51
|
Chen X, Qi Y, Wu Z, Wang X, Li J, Zhao D, Hou H, Li Y, Yu Z, Liu W, Wang M, Ren Y, Li Z, Yang H, Xu Y. Structural insights into preinitiation complex assembly on core promoters. Science 2021; 372:science.aba8490. [PMID: 33795473 DOI: 10.1126/science.aba8490] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 02/01/2021] [Accepted: 03/25/2021] [Indexed: 12/24/2022]
Abstract
Transcription factor IID (TFIID) recognizes core promoters and supports preinitiation complex (PIC) assembly for RNA polymerase II (Pol II)-mediated eukaryotic transcription. We determined the structures of human TFIID-based PIC in three stepwise assembly states and revealed two-track PIC assembly: stepwise promoter deposition to Pol II and extensive modular reorganization on track I (on TATA-TFIID-binding element promoters) versus direct promoter deposition on track II (on TATA-only and TATA-less promoters). The two tracks converge at an ~50-subunit holo PIC in identical conformation, whereby TFIID stabilizes PIC organization and supports loading of cyclin-dependent kinase (CDK)-activating kinase (CAK) onto Pol II and CAK-mediated phosphorylation of the Pol II carboxyl-terminal domain. Unexpectedly, TBP of TFIID similarly bends TATA box and TATA-less promoters in PIC. Our study provides structural visualization of stepwise PIC assembly on highly diversified promoters.
Collapse
Affiliation(s)
- Xizi Chen
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yilun Qi
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Zihan Wu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Xinxin Wang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Jiabei Li
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Dan Zhao
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Haifeng Hou
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yan Li
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Zishuo Yu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Weida Liu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Mo Wang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yulei Ren
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Ze Li
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Huirong Yang
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China
| | - Yanhui Xu
- Fudan University Shanghai Cancer Center, Institutes of Biomedical Sciences, State Key Laboratory of Genetic Engineering and Shanghai Key Laboratory of Medical Epigenetics, Shanghai Medical College of Fudan University, Shanghai 200032, China. .,The International Co-laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, China, Department of Systems Biology for Medicine, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, China.,Human Phenome Institute, Collaborative Innovation Center of Genetics and Development, School of Life Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
52
|
PUMA facilitates EMI1-promoted cytoplasmic Rad51 ubiquitination and inhibits DNA repair in stem and progenitor cells. Signal Transduct Target Ther 2021; 6:129. [PMID: 33785736 PMCID: PMC8009889 DOI: 10.1038/s41392-021-00510-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/20/2020] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Maintenance of genetic stability via proper DNA repair in stem and progenitor cells is essential for the tissue repair and regeneration, while preventing cell transformation after damage. Loss of PUMA dramatically increases the survival of mice after exposure to a lethal dose of ionizing radiation (IR), while without promoting tumorigenesis in the long-term survivors. This finding suggests that PUMA (p53 upregulated modulator of apoptosis) may have a function other than regulates apoptosis. Here, we identify a novel role of PUMA in regulation of DNA repair in embryonic or induced pluripotent stem cells (PSCs) and immortalized hematopoietic progenitor cells (HPCs) after IR. We found that PUMA-deficient PSCs and HPCs exhibited a significant higher double-strand break (DSB) DNA repair activity via Rad51-mediated homologous recombination (HR). This is because PUMA can be associated with early mitotic inhibitor 1 (EMI1) and Rad51 in the cytoplasm to facilitate EMI1-mediated cytoplasmic Rad51 ubiquitination and degradation, thereby inhibiting Rad51 nuclear translocation and HR DNA repair. Our data demonstrate that PUMA acts as a repressor for DSB DNA repair and thus offers a new rationale for therapeutic targeting of PUMA in regenerative cells in the context of DNA damage.
Collapse
|
53
|
Chlorogenic acid inhibits proliferation in human hepatoma cells by suppressing noncanonical NF-κB signaling pathway and triggering mitochondrial apoptosis. Mol Biol Rep 2021; 48:2351-2364. [PMID: 33738723 DOI: 10.1007/s11033-021-06267-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 03/05/2021] [Indexed: 12/24/2022]
Abstract
Chlorogenic acid (CGA), a phenylpropanoid derived from Eucommia ulmoides Oliver, has been shown to exhibit potent cytotoxic and anti-proliferative activities against several human cancers. However, the effects of CGA on hepatocellular carcinoma (HCC) and the underlying mechanisms have not been intensively studied. In this study, the CGA treatment effects on the viability of human hepatoma cells were investigated by MTT assay. Our data showed that CGA could dose-dependently inhibit the activity of human hepatoma cells Hep-G2 and Huh-7, but did not affect the activity and growth of normal human hepatocyte QSG-7701. The genes and pathways influenced by CGA treatment were explored by RNA sequencing and bioinformatics analysis, which identified 323 differentially expressed genes (DEGs) involved in multiple pharmacological signaling pathways such as MAPK, NF-κB, apoptosis and TGF-β signaling pathways. Further analyses by real-time quantitative PCR, Western blot and flow cytometry revealed that CGA effectually suppressed the noncanonical NF-κB signaling pathway, meanwhile it activated the mitochondrial apoptosis of HCC by upregulation of the BH3-only protein Bcl-2 binding component 3 (BBC3). Our findings demonstrated the potential of CGA in suppressing human hepatoma cells and provided a new insight into the anti-cancer mechanism of CGA.
Collapse
|
54
|
Zanetti TA, Biazi BI, Coatti GC, Baranoski A, Marques LA, Corveloni AC, Mantovani MS. Dimethoxycurcumin reduces proliferation and induces apoptosis in renal tumor cells more efficiently than demethoxycurcumin and curcumin. Chem Biol Interact 2021; 338:109410. [PMID: 33582110 DOI: 10.1016/j.cbi.2021.109410] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/22/2021] [Accepted: 02/08/2021] [Indexed: 12/21/2022]
Abstract
Curcumin (Cur), is a pigment with antiproliferative activity but has some pharmacokinetic limitations, which led researchers to look for more effective structure analogs. This work investigated the effects of Cur and compared them with the two analogs, demethoxycurcumin (DeMC) and dimethoxycurcumin (DiMC), to elucidate their mechanisms of action. The cytotoxic, antiproliferative, and genotoxic effects these compounds were correlated based on gene expression analysis in the human renal adenocarcinoma cells (786-O). Cur decreased CYP2D6 expression and exhibited cytotoxic effects, such as inducing monopolar spindle formation and mitotic arrest mediated by the increase in CDKN1A (p21) mRNA. This dysregulation induced cell death through a caspase-independent pathway but was mediated by decrease in MTOR and BCL2 mRNA expression, suggesting that apoptosis occurred by autophagy. DeMC and DiMC had similar effects in that they induced monopolar spindle and mitotic arrest, were genotoxic, and activated GADD45A, an important molecule in repair mechanisms, and CDKN1A. However, the induction of apoptosis by DeMC was delayed and regulated by the decrease of antiapoptotic mRNA BCL.XL and subsequent activation of caspase 9 and caspase 3/7. DiMC treatment increased the expression of CYP1A2, CYP2C19, and CYP3A4 and exhibited higher cytotoxicity compared with other compounds. It induced apoptosis by increasing mRNA expression of BBC3, MYC, and CASP7 and activation of caspase 9 and caspase 3/7. These data revealed that different gene regulation processes are involved in cell death induced by Cur, DeMC, and DiMC. All three can be considered as promising chemotherapy candidates, with DiMC showing the greatest potency.
Collapse
Affiliation(s)
- Thalita Alves Zanetti
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil.
| | - Bruna Isabela Biazi
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | | | - Adrivanio Baranoski
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | - Lilian Areal Marques
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | - Amanda Cristina Corveloni
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| | - Mario Sergio Mantovani
- Department of General Biology, Center of Biological Sciences, Londrina State University - UEL, Rodovia Celso Garcia Cid, Pr 445 Km 380, Londrina, Paraná, Brazil
| |
Collapse
|
55
|
Ulmke PA, Xie Y, Sokpor G, Pham L, Shomroni O, Berulava T, Rosenbusch J, Basu U, Fischer A, Nguyen HP, Staiger JF, Tuoc T. Post-transcriptional regulation by the exosome complex is required for cell survival and forebrain development via repression of P53 signaling. Development 2021; 148:dev.188276. [PMID: 33462115 DOI: 10.1242/dev.188276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022]
Abstract
Fine-tuned gene expression is crucial for neurodevelopment. The gene expression program is tightly controlled at different levels, including RNA decay. N6-methyladenosine (m6A) methylation-mediated degradation of RNA is essential for brain development. However, m6A methylation impacts not only RNA stability, but also other RNA metabolism processes. How RNA decay contributes to brain development is largely unknown. Here, we show that Exosc10, a RNA exonuclease subunit of the RNA exosome complex, is indispensable for forebrain development. We report that cortical cells undergo overt apoptosis, culminating in cortical agenesis upon conditional deletion of Exosc10 in mouse cortex. Mechanistically, Exosc10 directly binds and degrades transcripts of the P53 signaling-related genes, such as Aen and Bbc3. Overall, our findings suggest a crucial role for Exosc10 in suppressing the P53 pathway, in which the rapid turnover of the apoptosis effectors Aen and Bbc3 mRNAs is essential for cell survival and normal cortical histogenesis.
Collapse
Affiliation(s)
- Pauline Antonie Ulmke
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Yuanbin Xie
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany.,Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Gannan Medical University, 341000 Ganzhou, The People's Republic of China
| | - Godwin Sokpor
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany.,Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| | - Linh Pham
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany.,Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| | - Orr Shomroni
- Microarray and Deep-Sequencing Core Facility, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Tea Berulava
- German Center for Neurodegenerative Diseases, Goettingen 37075, Germany
| | - Joachim Rosenbusch
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Uttiya Basu
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Andre Fischer
- German Center for Neurodegenerative Diseases, Goettingen 37075, Germany.,Department for Psychiatry and Psychotherapy, University Medical Center, Georg-August-University Goettingen, Goettingen 37075, Germany.,Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Goettingen, Goettingen 37075, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| | - Jochen F Staiger
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany
| | - Tran Tuoc
- University Medical Center, Georg-August- University Goettingen, Goettingen 37075, Germany .,Department of Human Genetics, Ruhr University of Bochum, Bochum 44801, Germany
| |
Collapse
|
56
|
Maor-Nof M, Shipony Z, Lopez-Gonzalez R, Nakayama L, Zhang YJ, Couthouis J, Blum JA, Castruita PA, Linares GR, Ruan K, Ramaswami G, Simon DJ, Nof A, Santana M, Han K, Sinnott-Armstrong N, Bassik MC, Geschwind DH, Tessier-Lavigne M, Attardi LD, Lloyd TE, Ichida JK, Gao FB, Greenleaf WJ, Yokoyama JS, Petrucelli L, Gitler AD. p53 is a central regulator driving neurodegeneration caused by C9orf72 poly(PR). Cell 2021; 184:689-708.e20. [PMID: 33482083 PMCID: PMC7886018 DOI: 10.1016/j.cell.2020.12.025] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 10/07/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022]
Abstract
The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is a GGGGCC repeat expansion in the C9orf72 gene. We developed a platform to interrogate the chromatin accessibility landscape and transcriptional program within neurons during degeneration. We provide evidence that neurons expressing the dipeptide repeat protein poly(proline-arginine), translated from the C9orf72 repeat expansion, activate a highly specific transcriptional program, exemplified by a single transcription factor, p53. Ablating p53 in mice completely rescued neurons from degeneration and markedly increased survival in a C9orf72 mouse model. p53 reduction also rescued axonal degeneration caused by poly(glycine-arginine), increased survival of C9orf72 ALS/FTD-patient-induced pluripotent stem cell (iPSC)-derived motor neurons, and mitigated neurodegeneration in a C9orf72 fly model. We show that p53 activates a downstream transcriptional program, including Puma, which drives neurodegeneration. These data demonstrate a neurodegenerative mechanism dynamically regulated through transcription-factor-binding events and provide a framework to apply chromatin accessibility and transcription program profiles to neurodegeneration.
Collapse
Affiliation(s)
- Maya Maor-Nof
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| | - Zohar Shipony
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Lisa Nakayama
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Yong-Jie Zhang
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacob A Blum
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Patricia A Castruita
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriel R Linares
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Kai Ruan
- Department of Neurology, Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Gokul Ramaswami
- Department of Neurology, Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - David J Simon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Aviv Nof
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Manuel Santana
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Kyuho Han
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Michael C Bassik
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Daniel H Geschwind
- Department of Neurology, Program in Neurogenetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Laura D Attardi
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas E Lloyd
- Department of Neurology, Solomon H. Snyder Department of Neuroscience, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Justin K Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | | | - Aaron D Gitler
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
57
|
Oxidative Stress and Gene Expression Modifications Mediated by Extracellular Vesicles: An In Vivo Study of the Radiation-Induced Bystander Effect. Antioxidants (Basel) 2021; 10:antiox10020156. [PMID: 33494540 PMCID: PMC7911176 DOI: 10.3390/antiox10020156] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/16/2021] [Indexed: 12/27/2022] Open
Abstract
Radiation-induced bystander effect is a biological response in nonirradiated cells receiving signals from cells exposed to ionising radiation. The aim of this in vivo study was to analyse whether extracellular vesicles (EVs) originating from irradiated mice could induce modifications in the redox status and expression of radiation-response genes in bystander mice. C57BL/6 mice were whole-body irradiated with 0.1-Gy and 2-Gy X-rays, and EVs originating from mice irradiated with the same doses were injected into naïve, bystander mice. Lipid peroxidation in the spleen and plasma reactive oxygen metabolite (ROM) levels increased 24 h after irradiation with 2 Gy. The expression of antioxidant enzyme genes and inducible nitric oxide synthase 2 (iNOS2) decreased, while cell cycle arrest-, senescence- and apoptosis-related genes were upregulated after irradiation with 2 Gy. In bystander mice, no significant alterations were observed in lipid peroxidation or in the expression of genes connected to cell cycle arrest, senescence and apoptosis. However, there was a systemic increase in the circulating ROM level after an intravenous EV injection, and EVs originating from 2-Gy-irradiated mice caused a reduced expression of antioxidant enzyme genes and iNOS2 in bystander mice. In conclusion, we showed that ionising radiation-induced alterations in the cellular antioxidant system can be transmitted in vivo in a bystander manner through EVs originating from directly irradiated animals.
Collapse
|
58
|
Hyaluronic acid is a negative regulator of mucosal fibroblast-mediated enhancement of HIV infection. Mucosal Immunol 2021; 14:1203-1213. [PMID: 33976386 PMCID: PMC8379073 DOI: 10.1038/s41385-021-00409-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/08/2021] [Accepted: 04/16/2021] [Indexed: 02/08/2023]
Abstract
The majority of HIV infections are established through the genital or rectal mucosa. Fibroblasts are abundant in these tissues, and although not susceptible to infection, can potently enhance HIV infection of CD4+ T cells. Hyaluronic acid (HA) is a major component of the extracellular matrix of fibroblasts, and its levels are influenced by the inflammatory state of the tissue. Since inflammation is known to facilitate HIV sexual transmission, we investigated the role of HA in genital mucosal fibroblast-mediated enhancement of HIV infection. Depletion of HA by CRISPR-Cas9 in primary foreskin fibroblasts augmented the ability of the fibroblasts to increase HIV infection of CD4+ T cells. This amplified enhancement required direct contact between the fibroblasts and CD4+ T cells, and could be attributed to both increased rates of trans-infection and the increased ability of HA-deficient fibroblasts to push CD4+ T cells into a state of higher permissivity to infection. This HIV-permissive state was characterized by differential expression of genes associated with regulation of cell metabolism and death. Our results suggest that conditions resulting in diminished cell-surface HA on fibroblasts, such as genital inflammation, can promote HIV transmission by conditioning CD4+ T cells toward a state more vulnerable to infection by HIV.
Collapse
|
59
|
Lee J, Choi A, Cho SY, Jun Y, Na D, Lee A, Jang G, Kwon JY, Kim J, Lee S, Lee C. Genome-scale CRISPR screening identifies cell cycle and protein ubiquitination processes as druggable targets for erlotinib-resistant lung cancer. Mol Oncol 2020; 15:487-502. [PMID: 33188726 PMCID: PMC7858278 DOI: 10.1002/1878-0261.12853] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 01/01/2023] Open
Abstract
Erlotinib is highly effective in lung cancer patients with epidermal growth factor receptor (EGFR) mutations. However, despite initial favorable responses, most patients rapidly develop resistance to erlotinib soon after the initial treatment. This study aims to identify new genes and pathways associated with erlotinib resistance mechanisms in order to develop novel therapeutic strategies. Here, we induced knockout (KO) mutations in erlotinib‐resistant human lung cancer cells (NCI‐H820) using a genome‐scale CRISPR‐Cas9 sgRNA library to screen for genes involved in erlotinib susceptibility. The spectrum of sgRNAs incorporated among erlotinib‐treated cells was substantially different to that of the untreated cells. Gene set analyses showed a significant depletion of ‘cell cycle process’ and ‘protein ubiquitination pathway’ genes among erlotinib‐treated cells. Chemical inhibitors targeting genes in these two pathways, such as nutlin‐3 and carfilzomib, increased cancer cell death when combined with erlotinib in both in vitro cell line and in vivo patient‐derived xenograft experiments. Therefore, we propose that targeting cell cycle processes or protein ubiquitination pathways are promising treatment strategies for overcoming resistance to EGFR inhibitors in lung cancer.
Collapse
Affiliation(s)
- Jieun Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea.,Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea
| | - Ahyoung Choi
- Department of Bio-Information Science, Ewha Womans University, Seoul, Korea
| | - Sung-Yup Cho
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Korea
| | - Yukyung Jun
- Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea.,The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Deukchae Na
- Ewha Institute of Convergence Medicine, Ewha Womans University Mokdong Hospital, Seoul, Korea
| | - Ahra Lee
- Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea
| | - Giyong Jang
- Department of Life Science, Ewha Womans University, Seoul, Korea.,Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea
| | - Jee Young Kwon
- Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea.,The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Jaesang Kim
- Department of Life Science, Ewha Womans University, Seoul, Korea.,Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea
| | - Sanghyuk Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea.,Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea.,Department of Bio-Information Science, Ewha Womans University, Seoul, Korea
| | - Charles Lee
- Department of Life Science, Ewha Womans University, Seoul, Korea.,Ewha-JAX Cancer Immunotherapy Research Center, Ewha Womans University, Seoul, Korea.,The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA.,Precision Medicine Center, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
60
|
Bou Sleiman M, Jha P, Houtkooper R, Williams RW, Wang X, Auwerx J. The Gene-Regulatory Footprint of Aging Highlights Conserved Central Regulators. Cell Rep 2020; 32:108203. [PMID: 32997995 PMCID: PMC7527782 DOI: 10.1016/j.celrep.2020.108203] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Many genes and pathways have been linked to aging, yet our understanding of underlying molecular mechanisms is still lacking. Here, we measure changes in the transcriptome, histone modifications, and DNA methylome in three metabolic tissues of adult and aged mice. Transcriptome and methylome changes dominate the liver aging footprint, whereas heart and muscle globally increase chromatin accessibility, especially in aging pathways. In mouse and human data from multiple tissues and regulatory layers, age-related transcription factor expression changes and binding site enrichment converge on putative aging modulators, including ZIC1, CXXC1, HMGA1, MECP2, SREBF1, SREBF2, ETS2, ZBTB7A, and ZNF518B. Using Mendelian randomization, we establish possible epidemiological links between expression of some of these transcription factors or their targets, including CXXC1, ZNF518B, and BBC3, and longevity. We conclude that conserved modulators are at the core of the molecular footprint of aging, and variation in tissue-specific expression of some may affect human longevity.
Collapse
Affiliation(s)
- Maroun Bou Sleiman
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Pooja Jha
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Riekelt Houtkooper
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee, Memphis, TN 38163, USA
| | - Xu Wang
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland.
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne 1015, Switzerland.
| |
Collapse
|
61
|
Wolf ER, Mabry AR, Damania B, Mayo LD. Mdm2-mediated neddylation of pVHL blocks the induction of antiangiogenic factors. Oncogene 2020; 39:5228-5239. [PMID: 32555333 PMCID: PMC7368819 DOI: 10.1038/s41388-020-1359-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 05/27/2020] [Accepted: 06/05/2020] [Indexed: 01/26/2023]
Abstract
Mutations in the tumor suppressor TP53 are rare in renal cell carcinomas. p53 is a key factor for inducing antiangiogenic genes and RCC are highly vascularized, which suggests that p53 is inactive in these tumors. One regulator of p53 is the Mdm2 oncogene, which is correlated with high-grade, metastatic tumors. However, the sole activity of Mdm2 is not just to regulate p53, but it can also function independent of p53 to regulate the early stages of metastasis. Here, we report that the oncoprotein Mdm2 can bind directly to the tumor suppressor VHL, and conjugate nedd8 to VHL within a region that is important for the p53-VHL interaction. Nedd8 conjugated VHL is unable to bind to p53 thereby preventing the induction of antiangiogenic factors. These results highlight a previously unknown oncogenic function of Mdm2 during the progression of cancer to promote angiogenesis through the regulation of VHL. Thus, the Mdm2-VHL interaction represents a pathway that impacts tumor angiogenesis.
Collapse
Affiliation(s)
- Eric R Wolf
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Alexander R Mabry
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Blossom Damania
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Lindsey D Mayo
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
62
|
Tulsawani R, Sharma P, Sethy NK, Kumari P, Ganju L, Prakash S, Chouhan S. Acute exposure of 532 nm laser differentially regulates skin tissue transcription factors. PLoS One 2020; 15:e0230175. [PMID: 32191734 PMCID: PMC7082019 DOI: 10.1371/journal.pone.0230175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 02/24/2020] [Indexed: 12/16/2022] Open
Abstract
High energy laser, particularly 532 nm, is widely used in defense and medical applications and there is need to address its occupational safety. Thermal and non-thermal effects of 532 nm high energy laser on skin are cause of concern. This study indicates impact of 532 nm laser on rat skin and first of its kind of attempt to understand transcriptional activation of genes as an early response following laser exposure. Skin of experimental rats were exposed to 532 nm radiance at 0.1, 0.25 and 0.50 W/cm2 for 10 sec. Thermographic changes of skin exposed to 532 nm laser exhibited increased Tmax temperature in radiance dependent manner. After thermal imaging, skin of experimental rats was collected 1 h post laser exposure for studying differential gene expression. The skin exposed to lower power density (0.1 W/cm2) did not show significant changes in expression of gene pathways studied. At moderate radiance (0.25 W/cm2), predominantly canonical wnt/B-catenin pathway genes notch1, axin2, ccdn1, wnt5a and redox homeostasis genes; txn1, nqo1 and txnrd1 were expressed. At higher radiance (0.5 W/cm2), significant repression of genes related to wound healing process particularly notch/wnt pathway viz. hes5, wnt1, wn3b with higher expression of dab2 was recorded. The data obtained from these studies would help in drawing safety limits for skin exposure to 532 nm laser. Further, genes expressed at moderate and high level of radiance exposure to skin were distinct and differential and provide new avenue to configure pathway to counteract laser induced delay in tissue injury and hair follicular damage.
Collapse
Affiliation(s)
| | - Purva Sharma
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | | | - Pooja Kumari
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Lilly Ganju
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| | - Satya Prakash
- Laser Science and Technology Centre, Metcalfe House, Delhi, India
| | - Satish Chouhan
- Defence Institute of Physiology and Allied Sciences, Delhi, India
| |
Collapse
|
63
|
Fluorescent Light Energy (FLE) Acts on Mitochondrial Physiology Improving Wound Healing. J Clin Med 2020; 9:jcm9020559. [PMID: 32085605 PMCID: PMC7073965 DOI: 10.3390/jcm9020559] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/30/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
Fluorescent light energy (FLE) has been used to treat various injured tissues in a non-pharmacological and non-thermal fashion. It was applied to stimulate cell proliferation, accelerate healing in chronic and acute wounds, and reduce pain and inflammation. FLE has been shown to reduce pro-inflammatory cytokines while promoting an environment conducive to healing. A possible mechanism of action of FLE is linked to regulation of mitochondrial homeostasis. This work aims to investigate the effect of FLE on mitochondrial homeostasis in an in vitro model of inflammation. Confocal microscopy and gene expression profiling were performed on cultures of inflamed human dermal fibroblasts treated with either direct light from a multi-LED lamp, or FLE from either an amorphous gel or sheet hydrogel matrix. Assessment using confocal microscopy revealed mitochondrial fragmentation in inflamed cells, likely due to exposure to inflammatory cytokines, however, mitochondrial networks were restored to normal 24-h after treatment with FLE. Moreover, gene expression analysis found that treatment with FLE resulted in upregulation of uncoupling protein 1 (UCP1) and carnitine palmitoyltransferase 1B (CPT1B) genes, which encode proteins favoring mitochondrial ATP production through oxidative phosphorylation and lipid β-oxidation, respectively. These observations demonstrate a beneficial effect of FLE on mitochondrial homeostasis in inflamed cells.
Collapse
|
64
|
Sudhan DR, Guerrero-Zotano A, Won H, González Ericsson P, Servetto A, Huerta-Rosario M, Ye D, Lee KM, Formisano L, Guo Y, Liu Q, Kinch LN, Red Brewer M, Dugger T, Koch J, Wick MJ, Cutler RE, Lalani AS, Bryce R, Auerbach A, Hanker AB, Arteaga CL. Hyperactivation of TORC1 Drives Resistance to the Pan-HER Tyrosine Kinase Inhibitor Neratinib in HER2-Mutant Cancers. Cancer Cell 2020; 37:183-199.e5. [PMID: 31978326 PMCID: PMC7301608 DOI: 10.1016/j.ccell.2019.12.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 09/30/2019] [Accepted: 12/24/2019] [Indexed: 02/07/2023]
Abstract
We developed neratinib-resistant HER2-mutant cancer cells by gradual dose escalation. RNA sequencing identified TORC1 signaling as an actionable mechanism of drug resistance. Primary and acquired neratinib resistance in HER2-mutant breast cancer patient-derived xenografts (PDXs) was also associated with TORC1 hyperactivity. Genetic suppression of RAPTOR or RHEB ablated P-S6 and restored sensitivity to the tyrosine kinase inhibitor. The combination of the TORC1 inhibitor everolimus and neratinib potently arrested the growth of neratinib-resistant xenografts and organoids established from neratinib-resistant PDXs. RNA and whole-exome sequencing revealed RAS-mediated TORC1 activation in a subset of neratinib-resistant models. DNA sequencing of HER2-mutant tumors clinically refractory to neratinib, as well as circulating tumor DNA profiling of patients who progressed on neratinib, showed enrichment of genomic alterations that converge to activate the mTOR pathway.
Collapse
Affiliation(s)
- Dhivya R Sudhan
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Helen Won
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Alberto Servetto
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mariela Huerta-Rosario
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dan Ye
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kyung-Min Lee
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luigi Formisano
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan Guo
- Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - Qi Liu
- Center for Quantitative Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lisa N Kinch
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Monica Red Brewer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Teresa Dugger
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - James Koch
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | | | | | | | | | - Ariella B Hanker
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Carlos L Arteaga
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
65
|
Sanphui P, Kumar Das A, Biswas SC. Forkhead Box O3a requires BAF57, a subunit of chromatin remodeler SWI/SNF complex for induction of p53 up‐regulated modulator of apoptosis (Puma) in a model of Parkinson’s disease. J Neurochem 2020; 154:547-561. [DOI: 10.1111/jnc.14969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 01/16/2020] [Accepted: 01/16/2020] [Indexed: 01/01/2023]
Affiliation(s)
- Priyankar Sanphui
- Cell Biology and Physiology Division CSIR‐Indian Institute of Chemical Biology Kolkata India
| | - Anoy Kumar Das
- Cell Biology and Physiology Division CSIR‐Indian Institute of Chemical Biology Kolkata India
| | - Subhas C. Biswas
- Cell Biology and Physiology Division CSIR‐Indian Institute of Chemical Biology Kolkata India
| |
Collapse
|
66
|
Chen W, Li X. MiR-222-3p Promotes Cell Proliferation and Inhibits Apoptosis by Targeting PUMA (BBC3) in Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2020; 19:1533033820922558. [PMID: 32588752 PMCID: PMC7325542 DOI: 10.1177/1533033820922558] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 03/12/2020] [Accepted: 03/26/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs have been demonstrated to be critical regulators in tumor progression, including non-small cell lung cancer. MicroRNA-222-3p has been reported to function as a tumor suppressor or oncogene in several types of cancer, but its function role in non-small cell lung cancer has not been uncovered. In this study, we first found the expression of microRNA-222-3p was significantly increased in non-small cell lung cancer tissues and cell lines. MicroRNA-222-3p inhibitor decreased the activity of non-small cell lung cancer cells to proliferate and increased cell apoptosis using cell counting kit-8, flow cytometry, and caspase-3 activity analysis. Overexpressed microRNA-222-3p in non-small cell lung cancer cells promoted cell proliferation, but decreased cell apoptosis. Moreover, Bcl-2-binding component 3 was the target gene of microRNA-222-3p, and its knockdown weakened the regulatory effect of microRNA-222-3p inhibitor on cell proliferation and apoptosis in non-small cell lung cancer cells. In conclusion, microRNA-222-3p plays a significant role in the regulation of Bcl-2-binding component 3 expression and might be a promising target for clinical non-small cell lung cancer therapy.
Collapse
MESH Headings
- Adenocarcinoma of Lung/genetics
- Adenocarcinoma of Lung/metabolism
- Adenocarcinoma of Lung/pathology
- Apoptosis
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Proliferation
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- MicroRNAs/genetics
- Middle Aged
- Prognosis
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Weijun Chen
- Department of Radiotherapy, Taizhou Center Hospital, Taizhou City, Zhejiang Province, China
| | - Xiaobo Li
- Department of Respiratory Medicine, Taizhou First People’s Hospital, Taizhou City, Zhejiang Province, China
| |
Collapse
|
67
|
Gao W, Gao J, Chen L, Ren Y, Ma J. Targeting XIST induced apoptosis of human osteosarcoma cells by activation of NF-kB/PUMA signal. Bioengineered 2019; 10:261-270. [PMID: 31189404 PMCID: PMC6592364 DOI: 10.1080/21655979.2019.1631104] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The long noncoding RNA X-inactive specific transcript (XIST) plays vital roles in tumor progression. However, the underlying mechanisms remain unclear. This study investigated the effects and mechanisms of targeting XIST on osteosarcoma (OS) cells in vitro and in vivo. We used shRNA to knockdown XIST to evaluate cell growth and apoptosis in U2OS cells in vitro and xenograft formation in vivo. An observed relationship between XIST and the p53 upregulated modulator of apoptosis (PUMA) and nuclear factor-kappa B (NF-kB) pathway was further explored by using small interfering RNA (siRNA). Our results showed that suppression of XIST by short hairpin RNA (shRNA) impeded U2OS cell growth, induced apoptosis and lessened OS xenograft tumor growth. Targeting XIST increased NF-kB-dependent PUMA upregulation in U2OS cells. Upregulation of PUMA is correlated with suppression of XIST-induced apoptosis in U2OS cells. Therefore, inhibition of XIST could promote U2OS cell death via activation of NF-kB/PUMA pathways.
Collapse
Affiliation(s)
- Weiliang Gao
- a Department of Spine Surgery , The 107 Hospital of the People's Liberation Army , Yantai , Shandong , China
| | - Jisheng Gao
- a Department of Spine Surgery , The 107 Hospital of the People's Liberation Army , Yantai , Shandong , China
| | - Longying Chen
- b Department of Orthopedics , Linyi Central Hospital , Linyi , Shangdong , China
| | - Yande Ren
- c Department of Radiology , The affiliated hospital of Qingdao University , Qingdao , Shandong , China
| | - Jinfeng Ma
- d Department of Spine Surgery , The affiliated hospital of Qingdao University , Qingdao , Shandong , China
| |
Collapse
|
68
|
Khosla R, Hemati H, Rastogi A, Ramakrishna G, Sarin SK, Trehanpati N. miR-26b-5p helps in EpCAM+cancer stem cells maintenance via HSC71/HSPA8 and augments malignant features in HCC. Liver Int 2019; 39:1692-1703. [PMID: 31276277 DOI: 10.1111/liv.14188] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Targeting cancer stem cells (CSCs) in hepatocellular carcinoma (HCC) is difficult because of their similarities with normal stem cells (NSCs). EpCAM can identify CSCs from EpCAM+AFP+HCC cases, but is also expressed on NSCs. We aimed to distinguish the two using integrated protein, mRNA and miRNA profiling. METHODS iTRAQ based protein profiling and Next Generation Sequencing (NGS) was performed on EpCAM+/EpCAM- cells isolated from HCC (Ep+CSC, Ep- HCC) and EpCAM+ cells from non-cancerous/non-cirrhotic control liver tissues (Ep+NSC). Validations were done using qRT-PCR, flowcytometry and western blotting followed by in vitro and in vivo functional studies. RESULTS 11 proteins were overexpressed (>3 fold) in Ep+CSCs compared to Ep- HCC and Ep+NSC cells. However, RNA-sequencing confirmed the Ep+CSC specific up-regulation of only HSPA8, HNRNPC, MPST and GAPDH mRNAs among these. Database search combined with miRNA profiling revealed Ep+ CSC specific down-regulation of 29 miRNAs targeting these four genes. Of these, only miR-26b-5p was found to target both HSPA8 and EpCAM. Validation of HSPA8 overexpression and miR-26b-5p down-regulation followed by linear regression analysis established a negative correlation between the two. Functional studies demonstrated that reduced miR-26b-5p expression increased the spheroid formation, migration, invasion and tumourigenicity of Ep+ CSCs. Furthermore, anti-miR-26b-5p increased the number of Ep+ CSCs with a concomitant overexpression of stemness genes and reduction of proapoptotic protein BBC3, which is a known substrate of HSPA8. CONCLUSION miR-26b-5p imparts metastatic properties and helps in maintenance of Ep+ CSCs via HSPA8. Thus, miR-26b-5p and HSPA8 could serve as molecular targets for selectively eliminating the Ep+ CSC population in human HCCs.
Collapse
Affiliation(s)
- Ritu Khosla
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Hamed Hemati
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India.,Department of Biotechnology, Punjab University, Chandigarh, India
| | | | - Gayatri Ramakrishna
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| | - Shiv K Sarin
- Department of Hepatology, ILBS, New Delhi, India
| | - Nirupma Trehanpati
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences (ILBS), New Delhi, India
| |
Collapse
|
69
|
Bandolik JJ, Hamacher A, Schrenk C, Weishaupt R, Kassack MU. Class I-Histone Deacetylase (HDAC) Inhibition is Superior to pan-HDAC Inhibition in Modulating Cisplatin Potency in High Grade Serous Ovarian Cancer Cell Lines. Int J Mol Sci 2019; 20:ijms20123052. [PMID: 31234549 PMCID: PMC6627993 DOI: 10.3390/ijms20123052] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 01/23/2023] Open
Abstract
High grade serous ovarian cancer (HGSOC) is the most common and aggressive ovarian cancer subtype with the worst clinical outcome due to intrinsic or acquired drug resistance. Standard treatment involves platinum compounds. Cancer development and chemoresistance is often associated with an increase in histone deacetylase (HDAC) activity. The purpose of this study was to examine the potential of HDAC inhibitors (HDACi) to increase platinum potency in HGSOC. Four HGSOC cell lines with different cisplatin sensitivity were treated with combinations of cisplatin and entinostat (class I HDACi), panobinostat (pan-HDACi), or nexturastat A (class IIb HDACi), respectively. Inhibition of class I HDACs by entinostat turned out superior in increasing cisplatin potency than pan-HDAC inhibition in cell viability assays (MTT), apoptosis induction (subG1), and caspase 3/7 activation. Entinostat was synergistic with cisplatin in all cell lines in MTT and caspase activation assays. MTT assays gave combination indices (CI values) < 0.9 indicating synergism. The effect of HDAC inhibitors could be attributed to the upregulation of pro-apoptotic genes (CDNK1A, APAF1, PUMA, BAK1) and downregulation of survivin. In conclusion, the combination of entinostat and cisplatin is synergistic in HGSOC and could be an effective strategy for the treatment of aggressive ovarian cancer.
Collapse
Affiliation(s)
- Jan J Bandolik
- Institute for Pharmaceutical and Medicinal Chemistry, University of Duesseldorf, 40225 Duesseldorf, Germany.
| | - Alexandra Hamacher
- Institute for Pharmaceutical and Medicinal Chemistry, University of Duesseldorf, 40225 Duesseldorf, Germany.
| | - Christian Schrenk
- Institute for Pharmaceutical and Medicinal Chemistry, University of Duesseldorf, 40225 Duesseldorf, Germany.
| | - Robin Weishaupt
- Institute for Computer Science, Computational Complexity and Cryptology, University of Duesseldorf, 40225 Duesseldorf, Germany.
| | - Matthias U Kassack
- Institute for Pharmaceutical and Medicinal Chemistry, University of Duesseldorf, 40225 Duesseldorf, Germany.
| |
Collapse
|
70
|
Ma L, Chen Y, Han R, Wang S. Benzyl isothiocyanate inhibits invasion and induces apoptosis via reducing S100A4 expression and increases PUMA expression in oral squamous cell carcinoma cells. ACTA ACUST UNITED AC 2019; 52:e8409. [PMID: 30970087 PMCID: PMC6459467 DOI: 10.1590/1414-431x20198409] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 02/01/2019] [Indexed: 01/17/2023]
Abstract
Benzyl isothiocyanate (BITC) has been shown to inhibit invasion and induce apoptosis of various types of cancer. However, its role on human oral squamous cell carcinoma (OSCC) cells is still not well elucidated. In the present study, we investigated the effect of BITC on apoptosis and invasion of SCC9 cells, and its underlying mechanisms in vitro and in vivo. SCC9 cells were exposed to BITC (5 and 25 μM) for 24 and 48 h. Cell growth, apoptosis, invasion, and migration were detected in vitro by MTT, FITC-conjugated annexin V/propidium iodide staining followed by flow cytometry, Matrigel-coated semi-permeable modified Boyden, and wound-healing assay. S100A4, PUMA, and MMP-9 expressions were detected to investigate its mechanisms. Xenotransplantation experiments were used to investigate the role of BITC on tumor growth and lung metastasis. BITC inhibited cell viability and induced cell apoptosis in a dose- and time-dependent manner through upregulation of PUMA signals. BITC inhibited cell invasion and migration by downregulation of S100A4 dependent MMP-9 signals. The ip administration of BITC reduced tumor growth but not lung metastasis of SCC9 cells subcutaneously implanted in nude mice. BITC treatment activated pro-apoptotic PUMA and inhibited S100A4-dependent MMP-9 signals, resulting in the inhibition of cell growth and invasion in cultured and xenografted SCC9 cells. Thereby, BITC is a potential therapeutic approach for OSCC.
Collapse
Affiliation(s)
- Lei Ma
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yongjun Chen
- Department of Traditional Chinese medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Rui Han
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Shuangyi Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
71
|
Kiuchi Z, Nishibori Y, Kutsuna S, Kotani M, Hada I, Kimura T, Fukutomi T, Fukuhara D, Ito-Nitta N, Kudo A, Takata T, Ishigaki Y, Tomosugi N, Tanaka H, Matsushima S, Ogasawara S, Hirayama Y, Takematsu H, Yan K. GLCCI1 is a novel protector against glucocorticoid‐induced apoptosis in T cells. FASEB J 2019; 33:7387-7402. [DOI: 10.1096/fj.201800344rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Zentaro Kiuchi
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Yukino Nishibori
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Satoru Kutsuna
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Masashi Kotani
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Ichiro Hada
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Toru Kimura
- Department of Toxicology and PharmacologyKyorin University School of MedicineTokyoJapan
| | - Toshiyuki Fukutomi
- Department of Toxicology and PharmacologyKyorin University School of MedicineTokyoJapan
| | - Daisuke Fukuhara
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Noriko Ito-Nitta
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| | - Akihiko Kudo
- Department of AnatomyKyorin University School of MedicineTokyoJapan
| | - Takanobu Takata
- Medical Research InstituteKanazawa Medical UniversityUchinada-machiJapan
| | - Yasuhito Ishigaki
- Medical Research InstituteKanazawa Medical UniversityUchinada-machiJapan
| | - Naohisa Tomosugi
- Medical Research InstituteKanazawa Medical UniversityUchinada-machiJapan
| | - Hirotoshi Tanaka
- Department of RheumatologyCenter for Antibody and Vaccine TherapyInstitute of Medical ScienceUniversity of TokyoTokyoJapan
| | - Satsuki Matsushima
- Department of Laboratory MedicineKyorin University School of MedicineTokyoJapan
| | - Shinya Ogasawara
- Research and Development DepartmentDenka Seiken Company, LimitedGosenJapan
| | - Yoshiaki Hirayama
- Research and Development DepartmentDenka Seiken Company, LimitedGosenJapan
| | - Hiromu Takematsu
- Department of Biological ChemistryHuman Health ScienceKyoto University Graduate School of MedicineKyotoJapan
- Department of Molecular Cell BiologyFaculty of Medical TechnologyGraduate School of Health SciencesFujita Health University
| | - Kunimasa Yan
- Department of PediatricsKyorin University School of MedicineTokyoJapan
| |
Collapse
|
72
|
Erdem Kuruca S, Çetin MB, Akgün Dar K, Özerkan D. Protective effects of cytokine combinations against the apoptotic activity of glucocorticoids on CD34 + hematopoietic stem/progenitor cells. Cytotechnology 2019; 71:67-77. [PMID: 30603917 DOI: 10.1007/s10616-018-0265-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 10/09/2018] [Indexed: 12/26/2022] Open
Abstract
Haematopoietic stem cells can self-renew and produce progenitor cells, which have a high proliferation capacity. Chemotherapeutic drugs are toxic to normal cells as well as cancer cells, and glucocorticoids (GCs), which are essential drugs for many chemotherapeutic protocols, efficiently induce apoptosis not only in malignant cells but also in normal haematopoietic cells. Studies have shown that haematopoietic cytokines can prevent the apoptosis induced by chemotherapy and decrease the toxic effects of these drugs. However, the apoptosis induction mechanism of GCs in CD34+ haematopoietic cells and the anti-apoptotic effects of cytokines have not been well elucidated. In this study, we investigated the apoptotic effects of GCs on CD34+, a haematopoietic stem/progenitor cell (HSPC) population, and demonstrated the protective effects of haematopoietic cytokines. We used a cytokine cocktail containing early-acting cytokines, namely, interleukin-3 (IL-3), thrombopoietin, stem cell factor and flt3/flk2 ligand, and dexamethasone and prednisolone were used as GCs. Apoptotic mechanisms were assessed by immunohistochemical staining and quantified using H-scoring. Dexamethasone and prednisolone induced apoptosis in CD34+ HSPCs. GC treatment caused a significant increase in apoptotic Fas, caspase-3, cytochrome c and Bax, but a significant decrease in anti-apoptotic Bcl-2. Furthermore, as expected, cytokines caused a significant decrease in all apoptotic markers and a significant increase in Bcl-2. Thus, our findings suggest that CD34+ HSPCs are an extremely sensitive target for GCs and that cytokines protect these cells from GC-induced apoptosis.
Collapse
Affiliation(s)
- Serap Erdem Kuruca
- Deparment of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Muzaffer Beyza Çetin
- Deparment of Physiology, Cerrahpasa Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Kadriye Akgün Dar
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Dilşad Özerkan
- Department of Genetic and Bioengineering, Faculty of Engineering and Architecture, Kastamonu University, Kastamonu, Turkey.
| |
Collapse
|
73
|
Cohn R, Thakar K, Lowe A, Ladha FA, Pettinato AM, Romano R, Meredith E, Chen YS, Atamanuk K, Huey BD, Hinson JT. A Contraction Stress Model of Hypertrophic Cardiomyopathy due to Sarcomere Mutations. Stem Cell Reports 2018; 12:71-83. [PMID: 30554920 PMCID: PMC6335568 DOI: 10.1016/j.stemcr.2018.11.015] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022] Open
Abstract
Thick-filament sarcomere mutations are a common cause of hypertrophic cardiomyopathy (HCM), a disorder of heart muscle thickening associated with sudden cardiac death and heart failure, with unclear mechanisms. We engineered four isogenic induced pluripotent stem cell (iPSC) models of β-myosin heavy chain and myosin-binding protein C3 mutations, and studied iPSC-derived cardiomyocytes in cardiac microtissue assays that resemble cardiac architecture and biomechanics. All HCM mutations resulted in hypercontractility with prolonged relaxation kinetics in proportion to mutation pathogenicity, but not changes in calcium handling. RNA sequencing and expression studies of HCM models identified p53 activation, oxidative stress, and cytotoxicity induced by metabolic stress that can be reversed by p53 genetic ablation. Our findings implicate hypercontractility as a direct consequence of thick-filament mutations, irrespective of mutation localization, and the p53 pathway as a molecular marker of contraction stress and candidate therapeutic target for HCM patients.
Collapse
Affiliation(s)
- Rachel Cohn
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Ketan Thakar
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Andre Lowe
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Feria A Ladha
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Anthony M Pettinato
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Robert Romano
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Emily Meredith
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA
| | - Yu-Sheng Chen
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA
| | - Katherine Atamanuk
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Bryan D Huey
- Department of Materials Science and Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, 10 Discovery Drive, Farmington, CT 06032, USA; University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT 06032, USA.
| |
Collapse
|
74
|
Chen J, Zhong J, Liu Y, Huang Y, Luo F, Zhou Y, Pan X, Cao S, Zhang L, Zhang Y, Wang J. Purified vitexin compound 1, a new neolignan isolated compound, promotes PUMA-dependent apoptosis in colorectal cancer. Cancer Med 2018; 7:6158-6169. [PMID: 30402948 PMCID: PMC6308053 DOI: 10.1002/cam4.1769] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 07/24/2018] [Accepted: 08/08/2018] [Indexed: 12/11/2022] Open
Abstract
Purified vitexin compound 1 (VB1, a neolignan isolated and extracted from the seed of Chinese herb Vitex negundo) is an effective antitumor agent and exhibits promising clinical activity against various cancers including colorectal cancer. However, it remains unknown about the precise underlying mechanism associated with the antitumor effect of VB1 and how it triggers apoptosis in cancer cells. Here, we demonstrated that VB1 promoted apoptosis via p53-dependent induction of p53 upregulated modulator of apoptosis (PUMA) and further to induce Bax (Bcl-2-associated X protein) activation and mitochondrial dysfunction in colon cancer HCT-116 and LoVo cells. Deficiency in p53, PUMA, or Bax abrogated VB1-induced apoptosis and promoted cell survival in HCT-116 cells. Furthermore, the combination of VB1 with chemotherapeutic drugs 5-fluorouracil (5-FU) or NVP-BZE235 resulted in a synergistic antitumor effect via PUMA induction in HCT-116 cells. VB1 significantly suppressed the cell proliferation of wild-type (WT) HCT-116 and LoVo cells in vitro and tumor growth in vivo. The results indicate that p53/PUMA/Bax axis plays a critical role in VB1-induced apoptosis and VB1 may have valuable clinical applications in cancer therapy as a novel anticancer agent used alone or in combination with other chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jingfei Chen
- Department of Internal MedicineThe Third Xiangya HospitalCentral South UniversityChangshaChina
- College of BiologyHunan UniversityChangshaChina
- Department of Laboratory MedicineXiangya School of MedicineCentral South UniversityChangshaChina
- Department of Obstetrics and GynecologyXiangya HospitalCentral South UniversityChangshaChina
| | | | - Yeying Liu
- Department of Internal MedicineThe Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Yuan Huang
- College of BiologyHunan UniversityChangshaChina
| | - Fei Luo
- Department of CardiologyThe Second Xiangya HospitalChangshaChina
| | - Yingjun Zhou
- School of Pharmaceutical ScienceCentral South UniversityChangshaChina
| | - Xi Pan
- Department of OncologyThe Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Shousong Cao
- Department of PharmacologySchool of PharmacySouthwest Medical UniversityLuzhouChina
| | - Lingling Zhang
- Department of Laboratory MedicineXiangya School of MedicineCentral South UniversityChangshaChina
| | - Yingjie Zhang
- College of BiologyHunan UniversityChangshaChina
- Shenzhen InstituteHunan UniversityShenzhenChina
| | - Jiangang Wang
- Department of Internal MedicineThe Third Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
75
|
An altered gene expression profile in tyramine-exposed intestinal cell cultures supports the genotoxicity of this biogenic amine at dietary concentrations. Sci Rep 2018; 8:17038. [PMID: 30451877 PMCID: PMC6242974 DOI: 10.1038/s41598-018-35125-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 10/31/2018] [Indexed: 01/06/2023] Open
Abstract
Tyramine, histamine and putrescine are the most commonly detected and most abundant biogenic amines (BA) in food. The consumption of food with high concentrations of these BA is discouraged by the main food safety agencies, but legal limits have only been set for histamine. The present work reports a transcriptomic investigation of the oncogenic potential of the above-mentioned BA, as assessed in the HT29 human intestinal epithelial cell line. Tyramine had a greater effect on the expression of genes involved in tumorigenesis than did histamine or putrescine. Since some of the genes that showed altered expression in tyramine-exposed cells are involved in DNA damage and repair, the effect of this BA on the expression of other genes involved in the DNA damage response was investigated. The results suggest that tyramine might be genotoxic for intestinal cells at concentrations easily found in BA-rich food. Moreover, a role in promoting intestinal cancer cannot be excluded.
Collapse
|
76
|
Sun L, Huang Y, Liu Y, Zhao Y, He X, Zhang L, Wang F, Zhang Y. Ipatasertib, a novel Akt inhibitor, induces transcription factor FoxO3a and NF-κB directly regulates PUMA-dependent apoptosis. Cell Death Dis 2018; 9:911. [PMID: 30185800 PMCID: PMC6125489 DOI: 10.1038/s41419-018-0943-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/27/2018] [Accepted: 07/30/2018] [Indexed: 12/27/2022]
Abstract
Colon cancer is one of the three common malignant tumors, with a lower survival rate. Ipatasertib, a novel highly selective ATP-competitive pan-Akt inhibitor, shows a strong antitumor effect in a variety of carcinoma, including colon cancer. However, there is a lack of knowledge about the precise underlying mechanism of clinical therapy for colon cancer. We conducted this study to determine that ipatasertib prevented colon cancer growth through PUMA-dependent apoptosis. Ipatasertib led to p53-independent PUMA activation by inhibiting Akt, thereby activating both FoxO3a and NF-κB synchronously that will directly bind to PUMA promoter, up-regulating PUMA transcription and Bax-mediated intrinsic mitochondrial apoptosis. Remarkably, Akt/FoxO3a/PUMA is the major pathway while Akt/NF-κB/PUMA is the secondary pathway of PUMA activation induced by ipatasertib in colon cancer. Knocking out PUMA eliminated ipatasertib-induced apoptosis both in vitro and in vivo (xenografts). Furthermore, PUMA is also indispensable in combinational therapies of ipatasertib with some conventional or novel drugs. Collectively, our study demonstrated that PUMA induction by FoxO3a and NF-κB is a critical step to suppress the growth of colon cancer under the therapy with ipatasertib, which provides some theoretical basis for clinical assessment.
Collapse
Affiliation(s)
- Li Sun
- College of Biology, Hunan University, Changsha, 410082, China.,Department of Out-patient, Affiliated Hospital of Hebei University of Engineering, Handan, 056002, China
| | - Yuan Huang
- College of Biology, Hunan University, Changsha, 410082, China
| | - Yeying Liu
- College of Biology, Hunan University, Changsha, 410082, China
| | - Yujie Zhao
- College of Biology, Hunan University, Changsha, 410082, China
| | - Xiaoxiao He
- College of Biology, Hunan University, Changsha, 410082, China
| | - Lingling Zhang
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, 410013, China.
| | - Feng Wang
- Department of Gastroenterology, The Tenth People's Hospital of Shanghai, Tongji University, Shanghai, 200072, China. .,Shenzhen Institute, Hunan University, Shenzhen, China.
| | - Yingjie Zhang
- College of Biology, Hunan University, Changsha, 410082, China. .,Shenzhen Institute, Hunan University, Shenzhen, China.
| |
Collapse
|
77
|
Zhang QA, Yang XH, Chen D, Yan X, Jing FC, Liu HQ, Zhang R. miR-34 increases in vitro PANC-1 cell sensitivity to gemcitabine via targeting Slug/PUMA. Cancer Biomark 2018; 21:755-762. [PMID: 29355113 DOI: 10.3233/cbm-170289] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
miR-34 was deregulated in tumor tissues compared with corresponding noncancerous tissue samples. Furthermore, miR-34 may contribute to cancer-stromal interaction associated with cancer progression. However, whether miR-34 could decrease chemoresistance of cancer cells to chemotherapeutic agent remains unclear. In our study, we examined whether overexpression of miR-34 could sensitize gemcitabine -mediated apoptosis in human pancreatic cancer PANC-1 cells. We found that miR-34 markedly induced gemcitabine -mediated apoptosis in PANC-1 cells. miR-34 induced down-regulation of Slug expression and upregulation of p53 up-regulated modulator of apoptosis (PUMA) expression. The over-expression of Slug or downregulation of PUMA by Slug cDNA or PUMA siRNA transfection markedly blocked miR-34-induced gemcitabine sensitization. Furthermore, miR-34 induced PUMA expression by downregulation of Slug. Taken together, our study demonstrates that miR-34 enhances sensitization against gemcitabine-mediated apoptosis through the down-regulation of Slug expression, and up-regulation of Slug-dependent PUMA expression.
Collapse
Affiliation(s)
- Qing-An Zhang
- Department of Clinical Laboratory, The Central Hospital of Linyi, Yishui, Shandong, China.,Department of Clinical Laboratory, The Central Hospital of Linyi, Yishui, Shandong, China
| | - Xu-Hai Yang
- Department of Oncology, Yantaiyuhuangding Hospital, Yantai, Shandong, China
| | - Dong Chen
- Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao,Shandong, China
| | - Xiang Yan
- Department of Oncology, Yantaiyuhuangding Hospital, Yantai, Shandong, China
| | - Fu-Chun Jing
- Department of Gastroenterology, Taian Central Hospital, Taian, Shandong, China
| | - Hong-Qian Liu
- Department Pharmacy, The Central Hospital of Linyi, Yishui, Shandong, China
| | - Ronghua Zhang
- Department of Clinical Laboratory, The Central Hospital of Linyi, Yishui, Shandong, China
| |
Collapse
|
78
|
Cromer MK, Vaidyanathan S, Ryan DE, Curry B, Lucas AB, Camarena J, Kaushik M, Hay SR, Martin RM, Steinfeld I, Bak RO, Dever DP, Hendel A, Bruhn L, Porteus MH. Global Transcriptional Response to CRISPR/Cas9-AAV6-Based Genome Editing in CD34 + Hematopoietic Stem and Progenitor Cells. Mol Ther 2018; 26:2431-2442. [PMID: 30005866 DOI: 10.1016/j.ymthe.2018.06.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/30/2018] [Accepted: 06/01/2018] [Indexed: 12/22/2022] Open
Abstract
Genome-editing technologies are currently being translated to the clinic. However, cellular effects of the editing machinery have yet to be fully elucidated. Here, we performed global microarray-based gene expression measurements on human CD34+ hematopoietic stem and progenitor cells that underwent editing. We probed effects of the entire editing process as well as each component individually, including electroporation, Cas9 (mRNA or protein) with chemically modified sgRNA, and AAV6 transduction. We identified differentially expressed genes relative to control treatments, which displayed enrichment for particular biological processes. All editing machinery components elicited immune, stress, and apoptotic responses. Cas9 mRNA invoked the greatest amount of transcriptional change, eliciting a distinct viral response and global transcriptional downregulation, particularly of metabolic and cell cycle processes. Electroporation also induced significant transcriptional change, with notable downregulation of metabolic processes. Surprisingly, AAV6 evoked no detectable viral response. We also found Cas9/sgRNA ribonucleoprotein treatment to be well tolerated, in spite of eliciting a DNA damage signature. Overall, this data establishes a benchmark for cellular tolerance of CRISPR/Cas9-AAV6-based genome editing, ensuring that the clinical protocol is as safe and efficient as possible.
Collapse
Affiliation(s)
- M Kyle Cromer
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | | | | | - Bo Curry
- Agilent Technologies, Santa Clara, CA 95051, USA
| | | | - Joab Camarena
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Milan Kaushik
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Sarah R Hay
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Renata M Martin
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | | | - Rasmus O Bak
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Daniel P Dever
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA
| | - Ayal Hendel
- Institute for Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan 5290002, Israel.
| | | | - Matthew H Porteus
- Department of Pediatrics, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
79
|
Jiang J, Shi Y, Yu R, Chen L, Zhao X. Biological response of zebrafish after short-term exposure to azoxystrobin. CHEMOSPHERE 2018; 202:56-64. [PMID: 29554508 DOI: 10.1016/j.chemosphere.2018.03.055] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 06/08/2023]
Abstract
Azoxystrobin (AZ) is a broad-spectrum systemic fungicide that widely used in the world. The present study investigated the toxicity effects on zebrafish after short-term exposure of AZ. Results demonstrated that the larval stage was most susceptible to AZ in the multiple life stages of zebrafish, with 96 h-LC50 value of 0.777 mg/L. Zebrafish larvae were exposed to different AZ concentrations (0, 0.1, 1, 10, 100 μg/L) and examined on 24, 48 and 72 h. It was found that AZ induced ROS accumulation, increased GST, GPX and POD activity and the transcriptions of antioxidant and stress response related genes, while the opposite trend occurred for SOD and CAT activity in 24-h or 48-h exposure period. The increased E2 and VTG levels in zebrafish larvae, and altered transcription levels of regulatory and steroidogenic genes in the hypothalamus-pituitary-gonad (HPG) axis indicated the endocrine disruption capacity of AZ. The transcripts of mdm2, p53, ogg1, bcl2, bbc3, cas8 and cas9 involved in cell apoptosis, and the mRNA levels of cytokines and chemokines such as cxcl-c1c, ccl, il-1β, il-8, ifn, and tnfα were in accordance with the trends of the examined genes involved in oxidative stress and endocrine system. The results suggested that short-term exposure to AZ might impose ecotoxicological effects on zebrafish larvae, and the information presented here also provide molecular strategies and increase mechanistic understanding of AZ-induced toxic response, and help elucidate the environmental risks of AZ.
Collapse
Affiliation(s)
- Jinhua Jiang
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Hangzhou, 310021, Zhejiang, China
| | - Yan Shi
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Hangzhou, 310021, Zhejiang, China
| | - Ruixian Yu
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Hangzhou, 310021, Zhejiang, China
| | - Liping Chen
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Hangzhou, 310021, Zhejiang, China
| | - Xueping Zhao
- Institute of Quality and Standard for Agro-products, Zhejiang Academy of Agricultural Sciences, State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Hangzhou, 310021, Zhejiang, China.
| |
Collapse
|
80
|
LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. Hemasphere 2018; 2:e54. [PMID: 31723778 PMCID: PMC6745991 DOI: 10.1097/hs9.0000000000000054] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022] Open
Abstract
Supplemental Digital Content is available in the text Despite recent advances, the myeloproliferative neoplasms (MPNs) are attended by considerable morbidity and mortality. Janus kinase (Jak) inhibitors such as ruxolitinib manage symptoms but do not substantially change the natural history of the disease. In this report, we show the effects of IMG-7289, an irreversible inhibitor of the epigenetically active lysine-specific demethylase 1 (LSD1) in mouse models of MPN. Once-daily treatment with IMG-7289 normalized or improved blood cell counts, reduced spleen volumes, restored normal splenic architecture, and reduced bone marrow fibrosis. Most importantly, LSD1 inhibition lowered mutant allele burden and improved survival. IMG-7289 selectively inhibited proliferation and induced apoptosis of JAK2V617F cells by concomitantly increasing expression and methylation of p53, and, independently, the pro-apoptotic factor PUMA and by decreasing the levels of its antiapoptotic antagonist BCLXL. These data provide a molecular understanding of the disease-modifying activity of the LSD1 inhibitor IMG-7289 that is currently undergoing clinical evaluation in patients with high-risk myelofibrosis. Moreover, low doses of IMG-7289 and ruxolitinib synergize in normalizing the MPN phenotype in mice, offering a rationale for investigating combination therapy.
Collapse
|
81
|
Zhou K, Zhang W, Zhang Q, Gui R, Zhao H, Chai X, Li Y, Wei X, Song Y. Loss of thyroid hormone receptor interactor 13 inhibits cell proliferation and survival in human chronic lymphocytic leukemia. Oncotarget 2018; 8:25469-25481. [PMID: 28424416 PMCID: PMC5421944 DOI: 10.18632/oncotarget.16038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
Background The genetic regulation of apoptosis and cell proliferation plays a role in the growth of chronic lymphocytic leukemia (CLL), the most common form of leukemia in the Western hemisphere. Although thyroid hormone receptor interactors (TRIPs) are known to play roles in cell cycle, the potential involvement of the novel family member TRIP13 in CLL has not yet been investigated. Methods Quantitative PCR (qPCR) was used to detect expression of TRIP13 in 36 CLL patients and 33 healthy donors CD19+ B cells. Loss-of-function (siRNA) assays were used to alter TRIP13 expression levels. The effect of TRIP13 on cell proliferation and apoptosis was measured by MTT, Annexin V-based flow cytometry and Caspase 3/7 activity assay. Affymetrix GeneChip and Ingenuity Pathway Analysis (IPA) were used to describe an overview of TRIP13 potential biological function and downstream pathways. Dual-luciferase reporter assay was performed to assess the promoting effect of c-MYC on TRIP13 transcription. RESULTS The qPCR data showed that TRIP13 is significantly over-expressed in CLL patients. Microarray analyses indicated that the biological function of TRIP13 in CLL is majorly cell apoptosis and cell proliferation associated. TRIP13 siRNA expressing cells exhibited a slower cell proliferation rate and underwent apoptosis compared with control cells. TRIP13 knockdown induced CLL cells apoptosis through PUMA independent of p53. TRIP13 up-regulation is induced by c-MYC dependent transcriptional activation. Conclusion Overall, our data suggest the bio-function of TRIP13 in CLL cell for the first time, and that this gene might be a therapeutic target for CLL.
Collapse
Affiliation(s)
- Keshu Zhou
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Wentao Zhang
- Armed Police Forces Hospital of Henan, People's Republic of China
| | - Qing Zhang
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Ruirui Gui
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Huifang Zhao
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Xiaofei Chai
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Yufu Li
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Xudong Wei
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| | - Yongping Song
- Department of Hematology, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, People's Republic of China
| |
Collapse
|
82
|
Zhang L, Wang H, Li W, Zhong J, Yu R, Huang X, Wang H, Tan Z, Wang J, Zhang Y. Pazopanib, a novel multi-kinase inhibitor, shows potent antitumor activity in colon cancer through PUMA-mediated apoptosis. Oncotarget 2018; 8:3289-3303. [PMID: 27924057 PMCID: PMC5356882 DOI: 10.18632/oncotarget.13753] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Accepted: 10/19/2016] [Indexed: 12/22/2022] Open
Abstract
Colon cancer is still the third most common cancer which has a high mortality but low five-year survival rate. Novel tyrosine kinase inhibitors (TKI) such as pazopanib become effective antineoplastic agents that show promising clinical activity in a variety of carcinoma, including colon cancer. However, the precise underlying mechanism against tumor is unclear. Here, we demonstrated that pazopanib promoted colon cancer cell apoptosis through inducing PUMA expression. Pazopanib induced p53-independent PUMA activation by inhibiting PI3K/Akt signaling pathway, thereby activating Foxo3a, which subsequently bound to the promoter of PUMA to activate its transcription. After induction, PUMA activated Bax and triggered the intrinsic mitochondrial apoptosis pathway. Furthermore, administration of pazopanib highly suppressed tumor growth in a xenograft model. PUMA deletion in cells and tumors led to resistance of pazopanib, indicating PUMA-mediated pro-apoptotic and anti-tumor effects in vitro and in vivo. Combing pazopanib with some conventional or novel drugs, produced heightened and synergistic antitumor effects that were associated with potentiated PUMA induction via different pathways. Taken together, these results establish a critical role of PUMA in mediating the anticancer effects of pazopanib in colon cancer cells and provide the rationale for clinical evaluation.
Collapse
Affiliation(s)
- Lingling Zhang
- College of Biology, Hunan University, Changsha, China.,Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China.,Department of Internal Medicine, The Third Xiangya Hospital, Central South University, Changsha, China.,School of Life Sciences, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Huanan Wang
- College of Biology, Hunan University, Changsha, China.,Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei Li
- Department of Laboratory Medicine, Xiangya School of Medicine, Central South University, Changsha, China.,Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Juchang Zhong
- College of Biology, Hunan University, Changsha, China
| | - Rongcheng Yu
- College of Biology, Hunan University, Changsha, China
| | - Xinfeng Huang
- College of Biology, Hunan University, Changsha, China
| | - Honghui Wang
- College of Biology, Hunan University, Changsha, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, China
| | - Jiangang Wang
- Department of Internal Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yingjie Zhang
- College of Biology, Hunan University, Changsha, China.,Shenzhen Institute, Hunan University, Shenzhen, China
| |
Collapse
|
83
|
Sladky V, Schuler F, Fava LL, Villunger A. The resurrection of the PIDDosome - emerging roles in the DNA-damage response and centrosome surveillance. J Cell Sci 2018; 130:3779-3787. [PMID: 29142064 DOI: 10.1242/jcs.203448] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The PIDDosome is often used as the alias for a multi-protein complex that includes the p53-induced death domain protein 1 (PIDD1), the bipartite linker protein CRADD (also known as RAIDD) and the pro-form of an endopeptidase belonging to the caspase family, i.e. caspase-2. Yet, PIDD1 variants can also interact with a number of other proteins that include RIPK1 (also known as RIP1) and IKBKG (also known as NEMO), PCNA and RFC5, as well as nucleolar components such as NPM1 or NCL. This promiscuity in protein binding is facilitated mainly by autoprocessing of the full-length protein into various fragments that contain different structural domains. As a result, multiple responses can be mediated by protein complexes that contain a PIDD1 domain. This suggests that PIDD1 acts as an integrator for multiple types of stress that need instant attention. Examples are various types of DNA lesion but also the presence of extra centrosomes that can foster aneuploidy and, ultimately, promote DNA damage. Here, we review the role of PIDD1 in response to DNA damage and also highlight novel functions of PIDD1, such as in centrosome surveillance and scheduled polyploidisation as part of a cellular differentiation program during organogenesis.
Collapse
Affiliation(s)
- Valentina Sladky
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Fabian Schuler
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| | - Luca L Fava
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria.,Center for Integrative Biology (CIBIO), University of Trento, Via Sommarive 9, 38123 Povo, Italy
| | - Andreas Villunger
- Division of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80, 6020 Innsbruck, Austria
| |
Collapse
|
84
|
The regulation of the mitochondrial apoptotic pathway by glucocorticoid receptor in collaboration with Bcl-2 family proteins in developing T cells. Apoptosis 2018; 22:239-253. [PMID: 27888447 PMCID: PMC5306359 DOI: 10.1007/s10495-016-1320-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Glucocorticoids (GC) are important in the regulation of selection and apoptosis of CD4+CD8+ double-positive (DP) thymocytes. The pronounced GC-sensitivity of DP thymocytes, observed earlier, might be due to the combination of classical (genomic) and alternative (non-genomic) glucocorticoid receptor (GR) signaling events modifying activation or apoptotic pathways. In particular, the previously demonstrated mitochondrial translocation of activated GR in DP thymocytes offered a fascinating explanation for their pronounced GC-induced apoptosis sensitivity. However, the fine molecular details how the mitochondrial translocation of GR might regulate apoptosis remained unclear. Therefore, in the present study, we intended to examine which apoptotic pathways could be involved in GC-induced thymocyte apoptosis. Furthermore we investigated the potential relationship between the GR and Bcl-2 proteins. Using an in vitro test system, thymocytes from 4-week-old BALB/c mice, were treated with the GC-analogue dexamethasone (DX). Bax accumulated in mitochondria upon DX treatment. Mitochondrial GR showed association with members of the Bcl-2 family: Bak, Bim, Bcl-xL. Elevated Cytochrome C, and active caspase-3, -8, and -9 levels were detected in thymocytes after DX treatment. These results support the hypothesis that in early phases of GC-induced thymocyte apoptosis, the mitochondrial pathway plays a crucial role, confirmed by the release of Cytochrome C and the activation of caspase-9. The activation of caspase-8 was presumably due to cross-talk between apoptotic signaling pathways. We propose that the GC-induced mitochondrial accumulation of Bax and the interaction between the GR and Bim, Bcl-xL and Bak could play a role in the regulation of thymocyte apoptosis.
Collapse
|
85
|
miR-663 sustains NSCLC by inhibiting mitochondrial outer membrane permeabilization (MOMP) through PUMA/BBC3 and BTG2. Cell Death Dis 2018; 9:49. [PMID: 29352138 PMCID: PMC5833438 DOI: 10.1038/s41419-017-0080-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/10/2017] [Accepted: 10/17/2017] [Indexed: 02/06/2023]
Abstract
Treatment of lung cancer is an unmet need as it accounts for the majority of cancer deaths worldwide. The development of new therapies urges the identification of potential targets. MicroRNAs' expression is often deregulated in cancer and their modulation has been proposed as a successful strategy to interfere with tumor cell growth and spread. We recently reported on an unbiased high-content approach to identify miRNAs regulating cell proliferation and tumorigenesis in non-small cell lung cancer (NSCLC). Here we studied the oncogenic role of miR-663 in NSCLC biology and analyzed the therapeutic potential of miR-663 targeting. We found that miR-663 regulates apoptosis by controlling mitochondrial outer membrane permeabilization (MOMP) through the expression of two novel direct targets PUMA/BBC3 and BTG2. Specifically, upon miR-663 knockdown the BH3-only protein PUMA/BBC3 directly activates mitochondrial depolarization and cell death, while BTG2 accumulation further enhances this effect by triggering p53 mitochondrial localization. Moreover, we show that miR-663 depletion is sufficient to elicit cell death in NSCLC cells and to impair tumor growth in vivo.
Collapse
|
86
|
Bhat P, Kriel J, Shubha Priya B, Basappa, Shivananju NS, Loos B. Modulating autophagy in cancer therapy: Advancements and challenges for cancer cell death sensitization. Biochem Pharmacol 2017; 147:170-182. [PMID: 29203368 DOI: 10.1016/j.bcp.2017.11.021] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023]
Abstract
Autophagy is a major protein degradation pathway capable of upholding cellular metabolism under nutrient limiting conditions, making it a valuable resource to highly proliferating tumour cells. Although the regulatory machinery of the autophagic pathway has been well characterized, accurate modulation of this pathway remains complex in the context of clinical translatability for improved cancer therapies. In particular, the dynamic relationship between the rate of protein degradation through autophagy, i.e. autophagic flux, and the susceptibility of tumours to undergo apoptosis remains largely unclear. Adding to inefficient clinical translation is the lack of measurement techniques that accurately depict autophagic flux. Paradoxically, both increased autophagic flux as well as autophagy inhibition have been shown to sensitize cancer cells to undergo cell death, indicating the highly context dependent nature of this pathway. In this article, we aim to disentangle the role of autophagy modulation in tumour suppression by assessing existing literature in the context of autophagic flux and cellular metabolism at the interface of mitochondrial function. We highlight the urgency to not only assess autophagic flux more accurately, but also to center autophagy manipulation within the unique and inherent metabolic properties of cancer cells. Lastly, we discuss the challenges faced when targeting autophagy in the clinical setting. In doing so, it is hoped that a better understanding of autophagy in cancer therapy is revealed in order to overcome tumour chemoresistance through more controlled autophagy modulation in the future.
Collapse
Affiliation(s)
- Punya Bhat
- DOS in Chemistry, University of Mysore, Manasgangotri, Mysuru 570006, Karnataka, India
| | - Jurgen Kriel
- Department of Physiological Sciences, Faculty of Science, University of Stellenbosch, Stellenbosch 7600, South Africa
| | - Babu Shubha Priya
- DOS in Chemistry, University of Mysore, Manasgangotri, Mysuru 570006, Karnataka, India
| | - Basappa
- Laboratory of Chemical Biology, Department of studies in Organic Chemistry, Manasagangotri, University of Mysore, Mysore 570006, India
| | - Nanjunda Swamy Shivananju
- Department of Biotechnology, Sri Jayachamarajendra College of Engineering, JSS Science and Technology University, JSS TEI Campus, Mysuru 57006, Karnataka, India.
| | - Ben Loos
- Department of Physiological Sciences, Faculty of Science, University of Stellenbosch, Stellenbosch 7600, South Africa.
| |
Collapse
|
87
|
Abstract
Apoptosis is often deregulated in a number of human diseases. Heat-induced apoptosis is a model system for studying the consequences of protein misfolding and is mediated by the Bcl-2 family of proteins. This family consists of both pro-apoptotic and anti-apoptotic members that control mitochondrial integrity. The BH3-only pro-apoptotic members are strong inducers of apoptotic cell death. Protein damaging stress can activate a process of cellular destruction known as apoptosis. The pro-apoptotic BH3-only proteins and transcription factors activate this death pathway by inhibiting the anti-apoptotic Bcl-2 family proteins eliminating cancer cells in a short period of time.
Collapse
Affiliation(s)
- Rabih Roufayel
- a Department of Science , American University of the Middle East , Egaila , Kuwait
| |
Collapse
|
88
|
Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ 2017; 25:104-113. [PMID: 29149101 DOI: 10.1038/cdd.2017.169] [Citation(s) in RCA: 901] [Impact Index Per Article: 112.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/05/2017] [Accepted: 09/08/2017] [Indexed: 01/10/2023] Open
Abstract
The tumour suppressor gene TP53 is mutated in ~50% of human cancers. In addition to its function in tumour suppression, p53 also plays a major role in the response of malignant as well as nontransformed cells to many anticancer therapeutics, particularly those that cause DNA damage. P53 forms a homotetrameric transcription factor that is reported to directly regulate ~500 target genes, thereby controlling a broad range of cellular processes, including cell cycle arrest, cell senescence, DNA repair, metabolic adaptation and cell death. For a long time, induction of apoptotic death in nascent neoplastic cells was regarded as the principal mechanism by which p53 prevents tumour development. This concept has, however, recently been challenged by the findings that in striking contrast to Trp53-deficient mice, gene-targeted mice that lack the critical effectors of p53-induced apoptosis do not develop tumours spontaneously. Remarkably, even mice lacking all mediators critical for p53-induced apoptosis, G1/S boundary cell cycle arrest and cell senescence do not develop any tumours spontaneously. In this review we discuss current understanding of the mechanisms by which p53 induces cell death and how this affects p53-mediated tumour suppression and the response of malignant cells to anticancer therapy.
Collapse
Affiliation(s)
- Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Ana Janic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Marco J Herold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
89
|
Hu B, Huo Y, Yang L, Chen G, Luo M, Yang J, Zhou J. ZIKV infection effects changes in gene splicing, isoform composition and lncRNA expression in human neural progenitor cells. Virol J 2017; 14:217. [PMID: 29116029 PMCID: PMC5688814 DOI: 10.1186/s12985-017-0882-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 10/30/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The Zika virus (ZIKV) is a mosquito-borne flavivirus that causes microcephaly and Guillain-Barré syndrome in infected individuals. To obtain insights into the mechanism of ZIKV infection and pathogenesis, we analyzed the transcriptome of ZIKV infected human neural progenitor cells (hNPCs) for changes in alternative splicing (AS), gene isoform (ISO) composition and long noncoding RNAs (lncRNAs) expression. METHODS We analyzed differentially expressed lncRNAs, AS, ISO from RNA-seq data in ZIKV infected hNPCs. RESULTS We obtained 149 differentially expressed lncRNAs, including potential viral targets to modulate cellular processes such as cell cycle, apoptosis and immune response. The infection induced 262 cases of AS occurring in 229 genes, which were enriched in cell death, RNA processing, transport, and neuron development. Among 691 differentially expressed ISOs, upregulated ISOs were enriched in signaling, regulation of transcription, and amino acid biosynthesis, while downregulated ISOs were mostly enriched in cell cycle. Importantly, these analyses revealed specific links between ZIKV induced changes in cellular pathways and the type of changes in the host transcriptome, suggesting important regulatory mechanisms. CONCLUSIONS Our analyses revealed candidate lncRNAs, AS events and ISOs which may function in ZIKV infection induced cell cycle disruption, apoptosis and attenuation of neurogenesis, and shed light on the roles of lncRNAs, AS and ISOs in virus-host interactions, and would facilitate future studies of ZIKV infection and pathogenesis.
Collapse
Affiliation(s)
- Benxia Hu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, 650204, China
| | - Yongxia Huo
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, China
| | - Liping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, China
| | - Guijun Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, China
| | - Minhua Luo
- State Key Laboratory of Virology, CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Wuhan Institute of Virology, Wuhan, 430071, China
| | - Jinlong Yang
- BGI-Yunnan, BGI-Shenzhen, Kunming, 650000, China.,College of Life Sciences, Yunnan University, Kunming, 650091, China
| | - Jumin Zhou
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, China.
| |
Collapse
|
90
|
Xu K, Chen G, Qiu Y, Yuan Z, Li H, Yuan X, Sun J, Xu J, Liang X, Yin P. miR-503-5p confers drug resistance by targeting PUMA in colorectal carcinoma. Oncotarget 2017; 8:21719-21732. [PMID: 28423513 PMCID: PMC5400618 DOI: 10.18632/oncotarget.15559] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 01/22/2017] [Indexed: 01/04/2023] Open
Abstract
The development of multidrug-resistance (MDR) is a major contributor to death in colorectal carcinoma (CRC). Here, we investigated the possible role of microRNA (miR)-503-5p in drug resistant CRC cells. Unbiased microRNA array screening revealed that miR-503-5p is up-regulated in two oxaliplatin (OXA)-resistant CRC cell lines. Overexpression of miR-503-5p conferred resistance to OXA-induced apoptosis and inhibition of tumor growth in vitro and in vivo through down-regulation of PUMA expression. miR-503-5p knockdown sensitized chemoresistant CRC cells to OXA. Our studies indicated that p53 suppresses miR-503-5p expression and that deletion of p53 upregulates miR-503-5p expression. Inhibition of miR-503-5p in p53 null cells increased their sensitivity to OXA treatment. Importantly, analysis of patient samples showed that expression of miR-503-5p negatively correlates with PUMA in CRC. These results indicate that a p53/miR-503-5p/PUMA signaling axis regulates the CRC response to chemotherapy, and suggest that miR-503-5p plays an important role in the development of MDR in CRC by modulating PUMA expression.
Collapse
Affiliation(s)
- Ke Xu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.,Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China
| | - Guo Chen
- Department of Radiation Oncology, School of Medicine and Winship Cancer Institute of Emory University, Atlanta, Georgia 30322, USA
| | - Yanyan Qiu
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.,Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China.,Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China State
| | - Zeting Yuan
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Hongchang Li
- Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China State
| | - Xia Yuan
- Department of Pharmacy, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China
| | - Jian Sun
- Central Laboratory, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China.,Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China
| | - Jianhua Xu
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, PR China
| | - Peihao Yin
- Interventional Cancer Institute of Chinese Integrative Medicine, Shanghai University of Traditional Medicine, Shanghai 200062, PR China.,Department of General Surgery, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, PR China State
| |
Collapse
|
91
|
Li KP, Shanmuganad S, Carroll K, Katz JD, Jordan MB, Hildeman DA. Dying to protect: cell death and the control of T-cell homeostasis. Immunol Rev 2017; 277:21-43. [PMID: 28462527 PMCID: PMC5416827 DOI: 10.1111/imr.12538] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 02/23/2017] [Accepted: 02/26/2017] [Indexed: 02/07/2023]
Abstract
T cells play a critical role in immune responses as they specifically recognize peptide/MHC complexes with their T-cell receptors and initiate adaptive immune responses. While T cells are critical for performing appropriate effector functions and maintaining immune memory, they also can cause autoimmunity or neoplasia if misdirected or dysregulated. Thus, T cells must be tightly regulated from their development onward. Maintenance of appropriate T-cell homeostasis is essential to promote protective immunity and limit autoimmunity and neoplasia. This review will focus on the role of cell death in maintenance of T-cell homeostasis and outline novel therapeutic strategies tailored to manipulate cell death to limit T-cell survival (eg, autoimmunity and transplantation) or enhance T-cell survival (eg, vaccination and immune deficiency).
Collapse
Affiliation(s)
- Kun-Po Li
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Sharmila Shanmuganad
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Kaitlin Carroll
- Immunology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Jonathan D. Katz
- Division of Immunobiology, Cincinnati, OH 45229, USA
- Division of Endocrinology, Diabetes Research Center, Cincinnati, OH 45229, USA
| | - Michael B. Jordan
- Division of Immunobiology, Cincinnati, OH 45229, USA
- Division of Bone Marrow Transplantation and Immune Deficiency, Department of Pediatrics, Cincinnati Children’s Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | | |
Collapse
|
92
|
Shan Z, Liu Q, Li Y, Wu J, Sun D, Gao Z. PUMA decreases the growth of prostate cancer PC-3 cells independent of p53. Oncol Lett 2017; 13:1885-1890. [PMID: 28454339 DOI: 10.3892/ol.2017.5657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/15/2016] [Indexed: 12/18/2022] Open
Abstract
PUMA (p53 upregulated modulator of apoptosis), a member of the B-cell lymphoma 2 (Bcl-2) protein family, is a pro-apoptotic protein. PUMA expression is modulated by the tumor suppressor p53. PUMA has a role in rapid cell death via p53-dependent and -independent mechanisms. To evaluate whether p53 is required for PUMA-mediated apoptosis in prostate cancer cells, p53 protein was silenced in human prostate cancer PC-3 cells by using p53 small interfering RNA (siRNA). The interference efficiency of p53 on RNA and protein levels was detected by reverse transcription-quantitative polymerase chain reaction and western blotting. Cell proliferation and p21 expression were subsequently examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and western blot analysis, respectively. p53-silenced or control PC-3 cells were transfected with pCEP4-(hemagglutinin)-PUMA plasmid, or non-carrier plasmid. Enzyme-linked immunosorbent assay was used to determine cell apoptosis by measuring histone release and caspase-3 activation, and MTT assay was used to measure cell viability. In addition, the expression of pro-apoptosis protein Bax and anti-apoptosis protein Bcl-2 were evaluated. The results of the present study revealed that p53 siRNA significantly suppressed p53 RNA and protein expression in PC-3 cells. Deficiency of p53 increased the cell growth rate and decreased p21 expression. However, PUMA overexpression remained able to induce apoptosis in p53-silenced and control cells by increasing Bax expression and decreasing Bcl-2 expression, leading to the activation of caspase-3. These results suggest that PUMA may mediate apoptosis of prostate cancer PC-3 cells, potentially independently of p53. Furthermore, PUMA gene treatment to induce cancer cell apoptosis may be more efficient compared with p53-dependent apoptosis, where loss of p53 expression or function may lead to limited efficacy of PUMA expression. Therefore, the present study proposes the significant hypothesis that increasing PUMA expression may be an effective approach for the treatment of prostate cancer, regardless of p53 status.
Collapse
Affiliation(s)
- Zhengfei Shan
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Qingzuo Liu
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Yuling Li
- Department of Pathophysiology, Binzhou Medical University, Yantai, Shandong 264000, P.R. China
| | - Jitao Wu
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Dekang Sun
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Zhenli Gao
- Department of Urology, Yantai Yuhuangding Hospital Affiliated to Medical College of Qingdao University, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
93
|
PUMA and NF-kB Are Cell Signaling Predictors of Reovirus Oncolysis of Breast Cancer. PLoS One 2017; 12:e0168233. [PMID: 28099441 PMCID: PMC5243128 DOI: 10.1371/journal.pone.0168233] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/28/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND PURPOSE Reovirus is a ubiquitous RNA virus that exploits aberrant signaling pathways for its replication. The oncolytic potential of reovirus against numerous cancers under pre-clinical/clinical conditions has been documented by us and others. Despite its proven clinical activity, the underlying mechanisms of reovirus oncolysis is still not well elucidated. If reovirus therapy is to be optimized for cancer, including breast cancer patients, it is imperative to understand the mechanisms of reovirus oncolysis, especially in treatment of resistant tumour. EXPERIMENTAL APPROACH AND RESULTS In the present study global gene expression profiling was utilized as a preliminary roadmap to tease-out pivotal molecules involved in reovirus induced apoptosis in breast cancer. Reovirus treated HTB133 and MCF7 breast cancer cells revealed transcriptional alteration of a defined subset of apoptotic genes and members of the nuclear factor-kappa B (NF-kB) family and p53 upregulated modulator of apoptosis (PUMA) were prominent. Since NF-kB can paradoxically suppress or promote apoptosis in cancer, the significance of NF-kB in reovirus oncolysis of breast cancer was investigated. Real time PCR analysis indicated a 2.9-4.3 fold increase in NF-kB p65 message levels following reovirus infection of MCF7 and HTB133, respectively. Nuclear translocation of NF-kB p65 protein was also dramatically augmented post reovirus treatment and correlated with enhanced DNA binding. Pharmacologic inhibition of NF-kB lead to oncolytic protection and significant down regulation of PUMA message levels. PUMA down regulation using siRNA suppressed reovirus oncolysis via significantly repressed apoptosis in p53 mutant HTB133 cells. CONCLUSIONS This study demonstrates for the first time that a prominent pathway of reovirus oncolysis of breast cancer is mediated through NF-kB and that PUMA upregulation is dependent on NF-kB activation. These findings represent potential therapeutic indicators of reovirus treatment in future clinical trials.
Collapse
|
94
|
Tang HM, Talbot CC, Fung MC, Tang HL. Molecular signature of anastasis for reversal of apoptosis. F1000Res 2017; 6:43. [PMID: 28299189 DOI: 10.12688/f1000research.10568.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/10/2017] [Indexed: 12/22/2022] Open
Abstract
Anastasis (Greek for "rising to life") is a cell recovery phenomenon that rescues dying cells from the brink of cell death. We recently discovered anastasis to occur after the execution-stage of apoptosis in vitro and in vivo. Promoting anastasis could in principle preserve injured cells that are difficult to replace, such as cardiomyocytes and neurons. Conversely, arresting anastasis in dying cancer cells after cancer therapies could improve treatment efficacy. To develop new therapies that promote or inhibit anastasis, it is essential to identify the key regulators and mediators of anastasis - the therapeutic targets. Therefore, we performed time-course microarray analysis to explore the molecular mechanisms of anastasis during reversal of ethanol-induced apoptosis in mouse primary liver cells. We found striking changes in transcription of genes involved in multiple pathways, including early activation of pro-cell survival, anti-oxidation, cell cycle arrest, histone modification, DNA-damage and stress-inducible responses, and at delayed times, angiogenesis and cell migration. Validation with RT-PCR confirmed similar changes in the human liver cancer cell line, HepG2, during anastasis. Here, we present the time-course whole-genome gene expression dataset revealing gene expression profiles during the reversal of apoptosis. This dataset provides important insights into the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA
| |
Collapse
|
95
|
Abstract
Anastasis (Greek for "rising to life") is a cell recovery phenomenon that rescues dying cells from the brink of cell death. We recently discovered anastasis to occur after the execution-stage of apoptosis
in vitro and
in vivo. Promoting anastasis could in principle preserve injured cells that are difficult to replace, such as cardiomyocytes and neurons. Conversely, arresting anastasis in dying cancer cells after cancer therapies could improve treatment efficacy. To develop new therapies that promote or inhibit anastasis, it is essential to identify the key regulators and mediators of anastasis – the therapeutic targets. Therefore, we performed time-course microarray analysis to explore the molecular mechanisms of anastasis during reversal of ethanol-induced apoptosis in mouse primary liver cells. We found striking changes in transcription of genes involved in multiple pathways, including early activation of pro-cell survival, anti-oxidation, cell cycle arrest, histone modification, DNA-damage and stress-inducible responses, and at delayed times, angiogenesis and cell migration. Validation with RT-PCR confirmed similar changes in the human liver cancer cell line, HepG2, during anastasis. Here, we present the time-course whole-genome gene expression dataset revealing gene expression profiles during the reversal of apoptosis. This dataset provides important insights into the physiological, pathological, and therapeutic implications of anastasis.
Collapse
Affiliation(s)
- Ho Man Tang
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - C Conover Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Ming Chiu Fung
- School of Life Sciences, Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ho Lam Tang
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA
| |
Collapse
|
96
|
Cycloheximide Can Induce Bax/Bak Dependent Myeloid Cell Death Independently of Multiple BH3-Only Proteins. PLoS One 2016; 11:e0164003. [PMID: 27806040 PMCID: PMC5091851 DOI: 10.1371/journal.pone.0164003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 09/19/2016] [Indexed: 01/25/2023] Open
Abstract
Apoptosis mediated by Bax or Bak is usually thought to be triggered by BH3-only members of the Bcl-2 protein family. BH3-only proteins can directly bind to and activate Bax or Bak, or indirectly activate them by binding to anti-apoptotic Bcl-2 family members, thereby relieving their inhibition of Bax and Bak. Here we describe a third way of activation of Bax/Bak dependent apoptosis that does not require triggering by multiple BH3-only proteins. In factor dependent myeloid (FDM) cell lines, cycloheximide induced apoptosis by a Bax/Bak dependent mechanism, because Bax-/-Bak-/- lines were profoundly resistant, whereas FDM lines lacking one or more genes for BH3-only proteins remained highly sensitive. Addition of cycloheximide led to the rapid loss of Mcl-1 but did not affect the expression of other Bcl-2 family proteins. In support of these findings, similar results were observed by treating FDM cells with the CDK inhibitor, roscovitine. Roscovitine reduced Mcl-1 abundance and caused Bax/Bak dependent cell death, yet FDM lines lacking one or more genes for BH3-only proteins remained highly sensitive. Therefore Bax/Bak dependent apoptosis can be regulated by the abundance of anti-apoptotic Bcl-2 family members such as Mcl-1, independently of several known BH3-only proteins.
Collapse
|
97
|
Transcriptome altered by latent human cytomegalovirus infection on THP-1 cells using RNA-seq. Gene 2016; 594:144-150. [PMID: 27623506 PMCID: PMC7126988 DOI: 10.1016/j.gene.2016.09.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 02/08/2023]
Abstract
Human cytomegalovirus (HCMV) has been recognized as a cause of severe, sometimes life-threatening disease in congenitally infected newborns as well as in immunocompromised individuals. However, the molecular mechanisms of the host-virus interaction remain poorly understood. Here, we profiled the expression of mRNAs and long noncoding RNAs (lncRNAs) in THP-1 cells using the emerging RNA-seq to investigate the transcriptional changes during HCMV latent infection. At 4 days post HCMV infection, a total of 169,008,624 sequence reads and 180,616 transcripts were obtained, respectively. Of these transcripts, 1,354 noncoding genes and 12,952 protein-coding genes were observed in Refseq database. Differential gene expression analysis identified 2,153 differentially expressed genes (DEGs) between HCMV-infected and mock-infected THP-1 cells, including 1,098 up-regulated genes and 1,055 down-regulated genes. These regulated genes were involved in pathways of apoptosis, inflammatory response and cell cycle progression, all of which may be implicated in viral pathogenesis. In addition, 646 lncRNAs (208 known lncRNAs and 438 novel lncRNAs) were upregulated and 424 (140 known and 284 novel) were downregulated in infected THP-1 cells. These findings have provided a dynamic scenario of DE candidate genes and lncRNAs at the virus-host interface and clearly warrant further experimental investigation associated with HCMV infection. Differential gene expression analysis identified 2,153 differentially expressed genes between HCMV-infected cells and mock-infected THP-1 cells. These regulated genes were involved in pathways of apoptosis, inflammatory response and cell cycle progression, all of which may be implicated in viral pathogenesis. lncRNAs may involved in regulation of HCMV latent infection.
Collapse
|
98
|
Ambroise G, Portier A, Roders N, Arnoult D, Vazquez A. Subcellular localization of PUMA regulates its pro-apoptotic activity in Burkitt's lymphoma B cells. Oncotarget 2016; 6:38181-94. [PMID: 26431330 PMCID: PMC4741992 DOI: 10.18632/oncotarget.5901] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 09/17/2015] [Indexed: 02/06/2023] Open
Abstract
The BH3-only protein PUMA (p53-upregulated modulator of apoptosis) is a major regulator of apoptosis. It belongs to the Bcl-2 family of proteins responsible for maintaining mitochondrial outer membrane integrity by controlling the intrinsic (mitochondrial) apoptotic pathway. We describe here a new pathway regulating PUMA activation through the control of its subcellular distribution. Surprisingly, neither PUMA upregulation in normal activated human B lymphocytes nor high levels of PUMA in Burkitt's lymphoma (BL) were associated with cell death. We show that PUMA is localized to the cytosol in these cells. By contrast, various apoptosis-triggering signals were found to promote the translocation of PUMA to the mitochondria in these cells, leading to their death by apoptosis. This apoptosis was associated with the binding of mitochondrial PUMA to anti-apoptotic members of the Bcl-2 family, such as Bcl-2 and Mcl-1. This translocation was caspase-independent but was prevented by inhibiting or knocking down the expression of the MAPK kinase p38. Our data suggest that the accumulation of PUMA in the cytosol may be important for the participation of this protein in apoptosis without the need for prior transcription. This regulatory pathway may be an important feature of differentiation and tumorigenic processes.
Collapse
Affiliation(s)
- Gorbatchev Ambroise
- INSERM, UMR_S 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, France.,Equipe Labellisée Ligue contre le Cancer, Villejuif, France
| | - Alain Portier
- INSERM, UMR_S 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, France.,Equipe Labellisée Ligue contre le Cancer, Villejuif, France
| | - Nathalie Roders
- INSERM, UMR_S 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, France.,Equipe Labellisée Ligue contre le Cancer, Villejuif, France
| | - Damien Arnoult
- INSERM, UMR_S 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, France.,Equipe Labellisée Ligue contre le Cancer, Villejuif, France
| | - Aimé Vazquez
- INSERM, UMR_S 1197, Hôpital Paul Brousse, Villejuif, France.,Université Paris-Saclay, France.,Equipe Labellisée Ligue contre le Cancer, Villejuif, France
| |
Collapse
|
99
|
Yang LY, Greig NH, Huang YN, Hsieh TH, Tweedie D, Yu QS, Hoffer BJ, Luo Y, Kao YC, Wang JY. Post-traumatic administration of the p53 inactivator pifithrin-α oxygen analogue reduces hippocampal neuronal loss and improves cognitive deficits after experimental traumatic brain injury. Neurobiol Dis 2016; 96:216-226. [PMID: 27553877 DOI: 10.1016/j.nbd.2016.08.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/04/2016] [Accepted: 08/18/2016] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Neuronal apoptosis in the hippocampus has been detected after TBI. The hippocampal dysfunction may result in cognitive deficits in learning, memory, and spatial information processing. Our previous studies demonstrated that a p53 inhibitor, pifithrin-α oxygen analogue (PFT-α (O)), significantly reduced cortical cell death, which is substantial following controlled cortical impact (CCI) TBI, and improved neurological functional outcomes via anti-apoptotic mechanisms. In the present study, we examined the effect of PFT-α (O) on CCI TBI-induced hippocampal cellular pathophysiology in light of this brain region's role in memory. To investigate whether p53-dependent apoptosis plays a role in hippocampal neuronal loss and associated cognitive deficits and to define underlying mechanisms, SD rats were subjected to experimental CCI TBI followed by the administration of PFT-α or PFT-α (O) (2mg/kg, i.v.) or vehicle at 5h after TBI. Magnetic resonance imaging (MRI) scans were acquired at 24h and 7days post-injury to assess evolving structural hippocampal damage. Fluoro-Jade C was used to stain hippocampal sub-regions, including CA1 and dentate gyrus (DG), for cellular degeneration. Neurological functions, including motor and recognition memory, were assessed by behavioral tests at 7days post injury. p53, p53 upregulated modulator of apoptosis (PUMA), 4-hydroxynonenal (4-HNE), cyclooxygenase-IV (COX IV), annexin V and NeuN were visualized by double immunofluorescence staining with cell-specific markers. Levels of mRNA encoding for caspase-3, p53, PUMA, Bcl-2, Bcl-2-associated X protein (BAX) and superoxide dismutase (SOD) were measured by RT-qPCR. Our results showed that post-injury administration of PFT-α and, particularly, PFT-α (O) at 5h dramatically reduced injury volumes in the ipsilateral hippocampus, improved motor outcomes, and ameliorated cognitive deficits at 7days after TBI, as evaluated by novel object recognition and open-field test. PFT-α and especially PFT-α (O) significantly reduced the number of FJC-positive cells in hippocampus CA1 and DG subregions, versus vehicle treatment, and significantly decreased caspase-3 and PUMA mRNA expression. PFT-α (O), but not PFT-α, treatment significantly lowered p53 and elevated SOD2 mRNA expression. Double immunofluorescence staining demonstrated that PFT-α (O) treatment decreased p53, annexin V and 4-HNE positive neurons in the hippocampal CA1 region. Furthermore, PUMA co-localization with the mitochondrial maker COX IV, and the upregulation of PUMA were inhibited by PFT-α (O) after TBI. Our data suggest that PFT-α and especially PFT-α (O) significantly reduce hippocampal neuronal degeneration, and ameliorate neurological and cognitive deficits in vivo via antiapoptotic and antioxidative properties.
Collapse
Affiliation(s)
- Ling-Yu Yang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Nigel H Greig
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Ya-Ni Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan, Taiwan
| | - Tsung-Hsun Hsieh
- Department of Physical Therapy and Graduate Institute of Rehabilitation Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Neural Regenerative Medicine, Taipei Medical University, Taipei, Taiwan
| | - David Tweedie
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Qian-Sheng Yu
- Drug Design & Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, USA
| | - Barry J Hoffer
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yu Luo
- Department of Neurosurgery, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yu-Chieh Kao
- Translational Imaging Research Center and Department of Radiology, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
100
|
Bahl V, Johnson K, Phandthong R, Zahedi A, Schick SF, Talbot P. From the Cover: Thirdhand Cigarette Smoke Causes Stress-Induced Mitochondrial Hyperfusion and Alters the Transcriptional Profile of Stem Cells. Toxicol Sci 2016; 153:55-69. [PMID: 27255386 DOI: 10.1093/toxsci/kfw102] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Thirdhand cigarette smoke (THS) was recently recognized as an environmental health hazard; however, little is known about it effects on cells. Mitochondria are sensitive monitors of cell health and report on environmentally induced stress. We tested the effects of low levels of THS extracted from terry cloth on mitochondrial morphology and function using stem cells with well-defined mitochondria. Concentrations of THS that did not kill cells caused stress-induced mitochondrial hyperfusion (SIMH), which was characterized by changes in mitochondrial morphology indicative of fusion, increased mitochondrial membrane potential (MMP), increased ATP levels, increased superoxide production, and increased oxidation of mitochondrial proteins. SIMH was accompanied by a decrease in Fis1 expression, a gene responsible for mitochondrial fission, and a decrease in apoptosis-related genes, including Aifm2, Bbc3, and Bid There was also down regulation of Ucp2, Ucp4, and Ucp5, genes that decrease MMP thereby reducing oxidative phosphorylation, while promoting glycolysis. These effects, which collectively accompany SIMH, are a prosurvival mechanism to rescue damaged mitochondria and protect cells from apoptosis. Prolonged exposure to THS caused a reduction in MMP and decreased cell proliferation, which likely leads to apoptosis.
Collapse
Affiliation(s)
- Vasundhra Bahl
- *Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521 Environmental Toxicology Graduate Program University of California Riverside
| | - Kimberly Johnson
- *Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521
| | - Rattapol Phandthong
- *Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521
| | - Atena Zahedi
- *Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521 Bioengineering Interdepartmental Graduate Program, University of California Riverside
| | - Suzaynn F Schick
- Division of Occupational and Environmental Medicine, Department of Medicine, University of California, San Francisco, California 94243
| | - Prue Talbot
- *Department of Cell Biology and Neuroscience, University of California Riverside, Riverside, California 92521
| |
Collapse
|