1
|
Lee CF, Pienta KJ, Amend SR. The involvement of cyclin-dependent kinase 7 (CDK7) and 9 (CDK9) in coordinating transcription and cell cycle checkpoint regulation. Cell Cycle 2025:1-13. [PMID: 40223539 DOI: 10.1080/15384101.2025.2485844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 09/24/2024] [Accepted: 10/08/2024] [Indexed: 04/15/2025] Open
Abstract
Cells regulate the expression of cell cycle-related genes, including cyclins essential for mitosis, through the transcriptional activity of the positive transcription elongation factor b (P-TEFb), a complex comprising CDK9, cyclin T, and transcription factors. P-TEFb cooperates with CDK7 to activate RNA polymerase. In response to DNA stress, the cell cycle shifts from mitosis to repair, triggering cell cycle arrest and the activation of DNA repair genes. This tight coordination between transcription, cell cycle progression, and DNA stress response is crucial for maintaining cellular integrity. Cyclin-dependent kinases CDK7 and CDK9 are central to both transcription and cell cycle regulation. CDK7 functions as the CDK-activating kinase (CAK), essential for activating other CDKs, while CDK9 acts as a critical integrator of signals from both the cell cycle and transcriptional machinery. This review elucidates the mechanisms by which CDK7 and CDK9 regulate the mitotic process and cell cycle checkpoints, emphasizing their roles in balancing cell growth, homeostasis, and DNA repair through transcriptional control.
Collapse
Affiliation(s)
- Cheng-Fan Lee
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - Kenneth J Pienta
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| | - Sarah R Amend
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, USA
| |
Collapse
|
2
|
Villoch‐Fernandez J, Martínez‐García N, Martín‐López M, Maeso‐Alonso L, López‐Ferreras L, Vazquez‐Jimenez A, Muñoz‐Hidalgo L, Garcia‐Romero N, Sanchez JM, Fernandez A, Ayuso‐Sacido A, Marques MM, Marin MC. A novel TAp73-inhibitory compound counteracts stemness features of glioblastoma stem cells. Mol Oncol 2025; 19:852-877. [PMID: 39090849 PMCID: PMC11887682 DOI: 10.1002/1878-0261.13694] [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: 12/11/2023] [Revised: 05/01/2024] [Accepted: 06/19/2024] [Indexed: 08/04/2024] Open
Abstract
Glioblastoma (GB) is the most common and fatal type of primary malignant brain tumor for which effective therapeutics are still lacking. GB stem cells, with tumor-initiating and self-renewal capacity, are mostly responsible for GB malignancy, representing a crucial target for therapies. The TP73 gene, which is highly expressed in GB, gives rise to the TAp73 isoform, a pleiotropic protein that regulates neural stem cell biology; however, its role in cancer has been highly controversial. We inactivated TP73 in human GB stem cells and revealed that TAp73 is required for their stemness potential, acting as a regulator of the transcriptional stemness signatures, highlighting TAp73 as a possible therapeutic target. As proof of concept, we identified a novel natural compound with TAp73-inhibitory capacity, which was highly effective against GB stem cells. The treatment reduced GB stem cell-invasion capacity and stem features, at least in part by TAp73 repression. Our data are consistent with a novel paradigm in which hijacking of p73-regulated neurodevelopmental programs, including neural stemness, might sustain tumor progression, pointing out TAp73 as a therapeutic strategy for GB.
Collapse
Affiliation(s)
| | | | | | - Laura Maeso‐Alonso
- Instituto de Biomedicina y Departamento de Biología MolecularUniversidad de LeónSpain
| | - Lorena López‐Ferreras
- Instituto de Biomedicina y Departamento de Biología MolecularUniversidad de LeónSpain
| | | | | | - Noemí Garcia‐Romero
- Faculty of Experimental SciencesUniversidad Francisco de VitoriaMadridSpain
- Brain Tumor Laboratory, Fundación VithasGrupo Hospitales VithasMadridSpain
- Faculty of MedicineUniversidad Francisco de VitoriaMadridSpain
| | | | | | - Angel Ayuso‐Sacido
- Faculty of Experimental SciencesUniversidad Francisco de VitoriaMadridSpain
- Brain Tumor Laboratory, Fundación VithasGrupo Hospitales VithasMadridSpain
- Faculty of MedicineUniversidad Francisco de VitoriaMadridSpain
| | - Margarita M. Marques
- Instituto de Desarrollo Ganadero y Sanidad Animal y Departamento de Producción AnimalUniversidad de LeónSpain
| | - Maria C. Marin
- Instituto de Biomedicina y Departamento de Biología MolecularUniversidad de LeónSpain
| |
Collapse
|
3
|
Thakur A, Rana N, Kumar R. Altered hormone expression induced genetic changes leads to breast cancer. Curr Opin Oncol 2024; 36:115-122. [PMID: 38441060 DOI: 10.1097/cco.0000000000001019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
PURPOSE OF REVIEW Breast cancer ranks first among gynecological cancer in India. It is associated with urbanization, changes in lifestyle and obesity. Hormones also play a crucial role in the development of breast cancer. Steroid hormones play critical role in development of breast cancer. RECENT FINDING Breast cancer is caused due to alteration in different hormone expressions leading to genetic instability. Loss or gains of functions due to genetic instability were associated with the alterations in housekeeping genes. Up-regulation in c-myc, signal transducer and activator of transcription (STAT), CREB-regulated transcription coactivator (CRTC), and eukaryotic translation initiation factor 4E (eIF4E) may cause the development of breast cancer. Peptide hormones are commonly following the phosphoinositide 3-kinases (PI3K) pathway for activation of cell cycle causing uncontrolled proliferation. Although steroid hormones are following the Ras/Raf/mitogen-activated protein kinase (MEK) pathway, their hyper-activation of these pathways causes extracellular-signal-regulated kinase (ERK) and MAPK activation, leading to carcinogenesis. SUMMARY Alteration in cell cycle proteins, oncogenes, tumor suppressor genes, transcription and translation factors lead to breast cancer. Apoptosis plays a vital role in the elimination of abnormal cells but failure in any of these apoptotic pathways may cause tumorigenesis. Hence, a complex interplay of hormonal and genetic factors is required to maintain homeostasis in breast cells. Imbalance in homeostasis of these hormone and genes may lead to breast cancer.
Collapse
Affiliation(s)
- Anchal Thakur
- Department of Animal sciences, Central University of Himachal Pradesh, Dharamshala, H.P
| | - Navya Rana
- Department of Animal sciences, Central University of Himachal Pradesh, Dharamshala, H.P
| | - Ranjit Kumar
- Department of Zoology, Nagaland University, Lumami, Nagaland
| |
Collapse
|
4
|
Hanalis-Miller T, Ricon-Becker I, Sakis N, Trachtenberg E, Ohayon F, Wadhawker S, Birnboim Y, Magen A, Sharon E, Tarrasch R, Goldzweig G, Cole SW, Jacoby R, Ben-Eliyahu S. Peri-operative individually tailored psychological intervention in breast cancer patients improves psychological indices and molecular biomarkers of metastasis in excised tumors. Brain Behav Immun 2024; 117:529-540. [PMID: 38346596 DOI: 10.1016/j.bbi.2024.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Perioperative stress and inflammatory signaling can invigorate pro-metastatic molecular processes in patients' tumors, potentially worsening long-term survival. Yet, it is unknown whether pre-operative psychotherapeutic interventions can attenuate such effects. Herein, three weeks before surgery, forty women diagnosed with stage I-III invasive ductal/lobular breast carcinoma were randomized to a 6-week one-on-one psychological intervention (6 meetings with a medical psychologist and bi-weekly phone calls) versus standard nursing-staff-attention. The intervention protocol was individually tailored based on evaluation of patients' emotional, cognitive, physiological, and behavioral stress response-patterns, and also included psychoeducation regarding medical treatments and recruitment of social support. Resected primary tumors were subjected to whole-genome RNA sequencing and bioinformatic analyses, assessing a priori hypothesized cancer-relevant molecular signatures. Self-report questionnaires (BSI-18, Hope-18, MSPSS, and a stress-scale) were collected three (T1) and one (T2) week before surgery, a day before (T3) and after (T4) surgery, and three weeks (T5) and 3-months (T6) following surgery. The intervention reduced distress (GSI), depression, and somatization scores (BSI-18: p < 0.01, p < 0.05, p < 0.05; T5 vs. T1). Additionally, tumors from treated patients (vs. controls) showed: (i) decreased activity of transcription control pathways involved in adrenergic and glucocorticoid signaling (CREB, GR) (p < 0.001), pro-inflammatory signaling (NFkB) (p < 0.01), and pro-malignant signaling (ETS1, STAT and GATA families) (p < 0.001, p < 0.01, p < 0.005); (ii) increased M1 macrophage polarization (p < 0.05), and CD4+ T cell activity (p < 0.01); and an unexpected increase in epithelial-to-mesenchymal-transition (EMT) signature (p < 0.005). This is the first randomized controlled trial to show beneficial effects of a psychological perioperative intervention on tumor pro-metastatic molecular biomarkers.
Collapse
Affiliation(s)
- Tsipi Hanalis-Miller
- Psychoneuroimmunology Laboratory, School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel; Stress, Hope and Cope Laboratory, School of Behavioral Sciences, The Academic College of Tel Aviv-Yafo, Tel Aviv, Israel
| | - Itay Ricon-Becker
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Nahida Sakis
- Psychoneuroimmunology Laboratory, School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Frida Ohayon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Sonya Wadhawker
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Yehudit Birnboim
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Ada Magen
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | - Eran Sharon
- Department of Surgery, Rabin Medical Center, Beilinson Hospital, Petah Tikva, Israel
| | | | - Gil Goldzweig
- Stress, Hope and Cope Laboratory, School of Behavioral Sciences, The Academic College of Tel Aviv-Yafo, Tel Aviv, Israel
| | - Steve W Cole
- Cousins Center for Psychoneuroimmunology, Semel Institute for Neuroscience and Human Behavior, Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Rebecca Jacoby
- Stress, Hope and Cope Laboratory, School of Behavioral Sciences, The Academic College of Tel Aviv-Yafo, Tel Aviv, Israel
| | - Shamgar Ben-Eliyahu
- Psychoneuroimmunology Laboratory, School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
Tomasso A, Koopmans T, Lijnzaad P, Bartscherer K, Seifert AW. An ERK-dependent molecular switch antagonizes fibrosis and promotes regeneration in spiny mice ( Acomys). SCIENCE ADVANCES 2023; 9:eadf2331. [PMID: 37126559 PMCID: PMC10132760 DOI: 10.1126/sciadv.adf2331] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Although most mammals heal injured tissues and organs with scarring, spiny mice (Acomys) naturally regenerate skin and complex musculoskeletal tissues. Now, the core signaling pathways driving mammalian tissue regeneration are poorly characterized. Here, we show that, while immediate extracellular signal-regulated kinase (ERK) activation is a shared feature of scarring (Mus) and regenerating (Acomys) injuries, ERK activity is only sustained at high levels during complex tissue regeneration. Following ERK inhibition, ear punch regeneration in Acomys shifted toward fibrotic repair. Using single-cell RNA sequencing, we identified ERK-responsive cell types. Loss- and gain-of-function experiments prompted us to uncover fibroblast growth factor and ErbB signaling as upstream ERK regulators of regeneration. The ectopic activation of ERK in scar-prone injuries induced a pro-regenerative response, including cell proliferation, extracellular matrix remodeling, and hair follicle neogenesis. Our data detail an important distinction in ERK activity between regenerating and poorly regenerating adult mammals and open avenues to redirect fibrotic repair toward regenerative healing.
Collapse
Affiliation(s)
- Antonio Tomasso
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster 48149, Germany
- Cells in Motion Cluster of Excellence-International Max Planck Research School (CiM-IMPRS Graduate Program), Münster 48149, Germany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506, USA
| | - Tim Koopmans
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
| | - Philip Lijnzaad
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, Utrecht 3584 CS, Netherlands
| | - Kerstin Bartscherer
- Max Planck Institute for Molecular Biomedicine, Röntgenstrasse 20, Münster 48149, Germany
- Cells in Motion Cluster of Excellence-International Max Planck Research School (CiM-IMPRS Graduate Program), Münster 48149, Germany
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Uppsalalaan 8, Utrecht 3584CT, Netherlands
- Department of Biology/Chemistry, Osnabrück University, Barbarastrasse 11, Osnabrück 49076, Germany
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, 101 T.H. Morgan Building, Lexington, KY 40506, USA
| |
Collapse
|
6
|
Øvrebø JI, Ma Y, Edgar BA. Cell growth and the cell cycle: New insights about persistent questions. Bioessays 2022; 44:e2200150. [PMID: 36222263 DOI: 10.1002/bies.202200150] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/08/2022]
Abstract
Before a cell divides into two daughter cells, it typically doubles not only its DNA, but also its mass. Numerous studies in cells ranging from yeast to mammals have shown that cellular growth, stimulated by nutrients and/or growth factor signaling, is a prerequisite for cell cycle progression in most types of cells. The textbook view of growth-regulated cell cycles is that growth signaling activates the transcription of G1 Cyclin genes to induce cell proliferation, and also stimulates anabolic metabolism and cell growth in parallel. However, genetic knockout tests in model organisms indicate that this is not the whole story, and new studies show that additional, "smarter" mechanisms help to coordinate the cell cycle with growth itself. Here we summarize recent advances in this field, and discuss current models in which growth signaling regulates cell proliferation by targeting core cell cycle regulators via non-transcriptional mechanisms.
Collapse
Affiliation(s)
- Jan Inge Øvrebø
- Computational Biology Unit, Department of Informatics, University of Bergen, Bergen, Norway
| | - Yiqin Ma
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| | - Bruce A Edgar
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
7
|
In vitro single molecule and bulk phase studies reveal the AP-1 transcription factor cFos binds to DNA without its partner cJun. J Biol Chem 2022; 298:102229. [PMID: 35787376 PMCID: PMC9364023 DOI: 10.1016/j.jbc.2022.102229] [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: 04/26/2022] [Revised: 06/27/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
The AP-1 transcription factor family crucially regulates progression of the cell cycle, as well as playing roles in proliferation, differentiation, and the stress response. The two best described AP-1 family members, cFos and cJun, are known to dimerize to form a functional AP-1 heterodimer that binds to a consensus response element sequence. Although cJun can also homodimerize and bind to DNA, the canonical view is that cFos cannot bind DNA without heterodimerizing with cJun. Here, we show that cFos can actually bind to DNA in the absence of cJun in vitro. Using dual color single molecule imaging of cFos alone, we directly visualize binding to and movement on DNA. Of all these DNA-bound proteins, detailed analysis suggested 30 to 46% were homodimers. Furthermore, we constructed fluorescent protein fusions of cFos and cJun for Förster resonance energy transfer experiments. These constructs indicated complete dimerization of cJun, but although cFos could dimerize, its extent was reduced. Finally, to provide orthogonal confirmation of cFos binding to DNA, we performed bulk-phase circular dichroism experiments that showed clear structural changes in DNA; these were found to be specific to the AP-1 consensus sequence. Taken together, our results clearly show cFos can interact with DNA both as monomers and dimers independently of its archetypal partner, cJun.
Collapse
|
8
|
Conversion of a Non-Cancer-Selective Promoter into a Cancer-Selective Promoter. Cancers (Basel) 2022; 14:cancers14061497. [PMID: 35326649 PMCID: PMC8946048 DOI: 10.3390/cancers14061497] [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: 01/08/2022] [Revised: 02/11/2022] [Accepted: 03/03/2022] [Indexed: 11/23/2022] Open
Abstract
Simple Summary The rat progression elevated gene-3 (PEG-3) promoter displays cancer-selective expression, whereas the rat growth arrest and DNA damage inducible gene-34 (GADD34) promoter lacks cancer specificity. PEG-3 and GADD34 minimal promoters display strong sequence homology except for two single point mutations. Since mutations are prevalent in many gene promoters resulting in significant alterations in promoter specificity and activity, we have explored the relevance of these two nucleotide alterations in determining cancer-selective gene expression. We demonstrate that these two point mutations are required to transform a non-cancer-specific promoter (pGADD) into a cancer-selective promoter (pGAPE). Additionally, we found GATA2 transcription factor binding sites in the GAPE-Prom, which regulates pGAPE activity selectively in cancer cells. This newly created pGAPE has all the necessary elements making it an appropriate genetic tool to noninvasively deliver imaging agents to follow tumor growth and progression to metastasis and for generating conditionally replicating adenoviruses that can express and deliver their payload exclusively in cancer. Abstract Progression-elevated gene-3 (PEG-3) and rat growth arrest and DNA damage-inducible gene-34 (GADD34) display significant sequence homology with regulation predominantly transcriptional. The rat full-length (FL) and minimal (min) PEG-3 promoter display cancer-selective expression in rodent and human tumors, allowing for cancer-directed regulation of transgenes, viral replication and in vivo imaging of tumors and metastases in animals, whereas the FL- and min-GADD34-Prom lack cancer specificity. Min-PEG-Prom and min-GADD34-Prom have identical sequences except for two single-point mutation differences (at −260 bp and +159 bp). Engineering double mutations in the min-GADD34-Prom produce the GAPE-Prom. Changing one base pair (+159) or both point mutations in the min-GADD34-Prom, but not the FL-GADD34-Prom, results in cancer-selective transgene expression in diverse cancer cells (including prostate, breast, pancreatic and neuroblastoma) vs. normal counterparts. Additionally, we identified a GATA2 transcription factor binding site, promoting cancer specificity when both min-PEG-Prom mutations are present in the GAPE-Prom. Taken together, introducing specific point mutations in a rat min-GADD34-Prom converts this non-cancer-specific promoter into a cancer-selective promoter, and the addition of GATA2 with existing AP1 and PEA3 transcription factors enhances further cancer-selective activity of the GAPE-Prom. The GAPE-Prom provides a genetic tool to specifically regulate transgene expression in cancer cells.
Collapse
|
9
|
Kumar A, Kothari J, Lokhande KB, Seethamma TN, Venkateswara Swamy K, Sharma NK. Novel Antiproliferative Tripeptides Inhibit AP-1 Transcriptional Complex. Int J Pept Res Ther 2021; 27:2163-2182. [DOI: 10.1007/s10989-021-10244-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/23/2021] [Indexed: 12/24/2022]
|
10
|
Chen H, Padia R, Li T, Li Y, Li B, Jin L, Huang S. Signaling of MK2 sustains robust AP1 activity for triple negative breast cancer tumorigenesis through direct phosphorylation of JAB1. NPJ Breast Cancer 2021; 7:91. [PMID: 34244488 PMCID: PMC8270897 DOI: 10.1038/s41523-021-00300-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 06/24/2021] [Indexed: 02/06/2023] Open
Abstract
Triple negative breast cancer (TNBC) cells are generally more invasive than estrogen receptor-positive (ER + ) breast cancer cells. Consistent with the importance of activator protein 1 (AP1) transcription factors in invasion, AP1 activity is much higher in TNBC lines than ER + lines. In TNBC cells, robust AP1 activity is facilitated by both ERK and p38MAPK signaling pathways. While ERK signaling pathway regulates AP1 activity by controlling the abundance of AP1 transcription factors, p38MAPK signaling pathway does it by enhancing AP1 binding to AP1 sites without altering their abundance. Here, we show that p38MAPK regulation of AP1 activity involves both MAPKAPK2 (MK2) and JAB1, a known JUN-binding protein. MK2 not only interacts with JAB1 but also directly phosphorylates JAB1 at Ser177 in TNBC cells. Interestingly, Ser177 phosphorylation does not affect JAB1 and JUN interaction. Instead, interfering with p38MAPK signaling pathway or introducing an S to A point mutation at Ser177 of JAB1 reduces JUN recruitment to the AP1 sites in cyclin D1, urokinase plasminogen activator (uPA) and uPA receptor promoters. Moreover, knockdown of JAB1 diminishes >60% of AP1 transcriptional activity in TNBC cells. Taken together, these results indicate that MK2-mediated phosphorylation of JAB1 facilitates JUN recruitment to AP1 sites, thus augmenting AP1 activity. In line with the role of JAB1 in AP1 activity, silencing JAB1 leads to dramatic reduction in TNBC cell growth, in vitro invasion and in vivo tumor outgrowth. This study suggests that the p38MAPK-MK2 signaling pathway promotes TNBC tumorigenesis by sustaining robust AP1 activity.
Collapse
Affiliation(s)
- Haoming Chen
- grid.8547.e0000 0001 0125 2443The Ministry of Education Key Laboratory of Contemporary Anthropology, College of Life Science, Fudan University, Shanghai, China
| | - Ravi Padia
- grid.15276.370000 0004 1936 8091Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL USA
| | - Tao Li
- grid.15276.370000 0004 1936 8091Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL USA
| | - Yue Li
- grid.15276.370000 0004 1936 8091Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL USA
| | - Bin Li
- grid.15276.370000 0004 1936 8091Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL USA
| | - Lingtao Jin
- grid.15276.370000 0004 1936 8091Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL USA
| | - Shuang Huang
- grid.15276.370000 0004 1936 8091Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL USA
| |
Collapse
|
11
|
Banfi F, Rubio A, Zaghi M, Massimino L, Fagnocchi G, Bellini E, Luoni M, Cancellieri C, Bagliani A, Di Resta C, Maffezzini C, Ianielli A, Ferrari M, Piazza R, Mologni L, Broccoli V, Sessa A. SETBP1 accumulation induces P53 inhibition and genotoxic stress in neural progenitors underlying neurodegeneration in Schinzel-Giedion syndrome. Nat Commun 2021; 12:4050. [PMID: 34193871 PMCID: PMC8245514 DOI: 10.1038/s41467-021-24391-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
The investigation of genetic forms of juvenile neurodegeneration could shed light on the causative mechanisms of neuronal loss. Schinzel-Giedion syndrome (SGS) is a fatal developmental syndrome caused by mutations in the SETBP1 gene, inducing the accumulation of its protein product. SGS features multi-organ involvement with severe intellectual and physical deficits due, at least in part, to early neurodegeneration. Here we introduce a human SGS model that displays disease-relevant phenotypes. We show that SGS neural progenitors exhibit aberrant proliferation, deregulation of oncogenes and suppressors, unresolved DNA damage, and resistance to apoptosis. Mechanistically, we demonstrate that high SETBP1 levels inhibit P53 function through the stabilization of SET, which in turn hinders P53 acetylation. We find that the inheritance of unresolved DNA damage in SGS neurons triggers the neurodegenerative process that can be alleviated either by PARP-1 inhibition or by NAD + supplementation. These results implicate that neuronal death in SGS originates from developmental alterations mainly in safeguarding cell identity and homeostasis.
Collapse
MESH Headings
- Abnormalities, Multiple/genetics
- Abnormalities, Multiple/metabolism
- Abnormalities, Multiple/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Cells, Cultured
- Craniofacial Abnormalities/genetics
- Craniofacial Abnormalities/metabolism
- Craniofacial Abnormalities/pathology
- DNA Damage
- Hand Deformities, Congenital/genetics
- Hand Deformities, Congenital/metabolism
- Hand Deformities, Congenital/pathology
- Heredodegenerative Disorders, Nervous System/genetics
- Heredodegenerative Disorders, Nervous System/metabolism
- Heredodegenerative Disorders, Nervous System/pathology
- Humans
- Intellectual Disability/genetics
- Intellectual Disability/metabolism
- Intellectual Disability/pathology
- Mutation
- Nails, Malformed/genetics
- Nails, Malformed/metabolism
- Nails, Malformed/pathology
- Neural Stem Cells/metabolism
- Neural Stem Cells/pathology
- Nuclear Proteins/genetics
- Nuclear Proteins/metabolism
- Organoids
- Tumor Suppressor Protein p53/antagonists & inhibitors
Collapse
Affiliation(s)
- Federica Banfi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | - Alicia Rubio
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | - Mattia Zaghi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Fagnocchi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Edoardo Bellini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Mirko Luoni
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cinzia Cancellieri
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Human Induced Pluripotent Stem Cells service, Istituto Italiano di Oncologia Molecolare (IFOM), Milan, Italy
| | - Anna Bagliani
- Medical Oncology Unit, ASST Ovest Milanese, Legnano Hospital, Legnano, Italy
| | - Chiara Di Resta
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Genomics for human disease diagnosis, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Camilla Maffezzini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelo Ianielli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | | | - Rocco Piazza
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Luca Mologni
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- CNR Institute of Neuroscience, Milan, Italy
| | - Alessandro Sessa
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
12
|
Memariani Z, Abbas SQ, Ul Hassan SS, Ahmadi A, Chabra A. Naringin and naringenin as anticancer agents and adjuvants in cancer combination therapy: Efficacy and molecular mechanisms of action, a comprehensive narrative review. Pharmacol Res 2020; 171:105264. [PMID: 33166734 DOI: 10.1016/j.phrs.2020.105264] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/10/2020] [Accepted: 10/19/2020] [Indexed: 12/24/2022]
Abstract
Although the rates of many cancers are controlled in Western countries, those of some cancers, such as lung, breast, and colorectal cancer are currently increasing in many low- and middle-income countries due to increases in risk factors caused by development and societal problems. Additionally, endogenous factors, such as inherited mutations, steroid hormones, insulin, and insulin-like growth factor systems, inflammation, oxidative stress, and exogenous factors (including tobacco, alcohol, infectious agents, and radiation), are believed to compromise cell functions and lead to carcinogenesis. Chemotherapy, surgery, radiation therapy, hormone therapy, and targeted therapies are some examples of the approaches used for cancer treatment. However, various short- and long-term side effects can also considerably impact patient prognosis based on clinical factors associated with treatments. Recently, increasing numbers of studies have been conducted to identify novel therapeutic agents from natural products, among which plant-derived bioactive compounds have been increasingly studied. Naringin (NG) and its aglycone naringenin (NGE) are abundantly present in citrus fruits, such as grapefruits and oranges. Their anti-carcinogenic activities have been shown to be exerted through several cell signal transduction pathways. Recently, different pharmacological strategies based on combination therapy, involving NG and NGE with the current anti-cancer agents have shown prodigious synergistic effects when compared to monotherapy. Besides, NG and NGE have been reported to overcome multidrug resistance, resulting from different defensive mechanisms in cancer, which is one of the major obstacles of clinical treatment. Thus, we comprehensively reviewed the inhibitory effects of NG and NGE on several types of cancers through different signal transduction pathways, the roles on sensitizing with the current anticancer medicines, and the efficacy of the cancer combination therapy.
Collapse
Affiliation(s)
- Zahra Memariani
- Traditional Medicine and History of Medical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Syed Qamar Abbas
- Department of Pharmacy, Sarhad University of Science and Technology, Peshawar, Pakistan.
| | - Syed Shams Ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, PR China.
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Aroona Chabra
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
13
|
Pennycook BR, Barr AR. Restriction point regulation at the crossroads between quiescence and cell proliferation. FEBS Lett 2020; 594:2046-2060. [PMID: 32564372 DOI: 10.1002/1873-3468.13867] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/11/2024]
Abstract
The coordination of cell proliferation with reversible cell cycle exit into quiescence is crucial for the development of multicellular organisms and for tissue homeostasis in the adult. The decision between quiescence and proliferation occurs at the restriction point, which is widely thought to be located in the G1 phase of the cell cycle, when cells integrate accumulated extracellular and intracellular signals to drive this binary cellular decision. On the molecular level, decision-making is exerted through the activation of cyclin-dependent kinases (CDKs). CDKs phosphorylate the retinoblastoma (Rb) transcriptional repressor to regulate the expression of cell cycle genes. Recently, the classical view of restriction point regulation has been challenged. Here, we review the latest findings on the activation of CDKs, Rb phosphorylation and the nature and position of the restriction point within the cell cycle.
Collapse
Affiliation(s)
- Betheney R Pennycook
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexis R Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| |
Collapse
|
14
|
Ali A, Al-Tobasei R, Lourenco D, Leeds T, Kenney B, Salem M. Genome-wide identification of loci associated with growth in rainbow trout. BMC Genomics 2020; 21:209. [PMID: 32138655 PMCID: PMC7059289 DOI: 10.1186/s12864-020-6617-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/24/2020] [Indexed: 12/22/2022] Open
Abstract
Background Growth is a major economic production trait in aquaculture. Improvements in growth performance will reduce time and cost for fish to reach market size. However, genes underlying growth have not been fully explored in rainbow trout. Results A previously developed 50 K gene-transcribed SNP chip, containing ~ 21 K SNPs showing allelic imbalances potentially associated with important aquaculture production traits including body weight, muscle yield, was used for genotyping a total of 789 fish with available phenotypic data for bodyweight gain. Genotyped fish were obtained from two consecutive generations produced in the NCCCWA growth-selection breeding program. Weighted single-step GBLUP (WssGBLUP) was used to perform a genome-wide association (GWA) analysis to identify quantitative trait loci (QTL) associated with bodyweight gain. Using genomic sliding windows of 50 adjacent SNPs, 247 SNPs associated with bodyweight gain were identified. SNP-harboring genes were involved in cell growth, cell proliferation, cell cycle, lipid metabolism, proteolytic activities, chromatin modification, and developmental processes. Chromosome 14 harbored the highest number of SNPs (n = 50). An SNP window explaining the highest additive genetic variance for bodyweight gain (~ 6.4%) included a nonsynonymous SNP in a gene encoding inositol polyphosphate 5-phosphatase OCRL-1. Additionally, based on a single-marker GWA analysis, 33 SNPs were identified in association with bodyweight gain. The highest SNP explaining variation in bodyweight gain was identified in a gene coding for thrombospondin-1 (THBS1) (R2 = 0.09). Conclusion The majority of SNP-harboring genes, including OCRL-1 and THBS1, were involved in developmental processes. Our results suggest that development-related genes are important determinants for growth and could be prioritized and used for genomic selection in breeding programs.
Collapse
Affiliation(s)
- Ali Ali
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Rafet Al-Tobasei
- Computational Science Program, Middle Tennessee State University, Murfreesboro, TN, 37132, USA
| | - Daniela Lourenco
- Department of Animal and Dairy Science, University of Georgia, Athens, GA, 30602, USA
| | - Tim Leeds
- United States Department of Agriculture Kearneysville, National Center for Cool and Cold Water Aquaculture, Agricultural Research Service, Kearneysville, WV, USA
| | - Brett Kenney
- Division of Animal and Nutritional Sciences, West Virginia University, Morgantown, WV, 26506, USA
| | - Mohamed Salem
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
15
|
Min DY, Jung E, Kim J, Lee YH, Shin SY. Leptin stimulates IGF-1 transcription by activating AP-1 in human breast cancer cells. BMB Rep 2020. [PMID: 30293548 PMCID: PMC6605521 DOI: 10.5483/bmbrep.2019.52.6.189] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leptin, an adipokine regulating energy metabolism, appears to be associated with breast cancer progression. Insulin-like growth factor-1 (IGF-1) mediates the pathogenesis of breast cancer. The regulation of IGF-1 expression by leptin in breast cancer cells is unclear. Here, we found that leptin upregulates IGF-1 expression at the transcriptional level in breast cancer cells. Activating protein-1 (AP-1)-binding element within the proximal region of IGF-1 was necessary for leptin-induced IGF-1 promoter activation. Forced expression of AP-1 components, c-FOS or c-JUN, enhanced leptin-induced IGF-1 expression, while knockdown of c-FOS or c-JUN abrogated leptin responsiveness. All three MAPKs (ERK1/2, JNK1/2, and p38 MAPK) mediated leptin-induced IGF-1 expression. These results suggest that leptin contributes to breast cancer progression through the transcriptional upregulation of leptin via the MAPK pathway. [BMB Reports 2019; 52(6): 385-390].
Collapse
Affiliation(s)
- Dong Yeong Min
- Department of Biological Sciences, Sanghuh College of Lifesciences, Koknkuk University, Seoul 05029, China
| | - Euitaek Jung
- Department of Biological Sciences, Sanghuh College of Lifesciences, Koknkuk University, Seoul 05029, China
| | - Juhwan Kim
- Department of Biological Sciences, Sanghuh College of Lifesciences, Koknkuk University, Seoul 05029, China
| | - Young Han Lee
- Department of Biological Sciences, Sanghuh College of Lifesciences, Koknkuk University, Seoul 05029; Cancer and Metabolism Institute, Konkuk University, Seoul 05029, Korea
| | - Soon Young Shin
- Department of Biological Sciences, Sanghuh College of Lifesciences, Koknkuk University, Seoul 05029; Cancer and Metabolism Institute, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
16
|
Wang X, Fan H, Xu C, Jiang G, Wang H, Zhang J. KDM3B suppresses APL progression by restricting chromatin accessibility and facilitating the ATRA-mediated degradation of PML/RARα. Cancer Cell Int 2019; 19:256. [PMID: 31592194 PMCID: PMC6778369 DOI: 10.1186/s12935-019-0979-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/26/2019] [Indexed: 12/01/2022] Open
Abstract
Background A hallmark of acute promyelocytic leukemia (APL) is the expression of PML/RARα fusion protein. Treatment with all-trans retinoic acid (ATRA) results in the terminal differentiation of neutrophil granulocytes. However, the underlying mechanisms remain largely unknown. Here, we identify and elucidate a novel differentiation-suppressive model of APL involving the histone demethylase KDM3B, which has been identified as a suppressor of the tumor genes involved in hematopoietic malignancies. Methods First, we established a KDM3B knockdown NB4 cell model to determine the functional characteristics of KDM3B by cell proliferation assay and flow cytometry. Then, we performed ChIP-seq and ATAC-seq to search for potential relationships among KDM3B, histone modification (H3K9me1/me2) and the chromatin state. Finally, molecular biological techniques and a multi-omics analysis were used to explore the role of KDM3B in differentiation of the leukemia cells after ATRA treatment. Results We found that knocking down KDM3B contributed to the growth of NB4 APL cells via the promotion of cell-cycle progression and blocked granulocytic differentiation. Through global and molecular approaches, we provided futher evidence that knocking down KDM3B altered the global distribution of H3K9me1/me2 and increased the chromatin accessibility. Moreover, knocking down KDM3B inhibited the ATRA-induced degradation of the PML/RARα oncoprotein. Conclusion Our study suggested that KDM3B was able to inhibit APL progression by maintaining chromatin in a compact state and facilitating the ATRA-mediated degradation of PML/RARα. Taken together, the results show that KDM3B may be an alternative target for the treatment regimens and the targeted therapy for APL by sustaining the function of PML/RARα fusion protein.
Collapse
Affiliation(s)
- Xinrui Wang
- 1State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Huiyong Fan
- 1State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Congling Xu
- 1State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Guojuan Jiang
- 1State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Haiwei Wang
- 2Institute of Health Sciences, Shanghai Institutes for Biological Sciences and Graduate School, Chinese Academy of Sciences, Shanghai, 200025 China
| | - Ji Zhang
- 1State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Rui-Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
17
|
Jallow F, O'Leary KA, Rugowski DE, Guerrero JF, Ponik SM, Schuler LA. Dynamic interactions between the extracellular matrix and estrogen activity in progression of ER+ breast cancer. Oncogene 2019; 38:6913-6925. [PMID: 31406251 PMCID: PMC6814534 DOI: 10.1038/s41388-019-0941-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 02/06/2023]
Abstract
Metastatic, anti-estrogen resistant estrogen receptor α positive (ER+) breast cancer is the leading cause of breast cancer deaths in U.S. women. While studies have demonstrated the importance of the stromal tumor microenvironment in cancer progression and therapeutic responses, effects on the responses of ER+ cancers to estrogen and anti-estrogens are poorly understood, particularly in the complex in vivo environment. In this study, we used an estrogen responsive syngeneic mouse model to interrogate how a COL1A1-enriched fibrotic ECM modulates integrated hormonal responses in cancer progression. We orthotopically transplanted the ER+ TC11 cell line into wild-type (WT) or collagen-dense (Col1a1tm1Jae/+, mCol1a1) syngeneic FVB/N female mice. Once tumors were established, recipients were supplemented with 17β-estradiol (E2), tamoxifen, or left untreated. Although the dense/stiff environment in mCol1a1 recipients did not alter the rate of E2-induced proliferation of the primary tumor, it fostered the agonist activity of tamoxifen to increase proliferation and AP-1 activity. Manipulation of estrogen activity did not alter the incidence of lung lesions in either WT or mCol1a1 hosts. However, the mCol1a1 environment enabled tamoxifen-stimulated growth of pulmonary metastases and further fueled estrogen-driven growth. Moreover, E2 remodeled peritumoral ECM architecture in WT animals, modifying alignment of collagen fibers and altering synthesis of ECM components associated with increased alignment and stiffness, and increasing FN1 and POSTN expression in the pulmonary metastatic niche. These studies demonstrate dynamic interactions between ECM properties and estrogen activity in progression of ER+ breast cancer, and support the need for therapeutics that target both ER and the tumor microenvironment.
Collapse
Affiliation(s)
- Fatou Jallow
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA.,Endocrinology-Reproductive Physiology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Kathleen A O'Leary
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Debra E Rugowski
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jorge F Guerrero
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Suzanne M Ponik
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Linda A Schuler
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA. .,University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
18
|
Chang C, Wang H, Liu J, Pan C, Zhang D, Li X, Pan Y. Porphyromonas gingivalis Infection Promoted the Proliferation of Oral Squamous Cell Carcinoma Cells through the miR-21/PDCD4/AP-1 Negative Signaling Pathway. ACS Infect Dis 2019; 5:1336-1347. [PMID: 31243990 DOI: 10.1021/acsinfecdis.9b00032] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Recent epidemiological studies have revealed that Porphyromonas gingivalis, a major pathogen in periodontal disease, is associated with the development of oral squamous cell carcinoma (OSCC). However, the underlying mechanisms induced by P. gingivalis have not been well-defined. We aimed to determine the role of P. gingivalis in OSCC proliferation and the relevant molecular mechanisms. A cellular proliferation model of OSCC Tca8113 cells infected by P. gingivalis at a multiplicity of infection (MOI) of 50 was established. Cell proliferation was drastically increased in the infected cells compared with the control cells, while the proportion of cells in S phase was increased and the proportion of cells in G1 phase was decreased in the infected cells compared with the control cells. Additionally, the levels of activator protein 1 (AP-1; c-Jun and c-Fos) and its target gene cyclin D1 were increased in P. gingivalis-infected Tca8113 cells compared with control cells. miR-21 expression was elevated when programmed cell death 4 (PDCD4) expression was downregulated. Cyclin D1 expression was regulated by miR-21, PDCD4, and AP-1. The disruption of the pathway by silencing c-Jun, blocking miR-21 expression, or overexpressing PDCD4 led to decreased cyclin D1 expression and inhibited cell proliferation. P. gingivalis DNA levels were positively correlated with miR-21 and c-Jun expression and negatively correlated with PDCD4 expression in clinical OSCC samples. Our findings indicated that P. gingivalis might promote OSCC proliferation by regulating cyclin D1 expression via the miR-21/PDCD4/AP-1 negative feedback signaling pathway.
Collapse
Affiliation(s)
- Chunrong Chang
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Hongyan Wang
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Junchao Liu
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Chunling Pan
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Dongmei Zhang
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Xin Li
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| | - Yaping Pan
- Department of Periodontics, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
- Department of Oral Biology, School of Stomatology, China Medical University, No. 117 Nanjing North Street, Shenyang, Liaoning 110002, China
| |
Collapse
|
19
|
Ou WB, Ni N, Zuo R, Zhuang W, Zhu M, Kyriazoglou A, Wu D, Eilers G, Demetri GD, Qiu H, Li B, Marino-Enriquez A, Fletcher JA. Cyclin D1 is a mediator of gastrointestinal stromal tumor KIT-independence. Oncogene 2019; 38:6615-6629. [PMID: 31371779 DOI: 10.1038/s41388-019-0894-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 02/22/2019] [Accepted: 04/03/2019] [Indexed: 12/19/2022]
Abstract
Oncogenic KIT or PDGFRA tyrosine kinase mutations are compelling therapeutic targets in most gastrointestinal stromal tumors (GISTs), and the KIT inhibitor, imatinib, is therefore standard of care for patients with metastatic GIST. However, some GISTs lose expression of KIT oncoproteins, and therefore become KIT-independent and are consequently resistant to KIT-inhibitor drugs. We identified distinctive biologic features in KIT-independent, imatinib-resistant GISTs as a step towards identifying drug targets in these poorly understood tumors. We developed isogenic GIST lines in which the parental forms were KIT oncoprotein-dependent, whereas sublines had loss of KIT oncoprotein expression, accompanied by markedly downregulated expression of the GIST biomarker, protein kinase C-theta (PRKCQ). Biologic mechanisms unique to KIT-independent GISTs were identified by transcriptome sequencing, qRT-PCR, immunoblotting, protein interaction studies, knockdown and expression assays, and dual-luciferase assays. Transcriptome sequencing showed that cyclin D1 expression was extremely low in two of three parental KIT-dependent GIST lines, whereas cyclin D1 expression was high in each of the KIT-independent GIST sublines. Cyclin D1 inhibition in KIT-independent GISTs had anti-proliferative and pro-apoptotic effects, associated with Rb activation and p27 upregulation. PRKCQ, but not KIT, was a negative regulator of cyclin D1 expression, whereas JUN and Hippo pathway effectors YAP and TAZ were positive regulators of cyclin D1 expression. PRKCQ, JUN, and the Hippo pathway coordinately regulate GIST cyclin D1 expression. These findings highlight the roles of PRKCQ, JUN, Hippo, and cyclin D1 as oncogenic mediators in GISTs that have converted, during TKI-therapy, to a KIT-independent state. Inhibitors of these pathways could be effective therapeutically for these now untreatable tumors.
Collapse
Affiliation(s)
- Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China. .,Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| | - Nan Ni
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Rui Zuo
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Weihao Zhuang
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Meijun Zhu
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Anastasios Kyriazoglou
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Duolin Wu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, Zhejiang, China
| | - Grant Eilers
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - George D Demetri
- Ludwig Center at Dana-Farber/Harvard Cancer Center and Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, 02115, USA
| | - Haibo Qiu
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Bin Li
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Division of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Adrian Marino-Enriquez
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
20
|
Moussa AY, Mostafa NM, Singab ANB. Pulchranin A: First report of isolation from an endophytic fungus and its inhibitory activity on cyclin dependent kinases. Nat Prod Res 2019; 34:2715-2722. [PMID: 30887847 DOI: 10.1080/14786419.2019.1585846] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Fungal factories emerge as a promising source for the production of bioactive natural products. This study reports the isolation and structure elucidation of pulchranin A, for the first time, from an endophytic fungus (Aspergillus TRL1), which was cultured from Tabebuia rosea (Bignoniaceae) stems and identified by DNA ITS sequencing. Pulchranin A showed promising in-vitro cytotoxic effects against breast (MCF-7), liver (Hep-G2) and colorectal (HCT) cell lines, with IC50 values of 63, 80 and 91 µg/mL, respectively. In addition, it inhibited three cyclin-dependent kinases (CDK1, CDK2 and CDK4) in MCF-7 cells with IC50 values of 9.82, 15.6 and 2.7 µg/mL, respectively. Results were further supported by in-silico molecular docking of pulchranin A to CDK1, CDK2, and CDK4 crystal structures, where it demonstrated good interactions by H-bonding, hydrophobic and Pi-Pi interactions with different amino acid residues of these enzymes. Pulchranin A might be a potential CDK inhibitor in human breast cancer cells.[Figure: see text].
Collapse
Affiliation(s)
- Ashaimaa Y Moussa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University , Cairo , Egypt
| | - Nada M Mostafa
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University , Cairo , Egypt
| | - Abdel Nasser B Singab
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University , Cairo , Egypt.,Center for Drug Discovery Research and Development, Ain Shams University , Cairo , Egypt
| |
Collapse
|
21
|
Xue Y, Meehan B, Fu Z, Wang XQD, Fiset PO, Rieker R, Levins C, Kong T, Zhu X, Morin G, Skerritt L, Herpel E, Venneti S, Martinez D, Judkins AR, Jung S, Camilleri-Broet S, Gonzalez AV, Guiot MC, Lockwood WW, Spicer JD, Agaimy A, Pastor WA, Dostie J, Rak J, Foulkes WD, Huang S. SMARCA4 loss is synthetic lethal with CDK4/6 inhibition in non-small cell lung cancer. Nat Commun 2019; 10:557. [PMID: 30718506 PMCID: PMC6362083 DOI: 10.1038/s41467-019-08380-1] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 01/03/2019] [Indexed: 12/23/2022] Open
Abstract
Tumor suppressor SMARCA4 (BRG1), a key SWI/SNF chromatin remodeling gene, is frequently inactivated in cancers and is not directly druggable. We recently uncovered that SMARCA4 loss in an ovarian cancer subtype causes cyclin D1 deficiency leading to susceptibility to CDK4/6 inhibition. Here, we show that this vulnerability is conserved in non-small cell lung cancer (NSCLC), where SMARCA4 loss also results in reduced cyclin D1 expression and selective sensitivity to CDK4/6 inhibitors. In addition, SMARCA2, another SWI/SNF subunit lost in a subset of NSCLCs, also regulates cyclin D1 and drug response when SMARCA4 is absent. Mechanistically, SMARCA4/2 loss reduces cyclin D1 expression by a combination of restricting CCND1 chromatin accessibility and suppressing c-Jun, a transcription activator of CCND1. Furthermore, SMARCA4 loss is synthetic lethal with CDK4/6 inhibition both in vitro and in vivo, suggesting that FDA-approved CDK4/6 inhibitors could be effective to treat this significant subgroup of NSCLCs.
Collapse
Affiliation(s)
- Yibo Xue
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Brian Meehan
- Department of Pediatrics, McGill University, and Research Institute of McGill University Health Centre, Montreal Children's Hospital, Montreal, QC, H4A 3J1, Canada
| | - Zheng Fu
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Xue Qing D Wang
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Pierre Olivier Fiset
- Department of Pathology, Glen Site, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Ralf Rieker
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, 91054, Erlangen, Germany
| | - Cameron Levins
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
| | - Tim Kong
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Xianbing Zhu
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Geneviève Morin
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Lashanda Skerritt
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Esther Herpel
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, and Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Sriram Venneti
- Pathology and Neuropathology, University of Michigan Medical School, Ann Arbor, MI, 48109-0605, USA
| | - Daniel Martinez
- Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, 19104, USA
| | - Alexander R Judkins
- Department of Pathology and Laboratory Medicine, Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Los Angeles, CA, 90027, USA
| | - Sungmi Jung
- Department of Pathology, Glen Site, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Sophie Camilleri-Broet
- Department of Pathology, Glen Site, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Anne V Gonzalez
- Division of Respiratory Medicine, Montreal Chest Institute, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - Marie-Christine Guiot
- Department of Pathology, Montreal Neurological Hospital/Institute, McGill University Health Centre, Montreal, QC, H4A 3J1, Canada
| | - William W Lockwood
- Department of Integrative Oncology, British Columbia Cancer Agency, Vancouver, BC, V5Z 1L3, Canada
- Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 2B5, Canada
| | - Jonathan D Spicer
- Department of Surgery, McGill University Health Center, Montreal, QC, H4A 3J1, Canada
| | - Abbas Agaimy
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital, 91054, Erlangen, Germany
| | - William A Pastor
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada
| | - Josée Dostie
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada
| | - Janusz Rak
- Department of Pediatrics, McGill University, and Research Institute of McGill University Health Centre, Montreal Children's Hospital, Montreal, QC, H4A 3J1, Canada
| | - William D Foulkes
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0C7, Canada
- Department of Medical Genetics, and Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, H3T 1E2, Canada
- Department of Medical Genetics and Cancer Research Program, Research Institute of the McGill University Health Centre, McGill University, Montreal, QC, H4A 3J1, Canada
| | - Sidong Huang
- Department of Biochemistry, McGill University, Montreal, QC, H3G 1Y6, Canada.
- The Rosalind & Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, H3A 1A3, Canada.
| |
Collapse
|
22
|
Luo L, Chi H, Ling J. MiR-124-3p suppresses glioma aggressiveness via targeting of Fra-2. Pathol Res Pract 2018; 214:1825-1834. [PMID: 30243808 DOI: 10.1016/j.prp.2018.09.017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/31/2018] [Accepted: 09/14/2018] [Indexed: 12/27/2022]
Abstract
Malignant glioma is the most common and deadly primary brain tumor in adults. However, the mechanisms underlying the malignancy of glioma remain unclear. In the present study, we found that Fos-related antigen-2 (Fra-2) was overexpressed in most glioma cells, and knockdown of Fra-2 prevented cell proliferation, migration, and invasion. Mechanistically, Fra-2 silencing led to a significant reduction in cell-cycle drivers (Cyclin D1 and Cyclin E1), one invasion-associated gene (MMP9), the mesenchymal marker (Vimentin), and induction of the epithelial marker (E-cadherin). Further study confirmed that miR-124-3p decreased the expression of Fra-2 via directly targeting the 3'-UTR, and transfection with miR-124-3p in glioma cells inhibited expression of the above cell-cycle and EMT promoters. Phenotypic experiments also showed that overexpression of Fra-2 weakened the inhibitory effects of miR-124-3p on the proliferation, migration, and invasion of glioma cells. In addition, Fra-2 knockdown impaired the malignant phenotypes enhanced by miR-124-3p inhibition, which suggested a crucial role for the miR-124-3p/Fra-2 pathway in glioma development. Consistently, high expression of Fra-2 was closely associated with low miR-124-3p level and indicated a poor prognosis in patients with glioma. In conclusion, this study indicates the existence of an aberrant miR-124-3p/Fra-2 pathway that results in glioma aggressiveness, which suggests novel therapeutic opportunities for this fatal disease.
Collapse
Affiliation(s)
- Lifei Luo
- Clinical Laboratory, Enze Hospital, Taizhou Enze Medical Center, Luqiao 318050, China
| | - Hongbo Chi
- Clinical Laboratory, Enze Hospital, Taizhou Enze Medical Center, Luqiao 318050, China
| | - Jie Ling
- Clinical Laboratory, Taizhou First People's Hospital, Huangyan Hospital of Wenzhou Medical University, Huangyan 318020, China.
| |
Collapse
|
23
|
Zhao S, Jiang Y, Zhao J, Li H, Yin X, Wang Y, Xie Y, Chen X, Lu J, Dong Z, Liu K. Quercetin‐3‐methyl ether inhibits esophageal carcinogenesis by targeting the AKT/mTOR/p70S6K and MAPK pathways. Mol Carcinog 2018; 57:1540-1552. [DOI: 10.1002/mc.22876] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/06/2018] [Accepted: 07/19/2018] [Indexed: 11/12/2022]
Affiliation(s)
- Simin Zhao
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Yanan Jiang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Jimin Zhao
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Honglin Li
- East China University of Science and TechnologyShanghaiChina
| | - Xueshan Yin
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
- The First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yanhong Wang
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Yifei Xie
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Xinhuan Chen
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Jing Lu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Ziming Dong
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
| | - Kangdong Liu
- Department of PathophysiologySchool of Basic Medical SciencesZhengzhou UniversityZhengzhouHenanChina
- Henan Provincial Cooperative Innovation Center for Cancer ChemopreventionZhengzhou UniversityZhengzhouHenanChina
- China‐US (Henan) Hormel Cancer InstituteZhengzhouHenanChina
| |
Collapse
|
24
|
Campbell MC, Pontiggia L, Russell AY, Schwarting R, Camacho J, Jasmin JF, Mercier I. CAPER as a therapeutic target for triple negative breast cancer. Oncotarget 2018; 9:30340-30354. [PMID: 30100993 PMCID: PMC6084388 DOI: 10.18632/oncotarget.25719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022] Open
Abstract
Breast cancers (BCas) that lack expression of the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) are referred to as triple negative breast cancers (TNBCs) and have the poorest clinical outcome. Once these aggressive tumors progress to distant organs, the median survival decreases to 12 months. With endocrine therapies being ineffective in this BCa subtype, highly toxic chemo- and radiation therapies are the only options. A better understanding of the functional role(s) of molecular targets contributing to TNBC progression could help in the design and development of new treatments that are more targeted with less toxicity. CAPER (Co-activator of AP-1 and ER) is a nuclear transcriptional co-activator that was recently involved in ER-positive BCa progression, however its role in hormone-independent cancers remains unknown. Our current report demonstrates that CAPER expression is upregulated in human TNBC specimens compared to normal breast tissue and that its selective downregulation through a lentiviral-mediated shRNA knockdown approach resulted in decreased cell numbers in MDA-MB-231 and BT549 TNBC cell lines without affecting the growth of non-tumorigenic cell line MCF-10A. Concordant with these observations, CAPER knockdown was also associated with a decrease in DNA repair proteins leading to a marked increase in apoptosis, through caspase-3/7 activation without any changes in cell cycle. Collectively, we propose CAPER as an important signaling molecule in the development of TNBC linked to DNA repair mechanisms, which could lead to new therapeutic modalities for the treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Mallory C Campbell
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA
| | - Laura Pontiggia
- Department of Mathematics, Physics and Statistics, Misher College of Arts and Sciences, University of the Sciences, Philadelphia, PA 19104, USA
| | - Ashley Y Russell
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA
| | - Roland Schwarting
- Department of Pathology, Cooper University Hospital, Camden, NJ 08103, USA
| | - Jeanette Camacho
- Department of Pathology, Cooper University Hospital, Camden, NJ 08103, USA
| | - Jean-Francois Jasmin
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA
| | - Isabelle Mercier
- Department of Pharmaceutical Sciences, Philadelphia College of Pharmacy, University of the Sciences, Philadelphia, PA 19104, USA.,Program in Personalized Medicine and Targeted Therapeutics, University of the Sciences, Philadelphia, PA 19104, USA
| |
Collapse
|
25
|
Qiu Z, Zhou J, Zhang C, Cheng Y, Hu J, Zheng G. Antiproliferative effect of urolithin A, the ellagic acid-derived colonic metabolite, on hepatocellular carcinoma HepG2.2.15 cells by targeting Lin28a/let-7a axis. ACTA ACUST UNITED AC 2018; 51:e7220. [PMID: 29742265 PMCID: PMC5972012 DOI: 10.1590/1414-431x20187220] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
Abstract
An abnormality in the Lin28/let-7a axis is relevant to the progression of
hepatitis B virus (HBV)-positive hepatocellular carcinoma (HCC), which could be
a novel therapeutic target for this malignant tumor. The present study aimed to
investigate the antiproliferative and anti-invasive effects of urolithin A in a
stable full-length HBV gene integrated cell line HepG2.2.15 using CCK-8 and
transwell assays. The RNA and protein expressions of targets were assessed by
quantitative PCR and western blot, respectively. Results revealed that urolithin
A induced cytotoxicity in HepG2.2.15 cells, which was accompanied by the
cleavage of caspase-3 protein and down-regulation of Bcl-2/Bax ratio. Moreover,
urolithin A suppressed the protein expressions of Sp-1, Lin28a, and Zcchc11, and
elevated the expression of microRNA let-7a. Importantly, urolithin A also
regulated the Lin28a/let-7a axis in transient HBx-transfected HCC HepG2 cells.
Furthermore, urolithin A decelerated the HepG2.2.15 cell invasion, which was
involved in suppressing the let-7a downstream factors HMGA2 and K-ras. These
findings indicated that urolithin A exerted the antiproliferative effect by
regulating the Lin28a/let-7a axis and may be a potential supplement for
HBV-infected HCC therapy.
Collapse
Affiliation(s)
- Zhenpeng Qiu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Junxuan Zhou
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Cong Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Ye Cheng
- Hubei Engineering Research Center of Viral Vector, Wuhan Institute of Bioengineering, Wuhan, China
| | - Junjie Hu
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China
| |
Collapse
|
26
|
The Role of Activator Protein-1 (AP-1) Family Members in CD30-Positive Lymphomas. Cancers (Basel) 2018; 10:cancers10040093. [PMID: 29597249 PMCID: PMC5923348 DOI: 10.3390/cancers10040093] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/21/2018] [Accepted: 03/25/2018] [Indexed: 12/14/2022] Open
Abstract
The Activator Protein-1 (AP-1) transcription factor (TF) family, composed of a variety of members including c-JUN, c-FOS and ATF, is involved in mediating many biological processes such as proliferation, differentiation and cell death. Since their discovery, the role of AP-1 TFs in cancer development has been extensively analysed. Multiple in vitro and in vivo studies have highlighted the complexity of these TFs, mainly due to their cell-type specific homo- or hetero-dimerization resulting in diverse transcriptional response profiles. However, as a result of the increasing knowledge of the role of AP-1 TFs in disease, these TFs are being recognized as promising therapeutic targets for various malignancies. In this review, we focus on the impact of deregulated expression of AP-1 TFs in CD30-positive lymphomas including Classical Hodgkin Lymphoma and Anaplastic Large Cell Lymphoma.
Collapse
|
27
|
Ouimet M, Drouin S, Lajoie M, Caron M, St-Onge P, Gioia R, Richer C, Sinnett D. A childhood acute lymphoblastic leukemia-specific lncRNA implicated in prednisolone resistance, cell proliferation, and migration. Oncotarget 2018; 8:7477-7488. [PMID: 27980230 PMCID: PMC5352336 DOI: 10.18632/oncotarget.13936] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 12/02/2016] [Indexed: 11/25/2022] Open
Abstract
Childhood acute lymphoblastic leukemia (cALL) is the most common pediatric cancer and, despite an 85% cure rate, still represents a major cause of disease-related death in children. Recent studies have implicated long non-coding RNAs (lncRNAs) in cALL etiology, progression, and treatment response. However, barring some exceptions little is known about the functional impact of lncRNAs on cancer biology, which limits their potential as potential therapeutic targets. We wanted to investigate the functional role of lncRNAs identified as specifically overexpressed in pre-B cALL by whole-transcriptome sequencing. Here we report five lncRNAs specifically upregulated in pre-B cALL that had significant impacts on cancer hallmark traits such as cell proliferation, migration, apoptosis, and treatment response. In particular, silencing of the RP11-137H2.4 lncRNA effectively restored normal glucocorticoid (GC) response in a GC-resistant pre-B cALL cell line and specifically modulated expression of members of both the NRAS/BRAF/NF-?B MAPK cascade and cell cycle pathways. Since GC form the cornerstone of cALL chemotherapy and resistance in cALL confers a dismal prognosis, characterizing RP11-137H2.4sexact role and function in this process will be critical to the development of new therapeutic approaches to overcome GC resistance in children treated for cALL.
Collapse
Affiliation(s)
- Manon Ouimet
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Simon Drouin
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Mathieu Lajoie
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Maxime Caron
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Pascal St-Onge
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Romain Gioia
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Chantal Richer
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada
| | - Daniel Sinnett
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, Montreal, QC, Canada.,Department of Pediatrics, University of Montreal, Montreal, QC, Canada
| |
Collapse
|
28
|
Tewari D, Nabavi SF, Nabavi SM, Sureda A, Farooqi AA, Atanasov AG, Vacca RA, Sethi G, Bishayee A. Targeting activator protein 1 signaling pathway by bioactive natural agents: Possible therapeutic strategy for cancer prevention and intervention. Pharmacol Res 2017; 128:366-375. [PMID: 28951297 DOI: 10.1016/j.phrs.2017.09.014] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/13/2017] [Accepted: 09/22/2017] [Indexed: 12/29/2022]
Abstract
Activator protein 1 (AP-1) is a key transcription factor in the control of several cellular processes responsible for cell survival proliferation and differentiation. Dysfunctional AP-1 expression and activity are involved in several severe diseases, especially inflammatory disorders and cancer. Therefore, targeting AP-1 has recently emerged as an attractive therapeutic strategy for cancer prevention and therapy. This review summarizes our current understanding of AP-1 biology and function as well as explores and discusses several natural bioactive compounds modulating AP-1-associated signaling pathways for cancer prevention and intervention. Current limitations, challenges, and future directions of research are also critically discussed.
Collapse
Affiliation(s)
- Devesh Tewari
- Department of Pharmaceutical Sciences, Faculty of Technology, Bhimtal Campus, Kumaun University, Nainital, 263 136, Uttarakhand, India
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, 1435916471, Iran
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, 1435916471, Iran.
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress and CIBEROBN Physiopathology of Obesity and Nutrition, University of Balearic Islands, E-07122, Palma de Mallorca, Balearic Islands, Spain
| | - Ammad Ahmad Farooqi
- Laboratory for Translational Oncology and Personalized Medicine, Rashid Latif Medical College, Lahore, 54000, Pakistan
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, 05-552, Jastrzebiec, Poland; Department of Pharmacognosy, University of Vienna, 1090, Vienna, Austria; Department of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, 1090, Vienna, Austria
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Council of Research, I-70126, Bari, Italy
| | - Gautam Sethi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Anupam Bishayee
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, 18301 N. Miami Avenue, Miami, FL, 33169, USA.
| |
Collapse
|
29
|
Satterfield L, Shuck R, Kurenbekova L, Allen-Rhoades W, Edwards D, Huang S, Rajapakshe K, Coarfa C, Donehower LA, Yustein JT. miR-130b directly targets ARHGAP1 to drive activation of a metastatic CDC42-PAK1-AP1 positive feedback loop in Ewing sarcoma. Int J Cancer 2017; 141:2062-2075. [PMID: 28748534 DOI: 10.1002/ijc.30909] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/30/2017] [Accepted: 07/17/2017] [Indexed: 01/08/2023]
Abstract
Ewing Sarcoma (ES) is a highly aggressive bone tumor with peak incidence in the adolescent population. It has a high propensity to metastasize, which is associated with dismal survival rates of approximately 25%. To further understand mechanisms of metastasis we investigated microRNA regulatory networks in ES. Our studies focused on miR-130b due to our analysis that enhanced expression of this microRNA has clinical relevance in multiple sarcomas, including ES. Our studies provide insights into a novel positive feedback network involving the direct regulation of miR-130b and activation of downstream signaling events contributing toward sarcoma metastasis. Specifically, we demonstrated miR-130b induces proliferation, invasion, and migration in vitro and increased metastatic potential in vivo. Using microarray analysis of ES cells with differential miR-130b expression we identified alterations in downstream signaling cascades including activation of the CDC42 pathway. We identified ARHGAP1, which is a negative regulator of CDC42, as a novel, direct target of miR-130b. In turn, downstream activation of PAK1 activated the JNK and AP-1 cascades and downstream transcriptional targets including IL-8, MMP1 and CCND1. Furthermore, chromatin immunoprecipitation of endogenous AP-1 in ES cells demonstrated direct binding to an upstream consensus binding site within the miR-130b promoter. Finally, small molecule inhibition of PAK1 blocked miR-130b activation of JNK and downstream AP-1 target genes, including primary miR-130b transcripts, and miR-130b oncogenic properties, thus identifying PAK1 as a novel therapeutic target for ES. Taken together, our findings identify and characterize a novel, targetable miR-130b regulatory network that promotes ES metastasis.
Collapse
Affiliation(s)
- Laura Satterfield
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX.,Integrative Molecular and Biological Sciences Program, Baylor College of Medicine, Houston, TX
| | - Ryan Shuck
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX
| | - Lyazat Kurenbekova
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX
| | - Wendy Allen-Rhoades
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX
| | - Dean Edwards
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Shixia Huang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Kimal Rajapakshe
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Cristian Coarfa
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Lawrence A Donehower
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX.,Integrative Molecular and Biological Sciences Program, Baylor College of Medicine, Houston, TX.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX.,Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX
| | - Jason T Yustein
- Texas Children's Cancer and Hematology Centers and The Faris D. Virani Ewing Sarcoma Center, Baylor College of Medicine, Houston, TX.,Integrative Molecular and Biological Sciences Program, Baylor College of Medicine, Houston, TX.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
30
|
Phelps CA, Lai SC, Mu D. Roles of Thyroid Transcription Factor 1 in Lung Cancer Biology. VITAMINS AND HORMONES 2017; 106:517-544. [PMID: 29407447 PMCID: PMC11528467 DOI: 10.1016/bs.vh.2017.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Thyroid transcription factor 1 (TTF-1 or NKX2-1) is a transcription factor of fundamental importance in driving lung maturation and morphogenesis. In the last decade, scientists began to appreciate the functional roles of TTF-1 in lung tumorigenesis. This movement was triggered by the discoveries of genetic alterations of TTF-1 in the form of gene amplification in lung cancer. Many downstream target genes of TTF-1 relevant to the lung cancer biology of TTF-1 have been documented. One of the most surprising findings was that TTF-1 may exhibit either pro- or antitumorigenic activities, an outcome with the complexity exceeding the original anticipation purely based on the fact that TTF-1 undergoes gene amplification in lung cancer. In the coming decade, we believe, we will witness additional surprises as the research exploring the cancer roles of TTF-1 progresses.
Collapse
Affiliation(s)
- Cody A Phelps
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Shao-Chiang Lai
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States
| | - David Mu
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|
31
|
Watanabe M, Iizumi Y, Sukeno M, Iizuka-Ohashi M, Sowa Y, Sakai T. The pleiotropic regulation of cyclin D1 by newly identified sesaminol-binding protein ANT2. Oncogenesis 2017; 6:e311. [PMID: 28368390 PMCID: PMC5520487 DOI: 10.1038/oncsis.2017.10] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 02/03/2017] [Accepted: 02/12/2017] [Indexed: 12/14/2022] Open
Abstract
The expression of cyclin D1 is upregulated in various cancer cells by diverse mechanisms, such as increases in mRNA levels, the promotion of the translation by mammalian target of rapamycin complex 1 (mTORC1) signaling and the protein stabilization. We here show that sesaminol, a sesame lignan, reduces the expression of cyclin D1 with decreasing mRNA expression levels, inhibiting mTORC1 signaling and promoting proteasomal degradation. We subsequently generated sesaminol-immobilized FG beads to newly identify sesaminol-binding proteins. As a consequence, we found that adenine nucleotide translocase 2 (ANT2), the inner mitochondrial membrane protein, directly bound to sesaminol. Consistent with the effects of sesaminol, the depletion of ANT2 caused a reduction in cyclin D1 with decreases in its mRNA levels, mTORC1 inhibition and the proteasomal degradation of its protein, suggesting that sesaminol negatively regulates the function of ANT2. Furthermore, we screened other ANT2-binding compounds and found that the proliferator-activated receptor-γ agonist troglitazone also reduced cyclin D1 expression in a multifaceted manner, analogous to that of the sesaminol treatment and ANT2 depletion. Therefore, the chemical biology approach using magnetic FG beads employed in the present study revealed that sesaminol bound to ANT2, which may pleiotropically upregulate cyclin D1 expression at the mRNA level and protein level with mTORC1 activation and protein stabilization. These results suggest the potential of ANT2 as a target against cyclin D1-overexpressing cancers.
Collapse
Affiliation(s)
- M Watanabe
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Y Iizumi
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - M Sukeno
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - M Iizuka-Ohashi
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Division of Endocrine and Breast Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Y Sowa
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - T Sakai
- Department of Molecular-Targeting Cancer Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
32
|
Chloroquine inhibits human CD4 + T-cell activation by AP-1 signaling modulation. Sci Rep 2017; 7:42191. [PMID: 28169350 PMCID: PMC5294581 DOI: 10.1038/srep42191] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/06/2017] [Indexed: 12/17/2022] Open
Abstract
Chloroquine (CQ) is widely used as an anti-inflammatory therapeutic for rheumatic diseases. Although its modes of action on the innate immune system are well described, there is still insufficient knowledge about its direct effects on the adaptive immune system. Thus, we evaluated the influence of CQ on activation parameters of human CD4+ T-cells. CQ directly suppressed proliferation, metabolic activity and cytokine secretion of T-cells following anti-CD3/anti-CD28 activation. In contrast, CQ showed no effect on up-regulation of T-cell activation markers. CQ inhibited activation of all T helper cell subsets, although IL-4 and IL-13 secretion by Th2 cells were less influenced compared to other Th-specific cytokines. Up to 10 μM, CQ did not reduce cell viability, suggesting specific suppressive effects on T-cells. These properties of CQ were fully reversible in re-stimulation experiments. Analyses of intracellular signaling showed that CQ specifically inhibited autophagic flux and additionally activation of AP-1 by reducing phosphorylation of c-JUN. This effect was mediated by inhibition of JNK catalytic activity. In summary, we characterized selective and reversible immunomodulatory effects of CQ on human CD4+ T-cells. These findings provide new insights into the biological actions of JNK/AP-1 signaling in T-cells and may help to expand the therapeutic spectrum of CQ.
Collapse
|
33
|
Papaioannou A, Chevet E. Driving Cancer Tumorigenesis and Metastasis Through UPR Signaling. Curr Top Microbiol Immunol 2017; 414:159-192. [PMID: 28710693 DOI: 10.1007/82_2017_36] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the tumor microenvironment, cancer cells encounter both external and internal factors that can lead to the accumulation of improperly folded proteins in the Endoplasmic Reticulum (ER) lumen, thus causing ER stress. When this happens, an adaptive mechanism named the Unfolded Protein Response (UPR) is triggered to help the cell cope with this change and restore protein homeostasis in the ER. Sequentially, one would expect that the activation of the three UPR branches, driven namely by IRE1, PERK, and ATF6, are crucial for the adaptation of cancer cells to the changing environment and thus for their survival and further propagation. Indeed, in the last few years, an increasing amount of studies has shown the implication of UPR signaling in different aspects of carcinogenesis and tumor progression. Features such as sustaining proliferation and resistance to cell death, genomic instability, altered metabolism, increased inflammation and tumor-immune infiltration, invasion and metastasis, and angiogenesis, defined as "the hallmarks of cancer", can be regulated by the UPR machinery. At the same time, new potential therapeutic interventions applicable to different kinds of cancers are being revealed. In order to describe the emerging role of UPR in cancer biology, these are the points that will be discussed in this chapter.
Collapse
Affiliation(s)
- Alexandra Papaioannou
- Inserm U1242 «Chemistry, Oncogenesis, Stress and Signaling», University of Rennes 1, Rennes, France.,Centre de Lutte contre le Cancer Eugène Marquis, Avenue de la bataille Flandres Dunkerque, 35000, Rennes, France
| | - Eric Chevet
- Inserm U1242 «Chemistry, Oncogenesis, Stress and Signaling», University of Rennes 1, Rennes, France.
| |
Collapse
|
34
|
Li X, Liang Q, Liu W, Zhang N, Xu L, Zhang X, Zhang J, Sung JJY, Yu J. Ras association domain family member 10 suppresses gastric cancer growth by cooperating with GSTP1 to regulate JNK/c-Jun/AP-1 pathway. Oncogene 2016; 35:2453-2464. [PMID: 26279301 DOI: 10.1038/onc.2015.300] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 06/16/2015] [Accepted: 06/28/2015] [Indexed: 11/08/2022]
Abstract
The Ras association domain family (RASSF) encodes several members with tumor-suppressive potentials. We aimed to investigate the biological function and clinical implication of RASSF10 in gastric cancer (GC). We found that RASSF10 was silenced in six of seven GC cell lines and in primary GC tissues, but was highly expressed in normal gastric tissues. The silence of RASSAF10 was mediated by promoter methylation as evaluated by bisulfite genomic sequencing. RASSF10 expression could be restored by demethylation treatment. A negative correlation between methylation and mRNA expression of RASSF10 was observed in 223 gastric samples of The Cancer Genome Atlas study (P<0.0001). Re-expression of RASSF10 in GC cell lines (AGS and MKN45) significantly suppressed cell viability, colony formation, migration and invasion, reduced cells in S phase, accumulated cells in G2 phase and induced cell apoptosis in vitro, and inhibited tumorigenicity in nude mice. These were confirmed by decreased expression of proliferation markers (proliferating cell nuclear antigen, p-CDC2 and p-CDC25) and increased apoptotic cascades (cleaved caspases-9, -8, -3 and cleaved poly (ADP-ribose) polymerase). Conversely, RASSF10 knockdown in normal gastric cell line yielded an opposing effect. Co-immunoprecipitation combined with mass spectrometry analyses were performed to reveal the downstream effectors of RASSF10. The result revealed that glutathione S-transferase Pi 1 (GSTP1) was a direct cooperator of RASSF10. The tumor-suppressive effect of RASSF10 was partially mediated by cooperating with GSTP1 to deregulate Jun N-terminal kinase (JNK)/c-Jun/AP-1 pathway. Importantly, RASSF10 methylation was detected in 56.6% (98/173) of primary GCs and is an independent risk factor for poor survival of GC patients (P=0.001). In conclusions, RASSF10 functions as a tumor suppressor by cooperating with GSTP1 to deregulate JNK/c-Jun/AP-1 pathway in GC. Promoter methylation of RASSF10 is associated with poor survival of GC patients.
Collapse
Affiliation(s)
- X Li
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Q Liang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - W Liu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - N Zhang
- Department of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - L Xu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
- Department of Gastroenterology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - X Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - J Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - J J Y Sung
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - J Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
35
|
Meckbach C, Tacke R, Hua X, Waack S, Wingender E, Gültas M. PC-TraFF: identification of potentially collaborating transcription factors using pointwise mutual information. BMC Bioinformatics 2015; 16:400. [PMID: 26627005 PMCID: PMC4667426 DOI: 10.1186/s12859-015-0827-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 11/17/2015] [Indexed: 01/06/2023] Open
Abstract
Background Transcription factors (TFs) are important regulatory proteins that govern transcriptional regulation. Today, it is known that in higher organisms different TFs have to cooperate rather than acting individually in order to control complex genetic programs. The identification of these interactions is an important challenge for understanding the molecular mechanisms of regulating biological processes. In this study, we present a new method based on pointwise mutual information, PC-TraFF, which considers the genome as a document, the sequences as sentences, and TF binding sites (TFBSs) as words to identify interacting TFs in a set of sequences. Results To demonstrate the effectiveness of PC-TraFF, we performed a genome-wide analysis and a breast cancer-associated sequence set analysis for protein coding and miRNA genes. Our results show that in any of these sequence sets, PC-TraFF is able to identify important interacting TF pairs, for most of which we found support by previously published experimental results. Further, we made a pairwise comparison between PC-TraFF and three conventional methods. The outcome of this comparison study strongly suggests that all these methods focus on different important aspects of interaction between TFs and thus the pairwise overlap between any of them is only marginal. Conclusions In this study, adopting the idea from the field of linguistics in the field of bioinformatics, we develop a new information theoretic method, PC-TraFF, for the identification of potentially collaborating transcription factors based on the idiosyncrasy of their binding site distributions on the genome. The results of our study show that PC-TraFF can succesfully identify known interacting TF pairs and thus its currently biologically uncorfirmed predictions could provide new hypotheses for further experimental validation. Additionally, the comparison of the results of PC-TraFF with the results of previous methods demonstrates that different methods with their specific scopes can perfectly supplement each other. Overall, our analyses indicate that PC-TraFF is a time-efficient method where its algorithm has a tractable computational time and memory consumption. The PC-TraFF server is freely accessible at http://pctraff.bioinf.med.uni-goettingen.de/ Electronic supplementary material The online version of this article (doi:10.1186/s12859-015-0827-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Cornelia Meckbach
- Institute of Bioinformatics, University of Göttingen, Goldschmidtstr. 1, Göttingen, 37077, Germany.
| | - Rebecca Tacke
- Institute of Bioinformatics, University of Göttingen, Goldschmidtstr. 1, Göttingen, 37077, Germany.
| | - Xu Hua
- Institute of Bioinformatics, University of Göttingen, Goldschmidtstr. 1, Göttingen, 37077, Germany.
| | - Stephan Waack
- Institute of Computer Science, University of Göttingen, Goldschmidtstr. 7, Göttingen, 37077, Germany.
| | - Edgar Wingender
- Institute of Bioinformatics, University of Göttingen, Goldschmidtstr. 1, Göttingen, 37077, Germany.
| | - Mehmet Gültas
- Institute of Bioinformatics, University of Göttingen, Goldschmidtstr. 1, Göttingen, 37077, Germany.
| |
Collapse
|
36
|
Sundar R, Gudey SK, Heldin CH, Landström M. TRAF6 promotes TGFβ-induced invasion and cell-cycle regulation via Lys63-linked polyubiquitination of Lys178 in TGFβ type I receptor. Cell Cycle 2015; 14:554-65. [PMID: 25622187 PMCID: PMC4347693 DOI: 10.4161/15384101.2014.990302] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Transforming growth factor β (TGFβ) can act either as a tumor promoter or a tumor suppressor in a context-dependent manner. High levels of TGFβ are found in prostate cancer tissues and correlate with poor patient prognosis. We recently identified a novel TGFβ-regulated signaling cascade in which TGFβ type I receptor (TβRI) is activated by the E3 ligase TNF-receptor-associated factor 6 (TRAF6) via the Lys63-linked polyubiquitination of TβRI. TRAF6 also contributes to activation of TNF-α-converting enzyme and presenilin-1, resulting in the proteolytic cleavage of TβRI and releasing the intracellular domain of TβRI, which is translocated to the nucleus to promote tumor invasiveness. In this report, we provide evidence that Lys178 of TβRI is polyubiquitinated by TRAF6. Moreover, our data suggest that TRAF6-mediated Lys63-linked ubiquitination of the TβRI intracellular domain is a prerequisite for TGFβ regulation of mRNA for cyclin D1 (CCND1), expression, as well as for the regulation of other genes controlling the cell cycle, differentiation, and invasiveness of prostate cancer cells.
Collapse
Affiliation(s)
- Reshma Sundar
- a Medical Biosciences ; Umeå University ; Umeå , Sweden
| | | | | | | |
Collapse
|
37
|
Hu Q, Chen J, Zhang J, Xu C, Yang S, Jiang H. IOX1, a JMJD2A inhibitor, suppresses the proliferation and migration of vascular smooth muscle cells induced by angiotensin II by regulating the expression of cell cycle-related proteins. Int J Mol Med 2015; 37:189-96. [PMID: 26530537 DOI: 10.3892/ijmm.2015.2393] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 10/01/2015] [Indexed: 11/06/2022] Open
Abstract
The epigenetic modification of vascular smooth muscle cell (VSMC) phenotypic switching, proliferation, migration, apoptosis and extracellular matrix synthesis is known to occur in atherosclerosis. The aim of the present study was to investigate the effects of IOX1, a Jumonji domain-containing 2A (JMJD2A) inhibitor, on regulation of the cell cycle in angiotensin II (Ang II)-stimulated VSMCs and to elucidate the possible mechanisms involved. The proliferation and migration of the Ang II-stimulated VSMCs in the presence or absence of IOX1 were evaluated in vitro. Flow cytometric analysis was used to determine the effects of IOX1 on cell cycle progression. RT-qPCR and western blot analysis were carried out to measure the expression levels of cell cycle-related genes. The trimethylation of histone H3 lysine 9 (H3K9me3) at the promoters of these genes was detected by chromatin immunoprecipitation (ChIP) assay. We confirmed that the JMJD2A levels were increased, whereas the H3K9me3 levels were decreased in the Ang II-stimulated VSMCs. The inhibition of JMJD2A by IOX1 suppressed the Ang II-induced cell proliferation, migration and cell cycle progression by inhibiting cyclin D1 expression and increasing p21 expression. The underlying mechanisms were related to the restoration of the H3K9me3 levels at the promoters of these genes. In conclusion, the findings of our study indicate that IOX1 exerts its anti-proliferative and anti-migratory effects by regulating the expression of the cell cycle-related proteins, cyclin D1 and p21.
Collapse
Affiliation(s)
- Qi Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Changwu Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shuo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
38
|
Lee-Rivera I, López E, Parrales A, Alvarez-Arce A, López-Colomé AM. Thrombin promotes the expression of Ccnd1 gene in RPE cells through the activation of converging signaling pathways. Exp Eye Res 2015; 139:81-9. [DOI: 10.1016/j.exer.2015.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 07/06/2015] [Accepted: 08/01/2015] [Indexed: 11/29/2022]
|
39
|
Bernhart E, Damm S, Heffeter P, Wintersperger A, Asslaber M, Frank S, Hammer A, Strohmaier H, DeVaney T, Mrfka M, Eder H, Windpassinger C, Ireson CR, Mischel PS, Berger W, Sattler W. Silencing of protein kinase D2 induces glioma cell senescence via p53-dependent and -independent pathways. Neuro Oncol 2015; 16:933-45. [PMID: 24463355 PMCID: PMC4057133 DOI: 10.1093/neuonc/not303] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Glioblastoma multiforme (GBM) is a highly aggressive tumor of the central nervous system with a dismal prognosis for affected patients. Aberrant protein kinase C (PKC) signaling has been implicated in gliomagenesis, and a member of the PKC-activated protein kinase D (PRKD) family, PRKD2, was identified as mediator of GBM growth in vitro and in vivo. Methods The outcome of PRKD2 silencing and pharmacological inhibition on glioma cell proliferation was established with different glioma cell lines. Western blotting, senescence assays, co-immunoprecipitation, fluorescence activated cell sorting, quantitative PCR, and immunofluorescence microscopy were utilized to analyze downstream signaling. Results RNA-interference (21-mer siRNA) and pharmacological inhibition (CRT0066101) of PRKD2 profoundly inhibited proliferation of p53wt (U87MG, A172, and primary GBM2), and p53mut (GM133, T98G, U251, and primary Gli25) glioma cells. In a xenograft experiment, PRKD2 silencing significantly delayed tumor growth of U87MG cells. PRKD2 silencing in p53wt and p53mut cells was associated with typical hallmarks of senescence and cell cycle arrest in G1. Attenuated AKT/PKB phosphorylation in response to PRKD2 silencing was a common observation made in p53wt and p53mut GBM cells. PRKD2 knockdown in p53wt cells induced upregulation of p53, p21, and p27 expression, decreased phosphorylation of CDK2 and/or CDK4, hypophosphorylation of retinoblastoma protein (pRb), and reduced transcription of E2F1. In p53mut GM133 and primary Gli25 cells, PRKD2 silencing increased p27 and p15 and reduced E2F1 transcription but did not affect pRb phosphorylation. Conclusions PRKD2 silencing induces glioma cell senescence via p53-dependent and -independent pathways.
Collapse
|
40
|
Jen J, Lin LL, Chen HT, Liao SY, Lo FY, Tang YA, Su WC, Salgia R, Hsu CL, Huang HC, Juan HF, Wang YC. Oncoprotein ZNF322A transcriptionally deregulates alpha-adducin, cyclin D1 and p53 to promote tumor growth and metastasis in lung cancer. Oncogene 2015; 35:2357-69. [PMID: 26279304 PMCID: PMC4865475 DOI: 10.1038/onc.2015.296] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 06/08/2015] [Accepted: 07/05/2015] [Indexed: 12/24/2022]
Abstract
ZNF322A encoding a classical Cys2His2 zinc finger transcription factor was previously revealed as a potential oncogene in lung cancer patients. However, the oncogenic role of ZNF322A and its underlying mechanism in lung tumorigenesis remain elusive. Here we show ZNF322A protein overexpression in 123 Asian and 74 Caucasian lung cancer patients. Multivariate Cox regression analysis indicated that ZNF322A was an independent risk factor for a poor outcome in lung cancer, corroborating the Kaplan–Meier results that patients with ZNF322A protein overexpression had significantly poorer overall survival than other patients. Overexpression of ZNF322A promoted cell proliferation and soft agar growth by prolonging cell cycle in S phase in multiple lung cell lines, including the immortalized lung cell BEAS-2B. In addition, ZNF322A overexpression enhanced cell migration and invasion, whereas knockdown of ZNF322A reduced cell growth, invasion and metastasis abilities in vitro and in vivo. Quantitative proteomic analysis revealed potential ZNF322A-regulated downstream targets, including alpha-adducin (ADD1), cyclin D1 (CCND1), and p53. Using luciferase promoter activity assay combined with site-directed mutagenesis and sequential chromatin immunoprecipitation-PCR assay, we found that ZNF322A could form a complex with c-Jun and cooperatively activate ADD1 and CCND1 but repress p53 gene transcription by recruiting differential chromatin modifiers, such as histone deacetylase 3, in an AP-1 element dependent manner. Reconstitution experiments indicated that CCND1 and p53 were important to ZNF322A-mediated promotion of cell proliferation, whereas ADD1 was necessary for ZNF322A-mediated cell migration and invasion. Our results provide compelling evidence that ZNF322A overexpression transcriptionally dysregulates genes involved in cell growth and motility therefore contributes to lung tumorigenesis and poor prognosis.
Collapse
Affiliation(s)
- J Jen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - L-L Lin
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - H-T Chen
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - S-Y Liao
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - F-Y Lo
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Y-A Tang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - W-C Su
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - R Salgia
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - C-L Hsu
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - H-C Huang
- Institute of Biomedical Informatics and Center for Systems and Synthetic Biology, National Yang-Ming University, Taipei, Taiwan
| | - H-F Juan
- Department of Life Science, Institute of Molecular and Cellular Biology, Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Y-C Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
41
|
HBx induces the proliferation of hepatocellular carcinoma cells via AP1 over-expressed as a result of ER stress. Biochem J 2015; 466:115-21. [PMID: 25428452 DOI: 10.1042/bj20140819] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide and chronic hepatitis B virus (HBV) infection is the most common risk factor for HCC. The HBV proteins can induce oncogenic or synergy effects with a hyperproliferative response on transformation into HCC. CREBH (cAMP-responsive, element-binding protein H), activated by stress in the endoplasmic reticulum (ER), is an ER-resident transmembrane bZIP (basic leucine zipper) transcription factor that is specifically expressed in the liver. In the present study, we address the role played by CREBH activated by ER stress in HBV-induced hepatic cell proliferation. We confirmed CREBH activation by ER stress and showed that it occurred as a result of/via hepatitis B virus X (HBx)-induced ER stress. CREBH activated by HBx increased the expression of AP-1 target genes through c-Jun induction. Under pathological conditions such as liver damage or liver regeneration, activated CREBH may have an important role to play in hepatic inflammation and cell proliferation, as an insulin receptor with dual functions under these conditions. We showed that CREBH activated by HBx interacted with HBx protein, leading to a synergistic effect on the expression of AP-1 target genes and the proliferation of HCC cells and mouse primary hepatocytes. In conclusion, in HBV-infected hepatic cells or patients with chronic HBV, CREBH may induce proliferation of hepatic cells in co-operation with HBx, resulting in HCC.
Collapse
|
42
|
Salusin-β Induces Smooth Muscle Cell Proliferation by Regulating Cyclins D1 and E Expression Through MAPKs Signaling Pathways. J Cardiovasc Pharmacol 2015; 65:377-85. [DOI: 10.1097/fjc.0000000000000209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Brown L, Wan H. Desmoglein 3: a help or a hindrance in cancer progression? Cancers (Basel) 2015; 7:266-86. [PMID: 25629808 PMCID: PMC4381258 DOI: 10.3390/cancers7010266] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/09/2015] [Accepted: 01/16/2015] [Indexed: 02/07/2023] Open
Abstract
Desmoglein 3 is one of seven desmosomal cadherins that mediate cell-cell adhesion in desmosomes. Desmosomes are the intercellular junctional complexes that anchor the intermediate filaments of adjacent cells and confer strong cell adhesion thus are essential in the maintenance of tissue architecture and structural integrity. Like adherens junctions, desmosomes function as tumour suppressors and are down regulated in the process of epithelial-mesenchymal transition and in tumour cell invasion and metastasis. However, recently several studies have shown that various desmosomal components, including desmoglein 3, are up-regulated in cancer with increased levels of expression correlating with the clinical stage of malignancy, implicating their potentiality to serve as a diagnostic and prognostic marker. Furthermore, in vitro studies have demonstrated that overexpression of desmoglein 3 in cancer cell lines activates several signal pathways that have an impact on cell morphology, adhesion and locomotion. These additional signalling roles of desmoglein 3 may not be associated to its adhesive function in desmosomes but rather function outside of the junctions, acting as a key regulator in the control of actin based cellular processes. This review will discuss recent advances which support the role of desmoglein 3 in cancer progression.
Collapse
Affiliation(s)
- Louise Brown
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK.
| | - Hong Wan
- Queen Mary University of London, Barts and the London School of Medicine and Dentistry, Center for Clinical and Diagnostic Oral Sciences, Institute of Dentistry, Blizard Building, London E1 2AT, UK.
| |
Collapse
|
44
|
Zhong J, Cao RX, Liu JH, Liu YB, Wang J, Liu LP, Chen YJ, Yang J, Zhang QH, Wu Y, Ding WJ, Hong T, Xiao XH, Zu XY, Wen GB. Nuclear loss of protein arginine N-methyltransferase 2 in breast carcinoma is associated with tumor grade and overexpression of cyclin D1 protein. Oncogene 2014; 33:5546-5558. [PMID: 24292672 DOI: 10.1038/onc.2013.500] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 09/17/2013] [Accepted: 10/18/2013] [Indexed: 11/08/2022]
Abstract
Human protein arginine N-methyltransferase 2 (PRMT2, HRMT1L1) is a protein that belongs to the arginine methyltransferase family, and it has diverse roles in transcriptional regulation through different mechanisms depending on its binding partners. In this study, we provide evidences for the negative effect of PRMT2 on breast cancer cell proliferation in vitro and in vivo. Morever, cyclin D1, one of the key modulators of cell cycle, was found to be downregulated by PRMT2, and PRMT2 was further shown to suppress the estrogen receptor α-binding affinity to the activator protein-1 (AP-1) site in cyclin D1 promoter through indirect binding with AP-1 site, resulting in the inhibition of cyclin D1 promoter activity in MCF-7 cells. Furthermore, a positive correlation between the expression of PRMT2 and cyclin D1 was confirmed in the breast cancer tissues by using tissue microarray assay. In addition, PRMT2 was found to show a high absent percentage in breast caner cell nuclei and the nuclear loss ratio of PRMT2 was demonstrated to positively correlate with cyclin D1 expression and the increasing tumor grade of invasive ductal carcinoma. Those results offer an essential insight into the effect of PRMT2 on breast carcinogenesis, and PRMT2 nuclear loss might be an important biological marker for the diagnosis of breast cancer.
Collapse
Affiliation(s)
- J Zhong
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - R-X Cao
- 1] Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China [2] Department of Metabolism and Endocrinology, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - J-H Liu
- 1] Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China [2] Department of Metabolism and Endocrinology, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - Y-B Liu
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - J Wang
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - L-P Liu
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - Y-J Chen
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - J Yang
- 1] Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China [2] Department of Metabolism and Endocrinology, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - Q-H Zhang
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - Y Wu
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - W-J Ding
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - T Hong
- 1] Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China [2] Department of Metabolism and Endocrinology, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - X-H Xiao
- 1] Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China [2] Department of Metabolism and Endocrinology, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - X-Y Zu
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| | - G-B Wen
- Institute of Clinical Medicine, First Affiliated Hospital of University of South China, Hengyang, PR China
| |
Collapse
|
45
|
Kaur G, Cholia RP, Mantha AK, Kumar R. DNA repair and redox activities and inhibitors of apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1): a comparative analysis and their scope and limitations toward anticancer drug development. J Med Chem 2014; 57:10241-56. [PMID: 25280182 DOI: 10.1021/jm500865u] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The apurinic/apyrimidinic endonuclease 1/redox effector factor 1 (APE1/Ref-1) is a multifunctional enzyme involved in DNA repair and activation of transcription factors through its redox function. The evolutionarily conserved C- and N-termini are involved in these functions independently. It is also reported that the activity of APE1/Ref-1 abruptly increases several-fold in various human cancers. The control over the outcomes of these two functions is emerging as a new strategy to combine enhanced DNA damage and chemotherapy in order to tackle the major hurdle of increased cancer cell growth and proliferation. Studies have targeted these two domains individually for the design and development of inhibitors for APE1/Ref-1. Here, we have made, for the first time, an attempt at a comparative analysis of APE1/Ref-1 inhibitors that target both DNA repair and redox activities simultaneously. We further discuss their scope and limitations with respect to the development of potential anticancer agents.
Collapse
Affiliation(s)
- Gagandeep Kaur
- Laboratory for Drug Design and Synthesis, Centre for Chemical and Pharmaceutical Sciences, School of Basic and Applied Sciences, Central University of Punjab , Bathinda, 151001, Punjab, India
| | | | | | | |
Collapse
|
46
|
Zhang Y, Baysac KC, Yee LF, Saporita AJ, Weber JD. Elevated DDX21 regulates c-Jun activity and rRNA processing in human breast cancers. Breast Cancer Res 2014; 16:449. [PMID: 25260534 PMCID: PMC4303128 DOI: 10.1186/s13058-014-0449-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 09/19/2014] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION The DDX21 RNA helicase has been shown to be a nucleolar and nuclear protein involved in ribosome RNA processing and AP-1 transcription. DDX21 is highly expressed in colon cancer, lymphomas, and some breast cancers, but little is known about how DDX21 might promote tumorigenesis. METHODS Immunohistochemistry was performed on a breast cancer tissue array of 187 patients. In order to study the subcellular localization of DDX21 in both tumor tissue and tumor cell lines, indirect immunofluorescence was applied. The effect of DDX21 knockdown was measured by cellular apoptosis, rRNA processing assays, soft agar growth and mouse xenograft imaging. AP-1 transcriptional activity was analyzed with a luciferase reporter and bioluminescence imaging, as well as qRT-PCR analysis of downstream target, cyclin D1, to determine the mechanism of action for DDX21 in breast tumorigenesis. RESULTS Herein, we show that DDX21 is highly expressed in breast cancer tissues and established cell lines. A significant number of mammary tumor tissues and established breast cancer cell lines exhibit nuclear but not nucleolar localization of DDX21. The protein expression level of DDX21 correlates with cell proliferation rate and is markedly induced by EGF signaling. Mechanistically, DDX21 is required for the phosphorylation of c-Jun on Ser73 and DDX21 deficiency markedly reduces the transcriptional activity of AP-1. Additionally, DDX21 promotes rRNA processing in multiple breast cancer cell lines. Tumor cells expressing high levels of endogenous DDX21 undergo apoptosis after acute DDX21 knockdown, resulting in significant reduction of tumorigenicity in vitro and in vivo. CONCLUSIONS Our findings indicate that DDX21 expression in breast cancer cells can promote AP-1 activity and rRNA processing, and thus, promote tumorigenesis by two independent mechanisms. DDX21 could serve as a marker for a subset of breast cancer patients with higher proliferation potential and may be used as a therapeutic target for a subset of breast cancer patients.
Collapse
|
47
|
Nakahata AM, Suzuki DE, Rodini CO, Fiuza ML, Okamoto OK. RNAi-mediated knockdown of E2F2 inhibits tumorigenicity of human glioblastoma cells. Oncol Lett 2014; 8:1487-1491. [PMID: 25202354 PMCID: PMC4156183 DOI: 10.3892/ol.2014.2369] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 06/19/2014] [Indexed: 02/06/2023] Open
Abstract
In a previous genome-wide expression profiling study, we identified E2F2 as a hyperexpressed gene in stem-like cells of distinct glioblastoma multiforme (GBM) specimens. Since the encoded E2F2 transcription factor has been implicated in both tumor suppression and tumor development, we conducted a functional study to investigate the pertinence of E2F2 to human gliomagenesis. E2F2 expression was knocked down by transfecting U87MG cells with plasmids carrying a specific silencing shRNA. Upon E2F2 silencing, in vitro cell proliferation was significantly reduced, as indicated by a time-course analysis of viable tumor cells. Anchorage-independent cell growth was also significantly inhibited after E2F2 silencing, based on cell colony formation in soft agar. Subcutaneous and orthotopic xenograft models of GBM in nude mice also indicated inhibition of tumor development in vivo, following E2F2 silencing. As expression of the E2F2 gene is associated with glioblastoma stem cells and is involved in the transformation of human astrocytes, the present findings suggest that E2F2 is involved in gliomagenesis and could be explored as a potential therapeutic target in malignant gliomas.
Collapse
Affiliation(s)
- Adriana M Nakahata
- Department of Neurology and Neurosurgery, Experimental Neurology Unit, Federal University of São Paulo, São Paulo 04023-900, Brazil ; Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Daniela E Suzuki
- Department of Neurology and Neurosurgery, Experimental Neurology Unit, Federal University of São Paulo, São Paulo 04023-900, Brazil ; Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Carolina O Rodini
- Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Mayara L Fiuza
- Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| | - Oswaldo K Okamoto
- Department of Genetics and Evolutionary Biology, Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo 05508-090, Brazil
| |
Collapse
|
48
|
Ye N, Ding Y, Wild C, Shen Q, Zhou J. Small molecule inhibitors targeting activator protein 1 (AP-1). J Med Chem 2014; 57:6930-48. [PMID: 24831826 PMCID: PMC4148154 DOI: 10.1021/jm5004733] [Citation(s) in RCA: 215] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Activator
protein 1 (AP-1) is a pivotal transcription factor that
regulates a wide range of cellular processes including proliferation,
apoptosis, differentiation, survival, cell migration, and transformation.
Accumulating evidence supports that AP-1 plays an important role in
several severe disorders including cancer, fibrosis, and organ injury,
as well as inflammatory disorders such as asthma, psoriasis, and rheumatoid
arthritis. AP-1 has emerged as an actively pursued drug discovery
target over the past decade. Excitingly, a selective AP-1 inhibitor
T-5224 (51) has been investigated in phase II human clinical
trials. Nevertheless, no effective AP-1 inhibitors have yet been approved
for clinical use. Despite significant advances achieved in understanding
AP-1 biology and function, as well as the identification of small
molecules modulating AP-1 associated signaling pathways, medicinal
chemistry efforts remain an urgent need to yield selective and efficacious
AP-1 inhibitors as a viable therapeutic strategy for human diseases.
Collapse
Affiliation(s)
- Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch , Galveston, Texas 77555, United States
| | | | | | | | | |
Collapse
|
49
|
Chen H, Yang Z, Ding C, Xiong A, Wild C, Wang L, Ye N, Cai G, Flores RM, Ding Y, Shen Q, Zhou J. Discovery of potent anticancer agent HJC0416, an orally bioavailable small molecule inhibitor of signal transducer and activator of transcription 3 (STAT3). Eur J Med Chem 2014; 82:195-203. [PMID: 24904966 DOI: 10.1016/j.ejmech.2014.05.049] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/15/2014] [Accepted: 05/21/2014] [Indexed: 12/15/2022]
Abstract
In a continuing effort to develop orally bioavailable small-molecule STAT3 inhibitors as potential therapeutic agents for human cancer, a series of novel diversified analogues based on our identified lead compound HJC0149 (1) (5-chloro-N-(1,1-dioxo-1H-1λ(6)-benzo[b]thiophen-6-yl)-2-hydroxybenzamide, Eur. J. Med. Chem. 2013, 62, 498-507) have been rationally designed, synthesized, and pharmacologically evaluated. Molecular docking studies and biological characterization supported our earlier findings that the O-alkylamino-tethered side chain on the hydroxyl group is an effective and essential structural determinant for improving biological activities and druglike properties of these molecules. Compounds with such modifications exhibited potent antiproliferative effects against breast and pancreatic cancer cell lines with IC50 values from low micromolar to nanomolar range. Among them, the newly discovered STAT3 inhibitor 12 (HJC0416) displayed an intriguing anticancer profile both in vitro and in vivo (i.p. & p.o.). More importantly, HJC0416 is an orally bioavailable anticancer agent as a promising candidate for further development.
Collapse
Affiliation(s)
- Haijun Chen
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States; College of Chemistry and Chemical Engineering, Fuzhou University, Fuzhou, Fujian 350002, China
| | - Zhengduo Yang
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, United States
| | - Chunyong Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Ailian Xiong
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, United States
| | - Christopher Wild
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Lili Wang
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, United States
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Guoshuai Cai
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, United States
| | - Rudolfo M Flores
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Ye Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Qiang Shen
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, United States.
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| |
Collapse
|
50
|
Pal D, Basu A. The unique protein kinase Cη: implications for breast cancer (review). Int J Oncol 2014; 45:493-8. [PMID: 24841225 DOI: 10.3892/ijo.2014.2443] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 02/18/2014] [Indexed: 11/05/2022] Open
Abstract
Deregulation of key signal transduction pathways that govern important cellular processes leads to cancer. The development of effective therapeutics for cancer warrants a comprehensive understanding of the signaling pathways that are deregulated in cancer. The protein kinase C (PKC) family has served as an attractive target for cancer therapy for decades owing to its crucial roles in several cellular processes. PKCη is a novel member of the PKC family that plays critical roles in various cellular processes such as growth, proliferation, differentiation and cell death. The regulation of PKCη appears to be unique compared to other PKC isozymes, and there are conflicting reports regarding its role in cancer. This review focuses on the unique aspects of PKCη in terms of its structure, regulation and subcellular distribution and speculates on how these features could account for its distinct functions. We have also discussed the functional implications of PKCη in cancer with particular emphasis on breast cancer.
Collapse
Affiliation(s)
- Deepanwita Pal
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center and Institute for Cancer Research, Fort Worth, TX 76107, USA
| | - Alakananda Basu
- Department of Molecular and Medical Genetics, University of North Texas Health Science Center and Institute for Cancer Research, Fort Worth, TX 76107, USA
| |
Collapse
|