important role in the progression of cancer, although the mechanism through which it functions is poorly understood. Here we show that Brachyury regulates the potential of Brachyury-positive colorectal cancer cells to proliferate and reduced levels of Brachyury result in inhibition of proliferation, with features consistent with the cells entering a quiescent-like state. This inhibition of proliferation is dependent upon p27Kip1 demonstrating that Brachyury acts to modulate cellular proliferative fate in colorectal cancer cells in a p27Kip1-dependent manner. Analysis of patient derived colorectal tumours reveals a heterogeneous localisation of Brachyury (in the nucleolus, nucleus and cytoplasm) indicating the potential complexity of the regulatory role of Brachyury in solid colorectal tumours.
Collapse
Affiliation(s)
- Jana Jezkova
- North West Cancer Research Institute, College of Natural Sciences, Bangor, Gwynedd, UK
| | | | | | | | | | | | | | - Ramsay J McFarlane
- North West Cancer Research Institute, College of Natural Sciences, Bangor, Gwynedd, UK; NISCHR Cancer Genetics Biomedical Research Unit, Welsh Government, Cathays Park, Cardiff, UK
| | - Jane A Wakeman
- North West Cancer Research Institute, College of Natural Sciences, Bangor, Gwynedd, UK
| |
Collapse
|
39
|
Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer. Oncogene 2015; 35:691-701. [PMID: 25639871 PMCID: PMC4522411 DOI: 10.1038/onc.2014.469] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 12/16/2014] [Accepted: 12/19/2014] [Indexed: 12/26/2022]
Abstract
The myc oncogene is overexpressed in almost half of all breast and ovarian cancers, but attempts at therapeutic interventions against myc have proven to be challenging. Myc regulates multiple biological processes, including the cell cycle, and as such is associated with cell proliferation and tumor progression. We identified a protein signature of high myc, low p27 and high phospho-Rb significantly correlated with poor patient survival in breast and ovarian cancers. Screening of a miRNA library by functional proteomics in multiple cell lines and integration of data from patient tumors revealed a panel of five microRNAs (miRNAs) (miR-124, miR-365, miR-34b*, miR-18a and miR-506) as potential tumor suppressors capable of reversing the p27/myc/phospho-Rb protein signature. Mechanistic studies revealed an RNA-activation function of miR-124 resulting in direct induction of p27 protein levels by binding to and inducing transcription on the p27 promoter region leading to a subsequent G1 arrest. Additionally, in vivo studies utilizing a xenograft model demonstrated that nanoparticle-mediated delivery of miR-124 could reduce tumor growth and sensitize cells to etoposide, suggesting a clinical application of miRNAs as therapeutics to target the functional effect of myc on tumor growth.
Collapse
|
40
|
Androgen suppresses the proliferation of androgen receptor-positive castration-resistant prostate cancer cells via inhibition of Cdk2, CyclinA, and Skp2. PLoS One 2014; 9:e109170. [PMID: 25271736 PMCID: PMC4182885 DOI: 10.1371/journal.pone.0109170] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/28/2014] [Indexed: 12/21/2022] Open
Abstract
The majority of prostate cancer (PCa) patient receiving androgen ablation therapy eventually develop castration-resistant prostate cancer (CRPC). We previously reported that androgen treatment suppresses Skp2 and c-Myc through androgen receptor (AR) and induced G1 cell cycle arrest in androgen-independent LNCaP 104-R2 cells, a late stage CRPC cell line model. However, the mechanism of androgenic regulation of Skp2 in CRPC cells was not fully understood. In this study, we investigated the androgenic regulation of Skp2 in two AR-positive CRPC cell line models, the LNCaP 104-R1 and PC-3AR Cells. The former one is an early stage androgen-independent LNCaP cells, while the later one is PC-3 cells re-expressing either wild type AR or mutant LNCaP AR. Proliferation of LNCaP 104-R1 and PC-3AR cells is not dependent on but is suppressed by androgen. We observed in this study that androgen treatment reduced protein expression of Cdk2, Cdk7, Cyclin A, cyclin H, Skp2, c-Myc, and E2F-1; lessened phosphorylation of Thr14, Tyr15, and Thr160 on Cdk2; decreased activity of Cdk2; induced protein level of p27Kip1; and caused G1 cell cycle arrest in LNCaP 104-R1 cells and PC-3AR cells. Overexpression of Skp2 protein in LNCaP 104-R1 or PC-3AR cells partially blocked accumulation of p27Kip1 and increased Cdk2 activity under androgen treatment, which partially blocked the androgenic suppressive effects on proliferation and cell cycle. Analyzing on-line gene array data of 214 normal and PCa samples indicated that gene expression of Skp2, Cdk2, and cyclin A positively correlates to each other, while Cdk7 negatively correlates to these genes. These observations suggested that androgen suppresses the proliferation of CRPC cells partially through inhibition of Cyclin A, Cdk2, and Skp2.
Collapse
|
41
|
Caraballo JM, Acosta JC, Cortés MA, Albajar M, Gómez-Casares MT, Batlle-López A, Cuadrado MA, Onaindia A, Bretones G, Llorca J, Piris MA, Colomer D, León J. High p27 protein levels in chronic lymphocytic leukemia are associated to low Myc and Skp2 expression, confer resistance to apoptosis and antagonize Myc effects on cell cycle. Oncotarget 2014; 5:4694-708. [PMID: 25051361 PMCID: PMC4148092 DOI: 10.18632/oncotarget.2100] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 06/09/2014] [Indexed: 12/29/2022] Open
Abstract
Myc (c-Myc) counteracts p27 effects, and low p27 usually correlates with high Myc expression in human cancer. However there is no information on the co-expression of both genes in chronic lymphocytic leukemia (CLL). We found a lack of correlation between RNA and protein levels of p27 and Myc in CLL cells, so we determined the protein levels by immunoblot in 107 cases of CLL. We observed a high p27 protein expression in CLL compared to normal B cells. Ectopic p27 expression in a CLL-derived cell line resulted in cell death resistance. Surprisingly, Myc expression was very low or undetectable in most CLL cases analyzed, with a clear correlation between high p27 and low Myc protein levels. This was associated with low Skp2 expression, which is consistent with the Skp2 role in p27 degradation and with SKP2 being a Myc target gene. High Myc expression did not correlate with leukemia progression, despite that cell cycle-related Myc target genes were upregulated. However, biochemical analysis showed that the high p27 levels inhibited cyclin-Cdk complexes even in Myc expressing CLL cells. Our data suggest that the combination of high p27 and low Myc is a marker of CLL cells which is mediated by Skp2.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Apoptosis/genetics
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Cycle/genetics
- Cell Line, Tumor
- Cyclin-Dependent Kinase Inhibitor p27/genetics
- Cyclin-Dependent Kinase Inhibitor p27/metabolism
- Cyclins/genetics
- Cyclins/metabolism
- Drug Resistance, Neoplasm/genetics
- Female
- Gene Expression Regulation, Leukemic
- Humans
- Immunoblotting
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Microscopy, Fluorescence
- Middle Aged
- Proto-Oncogene Proteins c-myb/genetics
- Proto-Oncogene Proteins c-myb/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- S-Phase Kinase-Associated Proteins/genetics
- S-Phase Kinase-Associated Proteins/metabolism
Collapse
Affiliation(s)
- Juan M. Caraballo
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-Sodercan, and Dpt. of. Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Juan C. Acosta
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-Sodercan, and Dpt. of. Biología Molecular, Universidad de Cantabria, Santander, Spain
- Present address: Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, UK
| | | | - Marta Albajar
- Servicio de Hematologia, Hospital Marqués de Valdecilla and Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | | | - Ana Batlle-López
- Servicio de Hematologia, Hospital Marqués de Valdecilla and Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - M. Angeles Cuadrado
- Servicio de Hematologia, Hospital Marqués de Valdecilla and Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Arantza Onaindia
- Servicio de Anatomía Patológica, Hospital Marqués de Valdecilla and Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Gabriel Bretones
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-Sodercan, and Dpt. of. Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Javier Llorca
- Group of Epidemiology and Computational Biology, Universidad de Cantabria-IDIVAL, Santander, Spain and CIBER Epidemiología y Salud Pública (CIBERESP), Spain
| | - Miguel A. Piris
- Servicio de Anatomía Patológica, Hospital Marqués de Valdecilla and Instituto de Investigación Marqués de Valdecilla (IDIVAL), Santander, Spain
| | - Dolors Colomer
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Barcelona, Spain
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-Sodercan, and Dpt. of. Biología Molecular, Universidad de Cantabria, Santander, Spain
| |
Collapse
|
42
|
Myc and its interactors take shape. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:469-83. [PMID: 24933113 DOI: 10.1016/j.bbagrm.2014.06.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 06/03/2014] [Accepted: 06/04/2014] [Indexed: 12/11/2022]
Abstract
The Myc oncoprotein is a key contributor to the development of many human cancers. As such, understanding its molecular activities and biological functions has been a field of active research since its discovery more than three decades ago. Genome-wide studies have revealed Myc to be a global regulator of gene expression. The identification of its DNA-binding partner protein, Max, launched an area of extensive research into both the protein-protein interactions and protein structure of Myc. In this review, we highlight key insights with respect to Myc interactors and protein structure that contribute to the understanding of Myc's roles in transcriptional regulation and cancer. Structural analyses of Myc show many critical regions with transient structures that mediate protein interactions and biological functions. Interactors, such as Max, TRRAP, and PTEF-b, provide mechanistic insight into Myc's transcriptional activities, while others, such as ubiquitin ligases, regulate the Myc protein itself. It is appreciated that Myc possesses a large interactome, yet the functional relevance of many interactors remains unknown. Here, we discuss future research trends that embrace advances in genome-wide and proteome-wide approaches to systematically elucidate mechanisms of Myc action. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.
Collapse
|
43
|
Bretones G, Delgado MD, León J. Myc and cell cycle control. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2014; 1849:506-16. [PMID: 24704206 DOI: 10.1016/j.bbagrm.2014.03.013] [Citation(s) in RCA: 483] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 03/18/2014] [Accepted: 03/23/2014] [Indexed: 12/12/2022]
Abstract
Soon after the discovery of the Myc gene (c-Myc), it became clear that Myc expression levels tightly correlate to cell proliferation. The entry in cell cycle of quiescent cells upon Myc enforced expression has been described in many models. Also, the downregulation or inactivation of Myc results in the impairment of cell cycle progression. Given the frequent deregulation of Myc oncogene in human cancer it is important to dissect out the mechanisms underlying the role of Myc on cell cycle control. Several parallel mechanisms account for Myc-mediated stimulation of the cell cycle. First, most of the critical positive cell cycle regulators are encoded by genes induced by Myc. These Myc target genes include Cdks, cyclins and E2F transcription factors. Apart from its direct effects on the transcription, Myc is able to hyperactivate cyclin/Cdk complexes through the induction of Cdk activating kinase (CAK) and Cdc25 phosphatases. Moreover, Myc antagonizes the activity of cell cycle inhibitors as p21 and p27 through different mechanisms. Thus, Myc is able to block p21 transcription or to induce Skp2, a protein involved in p27 degradation. Finally, Myc induces DNA replication by binding to replication origins and by upregulating genes encoding proteins required for replication initiation. Myc also regulates genes involved in the mitotic control. A promising approach to treat tumors with deregulated Myc is the synthetic lethality based on the inhibition of Cdks. Thus, the knowledge of the Myc-dependent cell cycle regulatory mechanisms will help to discover new therapeutic approaches directed against malignancies with deregulated Myc. This article is part of a Special Issue entitled: Myc proteins in cell biology and pathology.
Collapse
Affiliation(s)
- Gabriel Bretones
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-SODERCAN and Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - M Dolores Delgado
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-SODERCAN and Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain
| | - Javier León
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria-SODERCAN and Departamento de Biología Molecular, Universidad de Cantabria, Santander, Spain.
| |
Collapse
|
44
|
Dombrowski C, Helledie T, Ling L, Grünert M, Canning CA, Jones CM, Hui JH, Nurcombe V, van Wijnen AJ, Cool SM. FGFR1 Signaling Stimulates Proliferation of Human Mesenchymal Stem Cells by Inhibiting the Cyclin-Dependent Kinase Inhibitors p21Waf1and p27Kip1. Stem Cells 2013; 31:2724-36. [DOI: 10.1002/stem.1514] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 06/27/2013] [Accepted: 07/22/2013] [Indexed: 11/10/2022]
Affiliation(s)
- Christian Dombrowski
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - Torben Helledie
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - Ling Ling
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - Martin Grünert
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - Claire A. Canning
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - C. Michael Jones
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - James H. Hui
- Department of Orthopaedic Surgery; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| | - Victor Nurcombe
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
| | - Andre J. van Wijnen
- Department of Orthopaedic Surgery; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
- Department of Orthopedic Surgery; Mayo Clinic; Rochester Minnesota USA
| | - Simon M. Cool
- Institute of Medical Biology; Glycotherapeutics Group; 8A Biomedical Grove, #06-06 Immunos, A*STAR, Singapore Singapore
- Department of Orthopaedic Surgery; Yong Loo Lin School of Medicine, National University of Singapore; Singapore
| |
Collapse
|
45
|
Epstein-Barr virus latent membrane protein 2A enhances MYC-driven cell cycle progression in a mouse model of B lymphoma. Blood 2013; 123:530-40. [PMID: 24174629 DOI: 10.1182/blood-2013-07-517649] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Elevated expression of MYC is a shared property of many human cancers. Epstein-Barr virus (EBV) has been associated with lymphoid malignancies, yet collaborative roles between MYC and EBV in lymphomagenesis are unclear. EBV latent membrane protein 2A (LMP2A) functions as a B-cell receptor (BCR) mimic known to provide survival signals to infected B cells. Co-expression of human MYC and LMP2A in mice (LMP2A/λ-MYC) accelerates B lymphoma onset compared with mice expressing human MYC alone (λ-MYC mice). Here we show a novel role of LMP2A in potentiating MYC to promote G1-S transition and hyperproliferation by downregulating cyclin-dependent kinase inhibitor p27(kip1) in a proteasome-dependent manner. Expressing a gain-of-function S10A mutant of p27(kip1) has minor effect on tumor latency. However, pretumor B cells from λ-MYC mice expressing homozygous S10A mutant show a significant decrease in the percentage of S-phase cells. Interestingly, LMP2A is able to counteract the antiproliferative effect of the S10A mutant to promote S-phase entry. Finally, we show that LMP2A expression correlates with higher levels of MYC expression and suppression of p27(kip1) before lymphoma onset. Our study demonstrates a novel function of EBV LMP2A in maximizing MYC expression, resulting in hyperproliferation and cellular transformation into cancer cells in vivo.
Collapse
|
46
|
Wu YS, Chen YL, Yin SG, Wang HH. Role of transcription factors in regulation of gene expression in esophageal squamous cell carcinoma. Shijie Huaren Xiaohua Zazhi 2013; 21:1533-1538. [DOI: 10.11569/wcjd.v21.i16.1533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To study the role of transcription factors (TFs) in regulation of gene expression in esophageal squamous cell cancer.
METHODS: Two gene microarray data sets downloaded from the GEO BRB-ArrayTool were used to analyze differentially expressed genes between esophageal squamous cell cancer and normal esophageal tissue. TFs were enriched from differentially expressed genes, and the regulatory role of TFs was assessed by analyzing interactions among target genes, binding proteins and differentially expressed genes.
RESULTS: Eight TFs were obtained from 463 differentially expressed genes. There were 110 genes that may be regulated by TFs, including target genes, genes encoding interaction proteins, and upstream enzyme genes. These genes participated in multiple biological processes such as cell cycle, mitosis, cell division, cell migration, response to DNA damage, and DNA repair. Of these 110 genes, 37 were involved in KEGG pathways, including cancer pathways, focal adhesion, cell cycle, ECM-receptor interaction, small cell lung cancer, oocyte meiosis, p53 signaling pathway, and fatty acid metabolism. The pathways involving these 110 genes showed no significant difference with those involving the 463 differentially expressed genes. These 110 genes interact and form a molecular network which is composed of 110 nodes and 316 edges.
CONCLUSION: Eight TFs identify in this study constitute a complex molecular interaction network and play an important role in ESCC by regulating their target and interactive genes.
Collapse
|
47
|
Tezil T, Bodur C, Kutuk O, Basaga H. IKK-β mediates chemoresistance by sequestering FOXO3; a critical factor for cell survival and death. Cell Signal 2012; 24:1361-8. [PMID: 22313691 DOI: 10.1016/j.cellsig.2012.01.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 01/24/2012] [Accepted: 01/24/2012] [Indexed: 10/14/2022]
Abstract
Chemotherapeutic drugs proved only 50% successful in breast cancer because of cell type-dependent resistance mechanisms. FOXO3 is known to be involved in the regulation of several cell death-related genes; however, the extent of FOXO3 regulation in chemoresistance is still not fully understood. Here, we show that FOXO3 critically mediates cisplatin chemosensitivity of MCF-7 breast cancer cells which express higher levels of FOXO3 compared to resistant MDA-MB-231 cells. Administration of cisplatin induces apoptosis in MCF-7 cells in a FOXO3-dependent manner as indicated by RNA interference. On the other hand, IKK-β (IκB kinase) appears to inhibit FOXO3 action after cisplatin treatment and promotes chemoresistance in MDA-MB-231 cells. IKK-β directly interacts and sequesters FOXO3 in the cytosol preventing its nuclear localization. Moreover, cisplatin treatment induces autophagosome formation through LC-3 conversion while inhibiting the cleavage of caspase 9 and caspase 3 in MDA-MB-231 cells manipulated to overexpress FOXO3. In brief, our findings demonstrate that in addition to cellular level of active FOXO3, cisplatin chemoresistance is also regulated by IKK-β sequestration of FOXO3 in cytosol.
Collapse
Affiliation(s)
- Tugsan Tezil
- Sabanci University, Biological Sciences and Bioengineering Program, 34956, Tuzla, Istanbul, Turkey.
| | | | | | | |
Collapse
|
48
|
FoxO3A promotes metabolic adaptation to hypoxia by antagonizing Myc function. EMBO J 2012; 30:4554-70. [PMID: 21915097 DOI: 10.1038/emboj.2011.323] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Accepted: 08/15/2011] [Indexed: 12/23/2022] Open
Abstract
Exposure of metazoan organisms to hypoxia engages a metabolic switch orchestrated by the hypoxia-inducible factor 1 (HIF-1). HIF-1 mediates induction of glycolysis and active repression of mitochondrial respiration that reduces oxygen consumption and inhibits the production of potentially harmful reactive oxygen species (ROS). Here, we show that FoxO3A is activated in hypoxia downstream of HIF-1 and mediates the hypoxic repression of a set of nuclear-encoded mitochondrial genes. FoxO3A is required for hypoxic suppression of mitochondrial mass, oxygen consumption, and ROS production and promotes cell survival in hypoxia. FoxO3A is recruited to the promoters of nuclear-encoded mitochondrial genes where it directly antagonizes c-Myc function via a mechanism that does not require binding to the consensus FoxO recognition element. Furthermore, we show that FoxO3A is activated in human hypoxic tumour tissue in vivo and that FoxO3A short-hairpin RNA (shRNA)-expressing xenograft tumours are decreased in size and metabolically changed. Our findings define a novel mechanism by which FoxO3A promotes metabolic adaptation and stress resistance in hypoxia.
Collapse
|
49
|
Yan Y, Zhang X, Legerski RJ. Artemis interacts with the Cul4A-DDB1DDB2 ubiquitin E3 ligase and regulates degradation of the CDK inhibitor p27. Cell Cycle 2011; 10:4098-109. [PMID: 22134138 DOI: 10.4161/cc.10.23.18227] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Artemis, a member of the SNM1 gene family, is a multifunctional phospho-protein that has been shown to have important roles in V(D)J recombination, DNA double strand break repair, and stress-induced cell-cycle checkpoint regulation. We show here that Artemis interacts with the Cul4A-DDB1 E3 ubiquitin ligase via a direct interaction with the substrate-specificity receptor DDB2. Furthermore, Artemis also interacts with the CDK inhibitor and tumor suppressor p27, a substrate of the Cul4A-DDB1 ligase, and both DDB2 and Artemis are required for the degradation of p27 mediated by this complex. We also show that the regulation of p27 by Artemis and DDB2 is important for cell cycle progression in normally proliferating cells and in response to serum deprivation. These findings thus define a function for Artemis as an effector of Cullin-based E3 ligase-mediated ubiquitylation, demonstrate a novel pathway for the regulation of p27, and show that Cul4A-DDB1(DDB2-Artemis) regulates G1 phase cell cycle progression in mammalian cells.
Collapse
Affiliation(s)
- Yiyi Yan
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
50
|
Järvinen K, Hotti A, Santos L, Nummela P, Hölttä E. Caspase-8, c-FLIP, and caspase-9 in c-Myc-induced apoptosis of fibroblasts. Exp Cell Res 2011; 317:2602-15. [DOI: 10.1016/j.yexcr.2011.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/10/2011] [Accepted: 08/22/2011] [Indexed: 01/02/2023]
|