1
|
Gualandi N, Minisini M, Bertozzo A, Brancolini C. Dissecting transposable elements and endogenous retroviruses upregulation by HDAC inhibitors in leiomyosarcoma cells: Implications for the interferon response. Genomics 2024; 116:110909. [PMID: 39103003 DOI: 10.1016/j.ygeno.2024.110909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
Transposable elements (TEs) are of interest as immunomodulators for cancer therapies. TEs can fold into dsRNAs that trigger the interferon response. Here, we investigated the effect of different HDAC inhibitors (HDACIs) on the expression of TEs in leiomyosarcoma cells. Our data show that endogenous retroviruses (ERVs), especially ERV1 elements, are upregulated after treatment with HDAC1/2/3-specific inhibitors. Surprisingly, the interferon response was not activated. We observed an increase in A-to-I editing of upregulated ERV1. This could have an impact on the stability of dsRNAs and the activation of the interferon response. We also found that H3K27ac levels are increased in the LTR12 subfamilies, which could be regulatory elements controlling the expression of proapoptotic genes such as TNFRSF10B. In summary, we provide a detailed characterization of TEs modulation in response to HDACIs and suggest the use of HDACIs in combination with ADAR inhibitors to induce cell death and support immunotherapy in cancer.
Collapse
Affiliation(s)
- Nicolò Gualandi
- Department of Medicine, Università degli Studi di Udine, P.le Kolbe 4, 33100 Udine, Italy
| | - Martina Minisini
- Department of Medicine, Università degli Studi di Udine, P.le Kolbe 4, 33100 Udine, Italy
| | - Alessio Bertozzo
- Department of Medicine, Università degli Studi di Udine, P.le Kolbe 4, 33100 Udine, Italy
| | - Claudio Brancolini
- Department of Medicine, Università degli Studi di Udine, P.le Kolbe 4, 33100 Udine, Italy.
| |
Collapse
|
2
|
Choi J, Ceribelli M, Phelan JD, Häupl B, Huang DW, Wright GW, Hsiao T, Morris V, Ciccarese F, Wang B, Corcoran S, Scheich S, Yu X, Xu W, Yang Y, Zhao H, Zhou J, Zhang G, Muppidi J, Inghirami GG, Oellerich T, Wilson WH, Thomas CJ, Staudt LM. Molecular targets of glucocorticoids that elucidate their therapeutic efficacy in aggressive lymphomas. Cancer Cell 2024; 42:833-849.e12. [PMID: 38701792 PMCID: PMC11168741 DOI: 10.1016/j.ccell.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 05/05/2024]
Abstract
Glucocorticoids have been used for decades to treat lymphomas without an established mechanism of action. Using functional genomic, proteomic, and chemical screens, we discover that glucocorticoids inhibit oncogenic signaling by the B cell receptor (BCR), a recurrent feature of aggressive B cell malignancies, including diffuse large B cell lymphoma and Burkitt lymphoma. Glucocorticoids induce the glucocorticoid receptor (GR) to directly transactivate genes encoding negative regulators of BCR stability (LAPTM5; KLHL14) and the PI3 kinase pathway (INPP5D; DDIT4). GR directly represses transcription of CSK, a kinase that limits the activity of BCR-proximal Src-family kinases. CSK inhibition attenuates the constitutive BCR signaling of lymphomas by hyperactivating Src-family kinases, triggering their ubiquitination and degradation. With the knowledge that glucocorticoids disable oncogenic BCR signaling, they can now be deployed rationally to treat BCR-dependent aggressive lymphomas and used to construct mechanistically sound combination regimens with inhibitors of BTK, PI3 kinase, BCL2, and CSK.
Collapse
MESH Headings
- Humans
- Glucocorticoids/pharmacology
- Receptors, Antigen, B-Cell/metabolism
- Animals
- Signal Transduction/drug effects
- Receptors, Glucocorticoid/metabolism
- Mice
- Cell Line, Tumor
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Burkitt Lymphoma/drug therapy
- Burkitt Lymphoma/genetics
- Burkitt Lymphoma/metabolism
- Burkitt Lymphoma/pathology
- Molecular Targeted Therapy/methods
- Phosphatidylinositol 3-Kinases/metabolism
- src-Family Kinases/metabolism
- Gene Expression Regulation, Neoplastic/drug effects
Collapse
Affiliation(s)
- Jaewoo Choi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michele Ceribelli
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - James D Phelan
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Björn Häupl
- Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany
| | - Da Wei Huang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - George W Wright
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tony Hsiao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vivian Morris
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francesco Ciccarese
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA; Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, via Gattamelata 64, 35128 Padova, Italy
| | - Boya Wang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sean Corcoran
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sebastian Scheich
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany; University Cancer Center (UCT) Frankfurt, University Hospital, Goethe University, 60590 Frankfurt am Main, Germany; Frankfurt Cancer Institute, Goethe University, 60596 Frankfurt am Main, Germany
| | - Xin Yu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Weihong Xu
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yandan Yang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hong Zhao
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Joyce Zhou
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grace Zhang
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jagan Muppidi
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Giorgio G Inghirami
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10065, USA
| | - Thomas Oellerich
- Department of Medicine II, Hematology/Oncology, Goethe University, Frankfurt, Germany; German Cancer Consortium (DKTK), Partner Site Frankfurt/Mainz, 60528 Frankfurt am Main, Germany
| | - Wyndham H Wilson
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Craig J Thomas
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Louis M Staudt
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Lu Y, Tang K, Wang S, Gao P, Tian Z, Wang M, Chen J, Xiao C, Zhao J, Xie J. Genetic Programs Between Steroid-Sensitive and Steroid-Insensitive Interstitial Lung Disease. Inflammation 2023; 46:2120-2131. [PMID: 37561311 PMCID: PMC10673734 DOI: 10.1007/s10753-023-01866-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 08/11/2023]
Abstract
The effectiveness of corticosteroids (GCs) varies greatly in interstitial lung diseases (ILDs). In this study, we aimed to compare the gene expression profiles of patients with cryptogenic organizing pneumonia (COP), idiopathic pulmonary fibrosis (IPF), and non-specific interstitial pneumonia (NSIP) and identify the molecules and pathways responsible for GCs sensitivity in ILDs. Three datasets (GSE21411, GSE47460, and GSE32537) were selected. Differentially expressed genes (DEGs) among COP, IPF, NSIP, and healthy control (CTRL) groups were identified. Functional enrichment analysis and protein-protein interaction network analysis were performed to examine the potential functions of DEGs. There were 128 DEGs when COP versus CTRL, 257 DEGs when IPF versus CTRL, 205 DEGs when NSIP versus CTRL, and 270 DEGs when COP versus IPF. The DEGs in different ILDs groups were mainly enriched in the inflammatory response. Further pathway analysis showed that "interleukin (IL)-17 signaling pathway" (hsa04657) and "tumor necrosis factor (TNF) signaling pathway" were associated with different types of ILDs. A total of 10 genes associated with inflammatory response were identified as hub genes and their expression levels in the IPF group were higher than those in the COP group. Finally, we identified two GCs' response-related differently expressed genes (FOSL1 and DDIT4). Our bioinformatics analysis demonstrated that the inflammatory response played a pathogenic role in the progression of ILDs. We also illustrated that the inflammatory reaction was more severe in the IPF group compared to the COP group and identified two GCs' response-related differently expressed genes (FOSL1 and DDIT4) in ILDs.
Collapse
Affiliation(s)
- Yanjiao Lu
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kun Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Shanshan Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pengfei Gao
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Zhen Tian
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Meijia Wang
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jinkun Chen
- Western University, 1151 Richmond Street, London, ON, N6A 3K7, Canada
| | - Chengfeng Xiao
- Department of Biology, Queens University, Kingston, ON, K7L 3N6, Canada
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, National Clinical Research Center of Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Dai C, Lin B, Xing X, Liu JS. A Scale-free Approach for False Discovery Rate Control in Generalized Linear Models. J Am Stat Assoc 2023. [DOI: 10.1080/01621459.2023.2165930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Buyu Lin
- Department of Statistics, Harvard University
| | - Xin Xing
- Department of Statistics, Virginia Tech
| | - Jun S. Liu
- Department of Statistics, Harvard University
| |
Collapse
|
5
|
Nutritional Sensor REDD1 in Cancer and Inflammation: Friend or Foe? Int J Mol Sci 2022; 23:ijms23179686. [PMID: 36077083 PMCID: PMC9456073 DOI: 10.3390/ijms23179686] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022] Open
Abstract
Regulated in Development and DNA Damage Response 1 (REDD1)/DNA Damage-Induced Transcript 4 (DDIT4) is an immediate early response gene activated by different stress conditions, including growth factor depletion, hypoxia, DNA damage, and stress hormones, i.e., glucocorticoids. The most known functions of REDD1 are the inhibition of proliferative signaling and the regulation of metabolism via the repression of the central regulator of these processes, the mammalian target of rapamycin (mTOR). The involvement of REDD1 in cell growth, apoptosis, metabolism, and oxidative stress implies its role in various pathological conditions, including cancer and inflammatory diseases. Recently, REDD1 was identified as one of the central genes mechanistically involved in undesirable atrophic effects induced by chronic topical and systemic glucocorticoids widely used for the treatment of blood cancer and inflammatory diseases. In this review, we discuss the role of REDD1 in the regulation of cell signaling and processes in normal and cancer cells, its involvement in the pathogenesis of different diseases, and the approach to safer glucocorticoid receptor (GR)-targeted therapies via a combination of glucocorticoids and REDD1 inhibitors to decrease the adverse atrophogenic effects of these steroids.
Collapse
|
6
|
Zhang W, Deng X, Liu H, Ke J, Xiang M, Ma Y, Zhang L, Yang M, Liu Y, Huang F. Identification and Verification of Potential Hub Genes in Amphetamine-Type Stimulant (ATS) and Opioid Dependence by Bioinformatic Analysis. Front Genet 2022; 13:837123. [PMID: 35432486 PMCID: PMC9006114 DOI: 10.3389/fgene.2022.837123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: Amphetamine-type stimulant (ATS) and opioid dependencies are chronic inflammatory diseases with similar symptoms and common genomics. However, their coexpressive genes have not been thoroughly investigated. We aimed to identify and verify the coexpressive hub genes and pathway involved in the pathogenesis of ATS and opioid dependencies. Methods: The microarray of ATS- and opioid-treatment mouse models was obtained from the Gene Expression Omnibus database. GEO2R and Venn diagram were performed to identify differentially expressed genes (DEGs) and coexpressive DEGs (CDEGs). Functional annotation and protein–protein interaction network detected the potential functions. The hub genes were screened using the CytoHubba and MCODE plugin with different algorithms, and further validated by receiver operating characteristic analysis in the GSE15774 database. We also validated the hub genes mRNA levels in BV2 cells using qPCR. Result: Forty-four CDEGs were identified between ATS and opioid databases, which were prominently enriched in the PI3K/Akt signaling pathway. The top 10 hub genes were mainly enriched in apoptotic process (CD44, Dusp1, Sgk1, and Hspa1b), neuron differentiation, migration, and proliferation (Nr4a2 and Ddit4), response to external stimulation (Fos and Cdkn1a), and transcriptional regulation (Nr4a2 and Npas4). Receiver operating characteristic (ROC) analysis found that six hub genes (Fos, Dusp1, Sgk1, Ddit4, Cdkn1a, and Nr4a2) have an area under the curve (AUC) of more than 0.70 in GSE15774. The mRNA levels of Fos, Dusp1, Sgk1, Ddit4, Cdkn1a, PI3K, and Akt in BV2 cells and GSE15774 with METH and heroin treatments were higher than those of controls. However, the Nr4a2 mRNA levels increased in BV2 cells and decreased in the bioinformatic analysis. Conclusions: The identification of hub genes was associated with ATS and opioid dependencies, which were involved in apoptosis, neuron differentiation, migration, and proliferation. The PI3K/Akt signaling pathway might play a critical role in the pathogenesis of substance dependence.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Forensic Pathology, West China School of Basic Medical Science & Forensic Medicine, Sichuan University, Chengdu, China
| | - Xiaodong Deng
- Department of Forensic Pathology, West China School of Basic Medical Science & Forensic Medicine, Sichuan University, Chengdu, China
- Department of Forensic Pathology, School of Basic Medical Science & Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Huan Liu
- Department of Preventive Medicine, North Sichuan Medical College, Nanchong, China
| | - Jianlin Ke
- Department of Forensic Pathology, School of Basic Medical Science & Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Mingliang Xiang
- Department of Forensic Pathology, School of Basic Medical Science & Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Ying Ma
- Department of Neurology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Lixia Zhang
- Department of Forensic Pathology, School of Basic Medical Science & Forensic Medicine, North Sichuan Medical College, Nanchong, China
| | - Ming Yang
- Department of Forensic Pathology, School of Basic Medical Science & Forensic Medicine, North Sichuan Medical College, Nanchong, China
- Department of Criminal Investigation, Nanchong Municipal Public Security Bureau, Nanchong, China
| | - Yun Liu
- Department of Forensic Pathology, School of Basic Medical Science & Forensic Medicine, North Sichuan Medical College, Nanchong, China
- Medical Imaging Key Laboratory of Sichuan Province, North Sichuan Medical College, Nanchong, China
- *Correspondence: Yun Liu, ; Feijun Huang,
| | - Feijun Huang
- Department of Forensic Pathology, West China School of Basic Medical Science & Forensic Medicine, Sichuan University, Chengdu, China
- *Correspondence: Yun Liu, ; Feijun Huang,
| |
Collapse
|
7
|
Li W, Hu S, Tian C, Wan X, Yu W, Guo P, Zhao F, Hua C, Lu X, Xue G, Han S, Guo W, Wang D, Deng W. TRIP4 transcriptionally activates DDIT4 and subsequent mTOR signaling to promote glioma progression. Free Radic Biol Med 2021; 177:31-47. [PMID: 34648907 DOI: 10.1016/j.freeradbiomed.2021.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/25/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022]
Abstract
In spite of significant advances in the understanding of glioma biology and pathology, survival remains poor. Therefore, it is still of great significance to further explore the key factors involved in tumorigenesis and development in glioma and find potential new therapeutic targets. Here, we show that thyroid hormone receptor interactor 4 (TRIP4) is highly expressed in glioma cells and tissues. Patients of glioma with high expression of TRIP4 possess poor overall survival. Knockdown of TRIP4 inhibited tumor cell proliferation, metastasis, and apoptosis suppression, whereas overexpression of TRIP4 displays the opposite effects. Further research showed that TRIP4 promoted glioma progression through regulating DDIT4 expression and subsequent activation of mTOR signaling. DDIT4 overexpression restored the inhibition of tumor growth by TRIP4 knockdown in vitro and in vivo. Consistently, mTOR activity inhibition reversed TRIP4 overexpression-mediated tumor promotion in vitro and in vivo. Moreover, molecular mechanism exploration demonstrates that TRIP4 functions as a specific transcriptional activator to anchor at the promoter region of DDIT4 gene (-196 to -11) to regulate its transcription and such regulation was affected by HIF1α. Clinically, TRIP4 expression is positively correlated with DDIT4 expression in glioma samples based on tissue microarray analysis and both of their high expression predicts the malignancy of the disease. Altogether, our findings identify TRIP4 as a critical promoter of glioma progression by targeting DDIT4 and mTOR signaling successively and suggest that TRIP4-DDIT4 axis has potential to be a novel therapeutic target in glioma treatment.
Collapse
Affiliation(s)
- Wenyang Li
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Sheng Hu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Chunfang Tian
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Xinyu Wan
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Wendan Yu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Ping Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Feng Zhao
- Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Chunyu Hua
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Xiaona Lu
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Guoqing Xue
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Shilong Han
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China
| | - Wei Guo
- Institute of Cancer Stem Cells, Dalian Medical University, Dalian, China.
| | - Dong Wang
- Department of Hepatobiliary Surgery, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, China.
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center of Cancer Medicine, Guangzhou, China.
| |
Collapse
|
8
|
Todosenko N, Vulf M, Yurova K, Khaziakhmatova O, Mikhailova L, Litvinova L. Causal Links between Hypovitaminosis D and Dysregulation of the T Cell Connection of Immunity Associated with Obesity and Concomitant Pathologies. Biomedicines 2021; 9:1750. [PMID: 34944566 PMCID: PMC8698424 DOI: 10.3390/biomedicines9121750] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/17/2021] [Accepted: 11/20/2021] [Indexed: 12/12/2022] Open
Abstract
Subclinical inflammation in morbid obesity is associated with immune activation and the development of concomitant diseases. Impaired immune homeostasis and immune cell dysregulation in adipose tissue are associated with phenotypic and functional changes in the pool of T lymphocytes and the development of chronic hypovitaminosis D. Low vitamin D levels in obesity lead to the activation, proliferation and production of pro-inflammatory mediators by T cells. Hypovitaminosis D is the cause of a decrease in the functional potential of regulatory and anti-inflammatory lymphocytes and the maintenance of the inflammatory response. The exact molecular genetic mechanisms of the effect of vitamin D on T lymphocytes have not been fully elucidated. Therefore, uncovering the functional role of T cells and their relationship to vitamin D homeostasis in the context of obesity development may contribute to the development of new pathogenetic methods for clinical prediction of the risk of metabolic, oncologic, autoimmune and infectious complications. The review presents the molecular genetic mechanisms of the effect of vitamin D on adipose tissue resident T lymphocytes and the characteristics of vitamin D receptor expression, and analyzes the phenotypic and functional characteristics of potentially pathogenic T lymphocytes in relation to the development of obesity and its associated complications.
Collapse
Affiliation(s)
- Natalia Todosenko
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (K.Y.); (O.K.); (L.L.)
| | - Maria Vulf
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (K.Y.); (O.K.); (L.L.)
| | - Kristina Yurova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (K.Y.); (O.K.); (L.L.)
| | - Olga Khaziakhmatova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (K.Y.); (O.K.); (L.L.)
| | - Larisa Mikhailova
- Department of Therapy Medical Institute, Immanuel Kant Baltic Federal University, 236016 Kaliningrad, Russia;
| | - Larisa Litvinova
- Center for Immunology and Cellular Biotechnology, Immanuel Kant Baltic Federal University, 236001 Kaliningrad, Russia; (N.T.); (K.Y.); (O.K.); (L.L.)
| |
Collapse
|
9
|
Fattahi F, Saeednejad Zanjani L, Habibi Shams Z, Kiani J, Mehrazma M, Najafi M, Madjd Z. High expression of DNA damage-inducible transcript 4 (DDIT4) is associated with advanced pathological features in the patients with colorectal cancer. Sci Rep 2021; 11:13626. [PMID: 34211002 PMCID: PMC8249407 DOI: 10.1038/s41598-021-92720-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/14/2021] [Indexed: 02/08/2023] Open
Abstract
DNA damage-inducible transcript 4 (DDIT4) is induced in various cellular stress conditions. This study was conducted to investigate expression and prognostic significance of DDIT4 protein as a biomarker in the patients with colorectal cancer (CRC). PPI network and KEGG pathway analysis were applied to identify hub genes among obtained differentially expressed genes in CRC tissues from three GEO Series. In clinical, expression of DDIT4 as one of hub genes in three subcellular locations was evaluated in 198 CRC tissues using immunohistochemistry method on tissue microarrays. The association between DDIT4 expression and clinicopathological features as well as survival outcomes were analyzed. Results of bioinformatics analysis indicated 14 hub genes enriched in significant pathways according to KEGG pathways analysis among which DDIT4 was selected to evaluate CRC tissues. Overexpression of nuclear DDIT4 protein was found in CRC tissues compared to adjacent normal tissues (P = 0.003). Furthermore, higher nuclear expression of DDIT4 was found to be significantly associated with the reduced tumor differentiation and advanced TNM stages (all, P = 0.009). No significant association was observed between survival outcomes and nuclear expression of DDIT4 in CRC cases. Our findings indicated higher nuclear expression of DDIT4 was significantly associated with more aggressive tumor behavior and more advanced stage of disease in the patients with CRC.
Collapse
Affiliation(s)
- Fahimeh Fattahi
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Mehrazma
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.,Department of Pathology, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Najafi
- Biochemistry Department, Faculty of Medical Sciences, Iran University of Medical Sciences, Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran. .,Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran. .,Department of Pathology, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
The self-renewal dental pulp stem cell microtissues challenged by a toxic dental monomer. Biosci Rep 2021; 40:225156. [PMID: 32495822 PMCID: PMC7303350 DOI: 10.1042/bsr20200210] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/12/2020] [Accepted: 05/19/2020] [Indexed: 01/19/2023] Open
Abstract
Dental pulp stem cells (DPSCs) regenerate injured/diseased pulp tissue and deposit tertiary dentin. DPSCs stress response can be activated by exposing cells to the monomer triethyleneglycol dimethacrylate (TEGDMA) and inducing the DNA-damage inducible transcript 4 (DDIT4) protein expression. The goal of the present study was to determine the impact of TEGDMA on the ability of DPSCs to maintain their self-renewal capabilities, develop and preserve their 3D structures and deposit the mineral. Human primary and immortalized DPSCs were cultured in extracellular matrix/basement membrane (ECM/BM) to support stemness and to create multicellular interacting layers (microtissues). The microtissues were exposed to the toxic concentrations of TEGDMA (0.5 and 1.5 mmol/l). The DPSCs spatial architecture was assessed by confocal microscopy. Mineral deposition was detected by alizarin red staining and visualized by stereoscopy. Cellular self-renewal transcription factor SOX2 was determined by immunocytochemistry. The microtissue thicknesses/vertical growth, surface area of the mineralizing microtissues, the percentage of area covered by the deposited mineral, and the fluorescence intensity of the immunostained cells were quantified ImageJ. DDIT4 expression was determined by a single molecule RNA-FISH technique and the cell phenotype was determined morphologically. DDIT4 expression was correlated with the cytotoxic phenotype. TEGDMA affected the structures of developing and mature microtissues. It inhibited the deposition of the mineral in the matrix while not affecting the SOX2 expression. Our data demonstrate that DPSCs retained their self-renewal capacity although their other functions were impeded. Since the DPSCs pool remained preserved, properties effected by the irritant should be restored by a proper rescue therapy.
Collapse
|
11
|
Sævik ÅB, Wolff AB, Björnsdottir S, Simunkova K, Hynne MS, Dolan DWP, Bratland E, Knappskog PM, Methlie P, Carlsen S, Isaksson M, Bensing S, Kämpe O, Husebye ES, Løvås K, Øksnes M. Potential Transcriptional Biomarkers to Guide Glucocorticoid Replacement in Autoimmune Addison's Disease. J Endocr Soc 2021; 5:bvaa202. [PMID: 33553982 PMCID: PMC7853175 DOI: 10.1210/jendso/bvaa202] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Background No reliable biomarkers exist to guide glucocorticoid (GC) replacement treatment in autoimmune Addison's disease (AAD), leading to overtreatment with alarming and persistent side effects or undertreatment, which could be fatal. Objective To explore changes in gene expression following different GC replacement doses as a means of identifying candidate transcriptional biomarkers to guide GC replacement in AAD. Methods Step 1: Global microarray expression analysis on RNA from whole blood before and after intravenous infusion of 100 mg hydrocortisone (HC) in 10 patients with AAD. In 3 of the most highly upregulated genes, we performed real-time PCR (rt-PCR) to compare gene expression levels before and 3, 4, and 6 hours after the HC infusion. Step 2: Rt-PCR to compare expression levels of 93 GC-regulated genes in normal versus very low morning cortisol levels in 27 patients with AAD. Results Step 1: Two hours after infusion of 100 mg HC, there was a marked increase in FKBP5, MMP9, and DSIPI expression levels. MMP9 and DSIPI expression levels correlated with serum cortisol. Step 2: Expression levels of CEBPB, DDIT4, FKBP5, DSIPI, and VDR were increased and levels of ADARB1, ARIDB5, and POU2F1 decreased in normal versus very low morning cortisol. Normal serum cortisol levels positively correlated with DSIPI, DDIT4, and FKBP5 expression. Conclusions We introduce gene expression as a novel approach to guide GC replacement in AAD. We suggest that gene expression of DSIPI, DDIT4, and FKBP5 are particularly promising candidate biomarkers of GC replacement, followed by MMP9, CEBPB, VDR, ADARB1, ARID5B, and POU2F1.
Collapse
Affiliation(s)
- Åse Bjorvatn Sævik
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway
| | - Anette B Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway
| | - Sigridur Björnsdottir
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | | | | | | | - Eirik Bratland
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Per M Knappskog
- K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Paal Methlie
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Siri Carlsen
- Department of Endocrinology, Stavanger University Hospital, Stavanger, Norway
| | - Magnus Isaksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Sophie Bensing
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden
| | - Olle Kämpe
- K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine (Solna), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Eystein S Husebye
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway.,Department of Medicine (Solna), Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Kristian Løvås
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Marianne Øksnes
- Department of Clinical Science, University of Bergen, Bergen, Norway.,K.G. Jebsen Center for Autoimmune Disorders, University of Bergen, Bergen, Norway.,Department of Endocrinology, Karolinska University Hospital, Stockholm, Sweden.,Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
12
|
Britto FA, Dumas K, Giorgetti-Peraldi S, Ollendorff V, Favier FB. Is REDD1 a metabolic double agent? Lessons from physiology and pathology. Am J Physiol Cell Physiol 2020; 319:C807-C824. [PMID: 32877205 DOI: 10.1152/ajpcell.00340.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Akt/mechanistic target of rapamycin (mTOR) signaling pathway governs macromolecule synthesis, cell growth, and metabolism in response to nutrients and growth factors. Regulated in development and DNA damage response (REDD)1 is a conserved and ubiquitous protein, which is transiently induced in response to multiple stimuli. Acting like an endogenous inhibitor of the Akt/mTOR signaling pathway, REDD1 protein has been shown to regulate cell growth, mitochondrial function, oxidative stress, and apoptosis. Recent studies also indicate that timely REDD1 expression limits Akt/mTOR-dependent synthesis processes to spare energy during metabolic stresses, avoiding energy collapse and detrimental consequences. In contrast to this beneficial role for metabolic adaptation, REDD1 chronic expression appears involved in the pathogenesis of several diseases. Indeed, REDD1 expression is found as an early biomarker in many pathologies including inflammatory diseases, cancer, neurodegenerative disorders, depression, diabetes, and obesity. Moreover, prolonged REDD1 expression is associated with cell apoptosis, excessive reactive oxygen species (ROS) production, and inflammation activation leading to tissue damage. In this review, we decipher several mechanisms that make REDD1 a likely metabolic double agent depending on its duration of expression in different physiological and pathological contexts. We also discuss the role played by REDD1 in the cross talk between the Akt/mTOR signaling pathway and the energetic metabolism.
Collapse
Affiliation(s)
| | - Karine Dumas
- Université Cote d'Azur, INSERM, UMR1065, C3M, Nice, France
| | | | | | | |
Collapse
|
13
|
Valenzuela CA, Ponce C, Zuloaga R, González P, Avendaño-Herrera R, Valdés JA, Molina A. Effects of crowding on the three main proteolytic mechanisms of skeletal muscle in rainbow trout (Oncorhynchus mykiss). BMC Vet Res 2020; 16:294. [PMID: 32799856 PMCID: PMC7429773 DOI: 10.1186/s12917-020-02518-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 08/07/2020] [Indexed: 02/07/2023] Open
Abstract
Background Skeletal muscle is one of the tissues most affected by stress conditions. The protein degradation in this tissue is vital for the supply of energy mediated by different proteolytic pathways such as the ubiquitin-proteasome (UPS), autophagy-lysosome (ALS) and the calpain/calpastatin system (CCS). Nevertheless, the regulation of this proteolytic axis under stress conditions is not yet completely clear. Chile is the main producer of rainbow trout (Oncorhynchus mykiss) in the world. This intensive fish farming has resulted in growing problems as crowding and stress are one of the major problems in the freshwater stage. In this context, we evaluated the crowding effect in juvenile rainbow trout kept in high stocking density (30 kg/m3) for 15, 45 and 60 days, using a control group of fish (10 kg/m3). Results Plasmatic cortisol and glucose were evaluated by enzyme immunoassay. The mRNA levels of stress-related genes (gr1, gr2, mr, hsp70, klf15 and redd1), markers of the UPS (atrogin1 and murf1) and CCS (capn1, capn1, cast-l and cast-s) were evaluated using qPCR. ALS (LC3-I/II and P62/SQSTM1) and growth markers (4E-BP1 and ERK) were measured by Western blot analysis. The cortisol levels increased concomitantly with weight loss at 45 days of crowding. The UPS alone was upregulated at 15 days of high stocking density, while ALS activation was observed at 60 days. However, the CCS was inactivated during the entire trial. Conclusion All these data suggest that stress conditions, such as crowding, promote muscle degradation in a time-dependent manner through the upregulation of the UPS at early stages of chronic stress and activation of the ALS in long-term stress, while the CCS is strongly inhibited by stress conditions in the rainbow trout muscle farmed during freshwater stage. Our descriptive study will allow perform functional analysis to determine, in a more detailed way, the effect of stress on skeletal muscle physiology as well as in the animal welfare in rainbow trout. Moreover, it is the first step to elucidate the optimal crop density in the freshwater stage and improve the standards of Chilean aquaculture.
Collapse
Affiliation(s)
- Cristián A Valenzuela
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370146, Santiago, Chile.,Laboratorio de Genética e Inmunología Molecular, Instituto de Biología, Pontificia Universidad Católica de Valparaíso (PUCV), Valparaíso, Chile
| | - Claudia Ponce
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370146, Santiago, Chile
| | - Rodrigo Zuloaga
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370146, Santiago, Chile.,Interdisciplinary Center for Aquaculture Research (INCAR), 4030000, Concepción, Chile
| | - Pamela González
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370146, Santiago, Chile.,Interdisciplinary Center for Aquaculture Research (INCAR), 4030000, Concepción, Chile
| | - Ruben Avendaño-Herrera
- Interdisciplinary Center for Aquaculture Research (INCAR), 4030000, Concepción, Chile. .,Laboratorio de Patología de Organismos Acuáticos y Biotecnología Acuícola, Universidad Andrés Bello, 2520000, Viña del Mar, Chile. .,Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, 2340000, Quintay, Chile.
| | - Juan A Valdés
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370146, Santiago, Chile.,Interdisciplinary Center for Aquaculture Research (INCAR), 4030000, Concepción, Chile.,Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, 2340000, Quintay, Chile
| | - Alfredo Molina
- Laboratorio de Biotecnología Molecular, Facultad de Ciencias de la Vida, Universidad Andrés Bello, 8370146, Santiago, Chile. .,Interdisciplinary Center for Aquaculture Research (INCAR), 4030000, Concepción, Chile. .,Centro de Investigación Marina Quintay (CIMARQ), Universidad Andrés Bello, 2340000, Quintay, Chile.
| |
Collapse
|
14
|
Ricci A, Galluzzi L, Magnani M, Menotta M. DDIT4 gene expression is switched on by a new HDAC4 function in ataxia telangiectasia. FASEB J 2019; 34:1802-1818. [PMID: 31914654 DOI: 10.1096/fj.201902039r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 10/12/2019] [Accepted: 11/06/2019] [Indexed: 12/21/2022]
Abstract
Ataxia telangiectasia (AT) is a rare, severe, and ineluctably progressive multisystemic neurodegenerative disease. Histone deacetylase 4 (HDAC4) nuclear accumulation has been related to neurodegeneration in AT. Since treatment with glucocorticoid analogues has been shown to improve the neurological symptoms that characterize this syndrome, the effects of dexamethasone on HDAC4 were investigated. In this paper, we describe a novel nonepigenetic function of HDAC4 induced by dexamethasone, through which it can directly modulate HIF-1a activity and promote the upregulation of the DDIT4 gene and protein expression. This new HDAC4 transcription regulation mechanism leads to a positive effect on autophagic flux, an AT-compromised biological pathway. This signaling was specifically induced by dexamethasone only in AT cell lines and can contribute in explaining the positive effects of dexamethasone observed in AT-treated patients.
Collapse
Affiliation(s)
- Anastasia Ricci
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Luca Galluzzi
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino "Carlo Bo", Urbino, Italy
| |
Collapse
|
15
|
Nishi M, Ogata T, Cannistraci CV, Ciucci S, Nakanishi N, Higuchi Y, Sakamoto A, Tsuji Y, Mizushima K, Matoba S. Systems Network Genomic Analysis Reveals Cardioprotective Effect of MURC/Cavin-4 Deletion Against Ischemia/Reperfusion Injury. J Am Heart Assoc 2019; 8:e012047. [PMID: 31364493 PMCID: PMC6761664 DOI: 10.1161/jaha.119.012047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Ischemia/reperfusion (I/R) injury is a critical issue in the development of treatment strategies for ischemic heart disease. MURC (muscle‐restricted coiled‐coil protein)/Cavin‐4 (caveolae‐associated protein 4), which is a component of caveolae, is involved in the pathophysiology of dilated cardiomyopathy and cardiac hypertrophy. However, the role of MURC in cardiac I/R injury remains unknown. Methods and Results The systems network genomic analysis based on PC‐corr network inference on microarray data between wild‐type and MURC knockout mouse hearts predicted a network of discriminating genes associated with reactive oxygen species. To demonstrate the prediction, we analyzed I/R‐injured mouse hearts. MURC deletion decreased infarct size and preserved heart contraction with reactive oxygen species–related molecule EGR1 (early growth response protein 1) and DDIT4 (DNA‐damage‐inducible transcript 4) suppression in I/R‐injured hearts. Because PC‐corr network inference integrated with a protein–protein interaction network prediction also showed that MURC is involved in the apoptotic pathway, we confirmed the upregulation of STAT3 (signal transducer and activator of transcription 3) and BCL2 (B‐cell lymphoma 2) and the inactivation of caspase 3 in I/R‐injured hearts of MURC knockout mice compared with those of wild‐type mice. STAT3 inhibitor canceled the cardioprotective effect of MURC deletion in I/R‐injured hearts. In cardiomyocytes exposed to hydrogen peroxide, MURC overexpression promoted apoptosis and MURC knockdown inhibited apoptosis. STAT3 inhibitor canceled the antiapoptotic effect of MURC knockdown in cardiomyocytes. Conclusions Our findings, obtained by prediction from systems network genomic analysis followed by experimental validation, suggested that MURC modulates cardiac I/R injury through the regulation of reactive oxygen species–induced cell death and STAT3‐meditated antiapoptosis. Functional inhibition of MURC may be effective in reducing cardiac I/R injury.
Collapse
Affiliation(s)
- Masahiro Nishi
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Takehiro Ogata
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan.,Department of Pathology and Cell Regulation Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Carlo Vittorio Cannistraci
- Biomedical Cybernetics Group, Biotechnology Center (BIOTEC) Center for Molecular and Cellular Bioengineering (CMCB) Center for Systems Biology Dresden Department of Physics Technische Universität Dresden Dresden Germany.,Tsinghua Laboratory of Brain and Intelligence Tsinghua University Beijing China
| | - Sara Ciucci
- Biomedical Cybernetics Group, Biotechnology Center (BIOTEC) Center for Molecular and Cellular Bioengineering (CMCB) Center for Systems Biology Dresden Department of Physics Technische Universität Dresden Dresden Germany
| | - Naohiko Nakanishi
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Yusuke Higuchi
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Akira Sakamoto
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Yumika Tsuji
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Katsura Mizushima
- Department of Molecular Gastroenterology and Hepatology Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine Graduate School of Medical Science Kyoto Prefectural University of Medicine Kyoto Japan
| |
Collapse
|
16
|
Tirado-Hurtado I, Fajardo W, Pinto JA. DNA Damage Inducible Transcript 4 Gene: The Switch of the Metabolism as Potential Target in Cancer. Front Oncol 2018; 8:106. [PMID: 29707520 PMCID: PMC5906527 DOI: 10.3389/fonc.2018.00106] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 03/26/2018] [Indexed: 12/02/2022] Open
Abstract
DNA damage inducible transcript 4 (DDIT4) gene is expressed under stress situations turning off the metabolic activity triggered by the mammalian target of rapamycin (mTOR). Several in vitro and in vivo works have demonstrated the ability of DDIT4 to generate resistance to cancer therapy. The link between the metabolism suppression and aggressiveness features of cancer cells remains poorly understood since anti-mTOR agents who are part of the repertoire of drugs used for systemic treatment of cancer achieving variable results. Interestingly, the high DDIT4 expression is associated with worse outcomes compared to tumors with low DDIT4 expression, seen in a wide variety of solid and hematological tumors, which suggests the driver role of this gene and provide the basis to target it as part of a new therapeutic strategy. In this review, we highlight our current knowledge about the biology of DDIT4 and its role as a prognostic biomarker, encompassing the motives for the development of target drugs against DDIT4 as a better target than mTOR inhibitors.
Collapse
Affiliation(s)
| | - Williams Fajardo
- Escuela de Medicina Humana, Universidad Privada San Juan Bautista, Lima, Peru
| | - Joseph A Pinto
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Lima, Peru
| |
Collapse
|
17
|
Yang J, Zhang Y, Tong J, Lv H, Zhang C, Chen ZJ. Dysfunction of DNA damage-inducible transcript 4 in the decidua is relevant to the pathogenesis of preeclampsia†. Biol Reprod 2018; 98:821-833. [PMID: 29447340 DOI: 10.1093/biolre/ioy038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/12/2018] [Indexed: 12/25/2022] Open
Affiliation(s)
- Jieqiong Yang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yachao Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Ji’nan, Shandong, China
| | - Jing Tong
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Lv
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Cong Zhang
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Ji’nan, Shandong, China
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Center for Reproductive Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Pinto JA, Rolfo C, Raez LE, Prado A, Araujo JM, Bravo L, Fajardo W, Morante ZD, Aguilar A, Neciosup SP, Mas LA, Bretel D, Balko JM, Gomez HL. In silico evaluation of DNA Damage Inducible Transcript 4 gene (DDIT4) as prognostic biomarker in several malignancies. Sci Rep 2017; 7:1526. [PMID: 28484222 PMCID: PMC5431475 DOI: 10.1038/s41598-017-01207-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 03/27/2017] [Indexed: 01/17/2023] Open
Abstract
DDIT4 gene encodes a protein whose main action is to inhibit mTOR under stress conditions whilst several in vitro studies indicate that its expression favors cancer progression. We have previously described that DDIT4 expression is an independent prognostic factor for tripe negative breast cancer resistant to neoadjuvant chemotherapy. We herein report that high DDIT4 expression is related to the outcome (recurrence-free survival, time to progression and overall survival) in several cancer types. We performed in silico analysis in online platforms, in pooled datasets from KM Plotter and meta-analysis of individual datasets from SurvExpress. High levels of DDIT4 were significantly associated with a worse prognosis in acute myeloid leukemia, breast cancer, glioblastoma multiforme, colon, skin and lung cancer. Conversely, a high DDIT4 expression was associated with an improved prognostic in gastric cancer. DDIT4 was not associated with the outcome of ovarian cancers. Analysis with data from the Cell Miner Tool in 60 cancer cell lines indicated that although rapamycin activity was correlated with levels of MTOR, it is not influenced by DDIT4 expression. In summary, DDIT4 might serve as a novel prognostic biomarker in several malignancies. DDIT4 activity could be responsible for resistance to mTOR inhibitors and is a potential candidate for the development of targeted therapy.
Collapse
Affiliation(s)
- Joseph A Pinto
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Christian Rolfo
- Phase I-Early Clinical trials Unit, Antwerp University Hospital & Center for Oncological Research (CORE), Antwerp, Belgium
| | - Luis E Raez
- Thoracic Oncology Program, Memorial Cancer Institute, Memorial Health Care System, Pembroke Pines, FL, USA
| | - Alexandra Prado
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Jhajaira M Araujo
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Leny Bravo
- Escuela de Medicina Humana, Universidad Privada San Juan Bautista, Av. José Antonio Lavalle s/n Hacienda Villa, Chorrillos. Lima 09, Peru
| | - Williams Fajardo
- Escuela de Medicina Humana, Universidad Privada San Juan Bautista, Av. José Antonio Lavalle s/n Hacienda Villa, Chorrillos. Lima 09, Peru
| | - Zaida D Morante
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Alfredo Aguilar
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Silvia P Neciosup
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Luis A Mas
- Unidad de Investigación Básica y Traslacional, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru
| | - Denisse Bretel
- Grupo de Estudios Clínico Oncológicos Peruano (GECOPERU), Lima, Lima 33, Peru
| | - Justin M Balko
- Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, TN, 37232-6307, USA
| | - Henry L Gomez
- Departamento de Medicina Oncológica, Oncosalud-AUNA, Av. Guardia Civil 571, San Borja. Lima 41, Peru. .,Departamento de Medicina Oncológica, Instituto Peruano de Enfermedades Neoplásicas, Av. Angamos Este 2520, Surquillo. Lima 34, Peru.
| |
Collapse
|
19
|
Cheng YH, Chen IS, Lin YC, Tung CW, Chang HS, Wang CC. Attenuation of antigen-specific T helper 1 immunity by Neolitsea hiiranensis and its derived terpenoids. PeerJ 2016; 4:e2758. [PMID: 28344896 PMCID: PMC5363408 DOI: 10.7717/peerj.2758] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/04/2016] [Indexed: 01/10/2023] Open
Abstract
Background T cells play a pivotal role in the adaptive immunity that participates in a wide range of immune responses through a complicated cytokine network. Imbalance of T-cell responses is involved in several immune disorders. Neolitsea species, one of the biggest genera in the family Lauraceae, have been employed widely as folk medicines for a long time in Asia. Previous phytochemical investigations revealed the abundance of terpenes in the leaves of N. hiiranensis, an endemic Neolitsea in Taiwan, and demonstrated anti-inflammatory activities. However, the effect of N. hiiranensis on the functionality of immune cells, especially T cells, is still unclear. In this study, we utilize in vitro and in vivo approaches to characterize the effects of leaves of N. hiiranensis and its terpenoids on adaptive immune responses. Methods Dried leaves of N. hiiranensis were extracted three times with cold methanol to prepare crude extracts and to isolate its secondary metabolites. The ovalbumin (OVA)-sensitized BALB/c mice were administrated with N. hiiranensis extracts (5–20 mg/kg). The serum and splenocytes of treated mice were collected to evaluate the immunomodulatory effects of N. hiiranensis on the production of OVA-specific antibodies and cytokines. To further identify the N. hiiranensis-derived compounds with immunomodulatory potentials, OVA-primed splenocytes were treated with compounds isolated from N. hiiranensis by determining the cell viability, cytokine productions, and mRNA expression in the presence of OVA in vitro. Results Crude extracts of leaves of N. hiiranensis significantly inhibited IL-12, IFN-γ, and IL-2 cytokine productions as well as the serum levels of antigen-specific IgM and IgG2ain vivo. Two of fourteen selected terpenoids and one diterpenoid derived from the leaves of N. hiiranensis suppressed IFN-γ in vitro. In addition, β-caryophyllene oxide attenuated the expression of IFN-γ, T-bet, and IL-12Rβ2 in a dose-dependent manner. N. hiiranensis-derived β-caryophyllene oxide inhibited several aspects of adaptive immune responses, including T-cell differentiation, IFN-γ production, and Th1-assocaited genes. Conclusion As IFN-γ is the key cytokine secreted by T helper-1 cells and plays a pivotal role in Th1 immune responses, our results suggested that the N. hiiranensis and its terpenoids may possess potential therapeutic effects on Th1-mediated immune disorders.
Collapse
Affiliation(s)
- Yin-Hua Cheng
- Ph.D. Program in Toxicology, College of Pharmacy, Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Ih-Sheng Chen
- School of Pharmacy, Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Ying-Chi Lin
- Ph.D. Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Wei Tung
- Ph.D. Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsun-Shuo Chang
- School of Pharmacy, Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Chia-Chi Wang
- Ph.D. Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan; School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Park KM, Teoh JP, Wang Y, Broskova Z, Bayoumi AS, Tang Y, Su H, Weintraub NL, Kim IM. Carvedilol-responsive microRNAs, miR-199a-3p and -214 protect cardiomyocytes from simulated ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2016; 311:H371-83. [PMID: 27288437 PMCID: PMC5005281 DOI: 10.1152/ajpheart.00807.2015] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The nonselective β-adrenergic receptor antagonist (β-blocker) carvedilol has been shown to protect against myocardial injury, but the detailed underlying mechanisms are unclear. We recently reported that carvedilol stimulates the processing of microRNA (miR)-199a-3p and miR-214 in the heart via β-arrestin1-biased β1-adrenergic receptor (β1AR) cardioprotective signaling. Here, we investigate whether these β-arrestin1/β1AR-responsive miRs mediate the beneficial effects of carvedilol against simulated ischemia/reperfusion (sI/R). Using cultured cardiomyocyte cell lines and primary cardiomyocytes, we demonstrate that carvedilol upregulates miR-199a-3p and miR-214 in both ventricular and atrial cardiomyocytes subjected to sI/R. Overexpression of the two miRs in cardiomyocytes mimics the effects of carvedilol to activate p-AKT survival signaling and the expression of a downstream pluripotency marker Sox2 in response to sI/R. Moreover, carvedilol-mediated p-AKT activation is abolished by knockdown of either miR-199a-3p or miR-214. Along with previous studies to directly link the cardioprotective actions of carvedilol to upregulation of p-AKT/stem cell markers, our findings suggest that the protective roles of carvedilol during ischemic injury are in part attributed to activation of these two protective miRs. Loss of function of miR-199a-3p and miR-214 also increases cardiomyocyte apoptosis after sI/R. Mechanistically, we demonstrate that miR-199a-3p and miR-214 repress the predictive or known apoptotic target genes ddit4 and ing4, respectively, in cardiomyocytes. These findings suggest pivotal roles for miR-199a-3p and miR-214 as regulators of cardiomyocyte survival and contributors to the functional benefits of carvedilol therapy.
Collapse
Affiliation(s)
- Kyoung-Mi Park
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jian-Peng Teoh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yongchao Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zuzana Broskova
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ahmed S Bayoumi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Il-Man Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
21
|
Wolff NC, Pavía-Jiménez A, Tcheuyap VT, Alexander S, Vishwanath M, Christie A, Xie XJ, Williams NS, Kapur P, Posner B, McKay RM, Brugarolas J. High-throughput simultaneous screen and counterscreen identifies homoharringtonine as synthetic lethal with von Hippel-Lindau loss in renal cell carcinoma. Oncotarget 2016. [PMID: 26219258 PMCID: PMC4627284 DOI: 10.18632/oncotarget.4773] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Renal cell carcinoma (RCC) accounts for 85% of primary renal neoplasms, and is rarely curable when metastatic. Approximately 70% of RCCs are clear-cell type (ccRCC), and in >80% the von Hippel-Lindau (VHL) gene is mutated or silenced. We developed a novel, high-content, screening strategy for the identification of small molecules that are synthetic lethal with genes mutated in cancer. In this strategy, the screen and counterscreen are conducted simultaneously by differentially labeling mutant and reconstituted isogenic tumor cell line pairs with different fluorochromes and using a highly sensitive high-throughput imaging-based platform. This approach minimizes confounding factors from sequential screening, and more accurately replicates the in vivo cancer setting where cancer cells are adjacent to normal cells. A screen of ~12,800 small molecules identified homoharringtonine (HHT), an FDA-approved drug for treating chronic myeloid leukemia, as a VHL-synthetic lethal agent in ccRCC. HHT induced apoptosis in VHL-mutant, but not VHL-reconstituted, ccRCC cells, and inhibited tumor growth in 30% of VHL-mutant patient-derived ccRCC tumorgraft lines tested. Building on a novel screening strategy and utilizing a validated RCC tumorgraft model recapitulating the genetics and drug responsiveness of human RCC, these studies identify HHT as a potential therapeutic agent for a subset of VHL-deficient ccRCCs.
Collapse
Affiliation(s)
- Nicholas C Wolff
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine - Hematology-Oncology Division, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Andrea Pavía-Jiménez
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine - Hematology-Oncology Division, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vanina T Tcheuyap
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine - Hematology-Oncology Division, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Shane Alexander
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine - Hematology-Oncology Division, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Mridula Vishwanath
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,BioTek Instruments, Winooski, VT, USA
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xian-Jin Xie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Noelle S Williams
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Payal Kapur
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bruce Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Renée M McKay
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine - Hematology-Oncology Division, University of Texas Southwestern Medical Center, Dallas, Texas, USA.,Department of Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
22
|
Effects of DDIT4 in Methamphetamine-Induced Autophagy and Apoptosis in Dopaminergic Neurons. Mol Neurobiol 2016; 54:1642-1660. [PMID: 26873849 DOI: 10.1007/s12035-015-9637-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 12/15/2015] [Indexed: 01/14/2023]
Abstract
Methamphetamine (METH) is an illicit psychoactive drug that can cause a variety of detrimental effects to the nervous system, especially dopaminergic pathways. We hypothesized that DNA damage-inducible transcript 4 (DDIT4) is involved in METH-induced dopaminergic neuronal autophagy and apoptosis. To test the hypothesis, we determined changes of DDIT4 protein expression and the level of autophagy in rat catecholaminergic PC12 cells and human dopaminergic SH-SY5Y cells, and in the hippocampus, prefrontal cortex, and striatum of Sprague Dawley rats exposed to METH. We also examined the effects of silencing DDIT4 expression on METH-induced dopaminergic neuronal autophagy using fluorescence microscopy and electron microscopy. Flow cytometry and Western blot were used to determine apoptosis and the expression of apoptotic markers (cleaved caspase-3 and cleaved PARP) after blocking DDIT4 expression in PC12 cells and SH-SY5Y cells with synthetic siRNA, as well as in the striatum of rats by injecting LV-shDDIT4 lentivirus using a stereotaxic positioning system. Our results showed that METH exposure increased DDIT4 expression that was accompanied with increased autophagy and apoptosis in PC12 cells (3 mM) and SH-SY5Y cells (2 mM), and in the hippocampus, prefrontal cortex, and striatum of rats. Inhibition of DDIT4 expression reduced METH-induced autophagy and apoptosis in vitro and in vivo. However, DDIT4-related effects were not observed at a low concentration of METH (1 μM). These results suggest that DDIT4 plays an essential role in METH-induced dopaminergic neuronal autophagy and apoptosis at higher doses and may be a potential gene target for therapeutics in high-dose METH-induced neurotoxicity.
Collapse
|
23
|
Consolaro F, Ghaem-Maghami S, Bortolozzi R, Zona S, Khongkow M, Basso G, Viola G, Lam EWF. FOXO3a and Posttranslational Modifications Mediate Glucocorticoid Sensitivity in B-ALL. Mol Cancer Res 2015; 13:1578-90. [PMID: 26376801 DOI: 10.1158/1541-7786.mcr-15-0127] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/02/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Glucocorticoids are widely used to treat B acute lymphoblastic leukemia (B-ALL); however, the molecular mechanism underlying glucocorticoid response and resistance is unclear. In this study, the role and regulation of FOXO3a in mediating the dexamethasone response in B-ALL were investigated. The results show that FOXO3a mediates the cytotoxic function of dexamethasone. In response to dexamethasone, it was found that FOXO3a translocates into the nucleus, where it induces the expression of downstream targets, including p27Kip1 and Bim, important for proliferative arrest and cell death in the sensitive RS4;11 and SUP-B15 B-ALL cells. FOXO3a activation by dexamethasone is mediated partially through the suppression of the PI3K/Akt signaling cascade. Furthermore, two posttranslational modifications were uncovered, phosphorylation on Ser-7 and acetylation on Lys-242/5, that associated with FOXO3a activation by dexamethasone. Immunoblot analysis showed that the phosphorylation on Ser-7 of FOXO3a is associated with p38/JNK activation, whereas the acetylation on Lys-242/5 is correlated with the downregulation of SIRT1/2/6 and the induction of the acetyltransferase CBP/p300. Collectively, these results indicate that FOXO3a is essential for dexamethasone response in B-ALL cells, and its nuclear translocation and activation is associated with its phosphorylation on Ser-7 and acetylation on Lys-242/245. These posttranslational events can be exploited as biomarkers for B-ALL diagnosis and as drug targets for B-ALL treatment, particularly for overcoming the glucocorticoid resistance. IMPLICATIONS FOXO3a and its posttranslational regulation are essential for dexamethasone response, and targeting FOXO3a and sirtuins may enhance the dexamethasone-induced cytotoxicity in B-ALL cells.
Collapse
Affiliation(s)
- Francesca Consolaro
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, United Kingdom. Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, University of Padova, Padova, Italy
| | - Sadaf Ghaem-Maghami
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, United Kingdom
| | - Roberta Bortolozzi
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, University of Padova, Padova, Italy
| | - Stefania Zona
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, United Kingdom
| | - Mattaka Khongkow
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, United Kingdom
| | - Giuseppe Basso
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, University of Padova, Padova, Italy
| | - Giampietro Viola
- Dipartimento di Salute della Donna e del Bambino, Laboratorio di Oncoematologia, University of Padova, Padova, Italy.
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, United Kingdom.
| |
Collapse
|
24
|
Reuschel EL, Wang J, Shivers DK, Muthumani K, Weiner DB, Ma Z, Finkel TH. REDD1 Is Essential for Optimal T Cell Proliferation and Survival. PLoS One 2015; 10:e0136323. [PMID: 26301899 PMCID: PMC4547781 DOI: 10.1371/journal.pone.0136323] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 08/02/2015] [Indexed: 12/21/2022] Open
Abstract
REDD1 is a highly conserved stress response protein that is upregulated following many types of cellular stress, including hypoxia, DNA damage, energy stress, ER stress, and nutrient deprivation. Recently, REDD1 was shown to be involved in dexamethasone induced autophagy in murine thymocytes. However, we know little of REDD1’s function in mature T cells. Here we show for the first time that REDD1 is upregulated following T cell stimulation with PHA or CD3/CD28 beads. REDD1 knockout T cells exhibit a defect in proliferation and cell survival, although markers of activation appear normal. These findings demonstrate a previously unappreciated role for REDD1 in T cell function.
Collapse
Affiliation(s)
- Emma L. Reuschel
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - JiangFang Wang
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Debra K. Shivers
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Karuppiah Muthumani
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Zhengyu Ma
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Terri H. Finkel
- Division of Rheumatology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
25
|
Zhang Z, Li Z, Wu X, Zhang CF, Calway T, He TC, Du W, Chen J, Wang CZ, Yuan CS. TRAIL pathway is associated with inhibition of colon cancer by protopanaxadiol. J Pharmacol Sci 2015; 127:83-91. [PMID: 25704023 PMCID: PMC5053100 DOI: 10.1016/j.jphs.2014.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 10/20/2014] [Accepted: 11/04/2014] [Indexed: 12/26/2022] Open
Abstract
Among important components of American ginseng, protopanaxadiol (PPD) showed more active anticancer potential than other triterpenoid saponins. In this study, we determined the in vivo effects of PPD in a mouse cancer model first. Then, using human colorectal cancer cell lines, we observed significant cancer cell growth inhibition by promoting G1 cell cycle redistribution and apoptosis. Subsequently, we characterized the downstream genes targeted by PPD in HCT-116 cancer cells. Using Affymetrix high density GeneChips, we obtained the gene expression profile of the cells. Microarray data indicated that the expression levels of 76 genes were changed over two-fold after PPD, of which 52 were upregulated while the remaining 24 were downregulated. Ingenuity pathway analysis of top functions affected was carried out. Data suggested that by regulating the interactions between p53 and DR4/DR5, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) pathway played a key role in the action of PPD, a promising colon cancer inhibitory compound.
Collapse
Affiliation(s)
- Zhiyu Zhang
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Zejuan Li
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Xiaohui Wu
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Chun-Feng Zhang
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Tyler Calway
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Tong-Chuan He
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Wei Du
- Ben May Department for Cancer Research, University of Chicago, Chicago, IL 60637, USA
| | - Jianjun Chen
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Chong-Zhi Wang
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Chun-Su Yuan
- Tang Center for Herbal Medicine Research, University of Chicago, Chicago, IL 60637, USA; Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA; Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|