1
|
Shen J, Zhou L, Ye K, Gong J, Wu F, Mo K, Zhu Y, Chen C, Zhan R. The role of SPI1/VSIG4/THBS1 on glioblastoma progression through modulation of the PI3K/AKT pathway. J Adv Res 2024:S2090-1232(24)00260-1. [PMID: 38960279 DOI: 10.1016/j.jare.2024.06.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024] Open
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) poses a significant challenge in terms of treatment due to its high malignancy, necessitating the identification of additional molecular targets. VSIG4, an oncogenic gene participates in tumor growth and migration in various cancer types. Nevertheless, the precise process through which VSIG4 facilitates the malignant progression of glioma remains to be elucidated. OBJECTIVES This research aims to explore the function and molecular mechanism involving VSIG4 in the malignant progression of glioma. METHODS The amount of VSIG4 was measured using qPCR, western blotting, and immunohistochemistry. Lentivirus infections were applied for upregulating or downregulating molecules within glioma cells. The incorporation of 5-ethynyl-20-deoxyuridine, Transwell, cell counting kit-8, and clone formation experiments, were applied to assess the biological functions of molecules on glioma cells. Dual luciferase reporter gene, RNA immunoprecipitation, and chromatin immunoprecipitation assays were used to explore the functional relationship among relevant molecules. RESULTS The upregulation of VSIG4 was observed in GBM tissues, indicating an adverse prognosis. Silencing VSIG4 in glioma cells resulted in a decrease in cell viability, invasion, proliferation, and tumorigenesis, an increase in cell apoptosis, and a stagnation in the cell cycle progression at the G0/G1 phase. Mechanistically, SPI1-mediated upregulation of VSIG4 expression led to binding between VSIG4 and THBS1 protein, ultimately facilitating the malignant progression of glioma cells through the activation of the PI3K/AKT pathway. The inhibited proliferative and invasive capabilities of glioma cells were reversed by overexpressing THBS1 following the knockdown of VSIG4. CONCLUSION Our findings provide evidence for the role of VSIG4 as an oncogene and reveal the previously unidentified contribution of the SPI1/VSIG4/THBS1 axis in the malignant progression of glioma. This signaling cascade enhances tumor growth and invasion by modulating the PI3K/AKT pathway. VSIG4 as a potential biomarker may be a viable strategy in the development of tailored molecular therapies for GBM.
Collapse
Affiliation(s)
- Jie Shen
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China; College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China.
| | - Lihui Zhou
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China; College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China.
| | - Ke Ye
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China; College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China.
| | - Jiangbiao Gong
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China.
| | - Fan Wu
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China.
| | - Kangnan Mo
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China.
| | - Yu Zhu
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China.
| | - Chao Chen
- Department of Head and Neck Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, PR China.
| | - Renya Zhan
- Department of Neurosurgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China; College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, PR China.
| |
Collapse
|
2
|
Alvarez-Arzola R, Oliver L, Messmer MM, Twum DYF, Lee KP, Muhitch JB, Mesa C, Abrams SI. A Bacterial and Ganglioside-based Nanoparticle Initiates Reprogramming of Macrophages and Promotes Antitumor Phenotypes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:475-486. [PMID: 38117752 DOI: 10.4049/jimmunol.2300256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 11/19/2023] [Indexed: 12/22/2023]
Abstract
Macrophages represent the most abundant immune component of the tumor microenvironment and often exhibit protumorigenic (M2-like) phenotypes that contribute to disease progression. Despite their generally accepted protumorigenic role, macrophages can also display tumoricidal (or M1-like) behavior, revealing that macrophages can be functionally reprogrammed, depending on the cues received within the tumor microenvironment. Moreover, such plasticity may be achieved by pharmacologic or biologic interventions. To that end, we previously demonstrated that a novel immunomodulator termed the "very small size particle" (VSSP) facilitates maturation of dendritic cells and differentiation of myeloid-derived suppressor cells to APCs with reduced suppressive activity in cancer models. VSSP was further shown to act in the bone marrow to drive the differentiation of progenitors toward monocytes, macrophages, and dendritic cells during emergency myelopoiesis. However, the underlying mechanisms for VSSP-driven alterations in myeloid differentiation and function remained unclear. In this study, in mouse models, we focused on macrophages and tested the hypothesis that VSSP drives macrophages toward M1-like functional states via IRF8- and PU.1-dependent mechanisms. We further hypothesized that such VSSP-mediated actions would be accompanied by enhanced antitumor responses. Overall, we showed that (1) VSSP drives naive or M2-derived macrophages to M1-like states, (2) the M1-like state induced by VSSP occurs via IRF8- and PU.1-dependent mechanisms, and (3) single-agent VSSP induces an antitumor response that is accompanied by alterations in the intratumoral myeloid compartment. These results provide a deeper mechanistic underpinning of VSSP and strengthen its use to drive M1-like responses in host defense, including cancer.
Collapse
Affiliation(s)
- Rydell Alvarez-Arzola
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | - Liliana Oliver
- Department of Immunoregulation, Immunology and Immunotherapy Direction, Center of Molecular Immunology, Havana, Cuba
| | | | - Danielle Y F Twum
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Kelvin P Lee
- IU Simon Comprehensive Cancer Center, Division of Hematology/Oncology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Jason B Muhitch
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Circe Mesa
- Innovative Immunotherapy Alliance S.A., Mariel, Artemisa, Cuba
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| |
Collapse
|
3
|
Liu X, Xu M, Jia W, Duan Y, Ma J, Tai W. PU.1 negatively regulates tumorigenesis in non-small-cell lung cancer. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2023; 40:79. [PMID: 36648591 DOI: 10.1007/s12032-023-01946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023]
Abstract
PU.1 is a key transcription factor that modulates hematopoietic cell differentiation and is involved in various physiological and pathological processes. PU.1 has been described to have multiple roles in a diverse range of cancers, but its contribution in non-small-cell lung cancer (NSCLC) has not been clearly elucidated. Fifty pairs of lung adenocarcinoma (LUAD) tissues and paraneoplastic tissues were collected. RT-qPCR assay was used to test PU.1 expression. Expression of PU.1 in LUAD cell lines and control cell lines was detected by RT-qPCR, and the role of PU.1 in LUAD was investigated by in vitro experiment. Levels of the major proteins in the apoptotic pathway were also detected by Western blot. The expression of PU.1 was remarkably downregulated in LUAD. Overexpression of PU.1 impaired the viability of LUAD cells as well as their metastatic function. In addition, PU.1 promoted apoptosis of LUAD cells by decreasing Bcl2 and increasing Bax/Bak1 expression. PU.1 plays an inhibitory role in LUAD, mainly promoting the apoptosis of LUAD cells.
Collapse
Affiliation(s)
- Xiaoxiao Liu
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Muli Xu
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wanting Jia
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yu Duan
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiaxuan Ma
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenlin Tai
- Department of Clinical Laboratory, Yunnan Molecular Diagnostic Center, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
4
|
Sunthankar KI, Jenkins MT, Cote CH, Patel SB, Welner RS, Ferrell PB. Isocitrate dehydrogenase mutations are associated with altered IL-1β responses in acute myeloid leukemia. Leukemia 2022; 36:923-934. [PMID: 34857894 PMCID: PMC9066619 DOI: 10.1038/s41375-021-01487-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/08/2021] [Accepted: 11/22/2021] [Indexed: 12/20/2022]
Abstract
Mutations in isocitrate dehydrogenase 2 (IDH2) have been noted to impact cellular differentiation in addition to DNA and histone methylation. However, little is known about the impact of IDH2 mutations on intracellular signaling. Using an isogenic cell line model, we investigated both differentiation and signaling responses in IDH2 mutant cells and show augmented responses to inflammatory immune ligands. Using phospho-specific flow and mass cytometry, we demonstrate IDH2 mutant cells were significantly more sensitive to IL-1β at multiple downstream readouts. Further, bulk RNA sequencing confirmed increases in cytokine-related signaling pathways and NF-κB target genes. Single-cell RNA sequencing of unstimulated and stimulated cells confirmed altered IL-1β transcriptional responses in the IDH2 mutant cells. Targeted inhibition of the IKK complex reduced IL-1β responses and induced cell death in primary IDH-mutated leukemia samples. Together, these results confirm altered IL-1β signaling in IDH2 mutant cells and identify this pathway as a potential therapeutic target.
Collapse
Affiliation(s)
- Kathryn I. Sunthankar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew T. Jenkins
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Candace H. Cote
- University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Sweta B. Patel
- Division of Hematology/Oncology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Robert S. Welner
- Division of Hematology/Oncology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - P. Brent Ferrell
- Division of Hematology/Oncology, Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Corresponding Author (PBF),
| |
Collapse
|
5
|
Kaempferol sensitizes tumor necrosis factor-related apoptosis-inducing ligand-resistance chronic myelogenous leukemia cells to apoptosis. Mol Biol Rep 2021; 49:19-29. [PMID: 34820749 DOI: 10.1007/s11033-021-06778-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND The tumor necrosis factor (TNF)-related apoptosis-inducing ligand, TRAIL, an apoptosis-inducing cytokine, has attracted much attention in the treatment of cancer for its selective toxicity to malignant rather than normal cells. However, the apoptosis-inducing ability of TRAIL is weaker than expected primarily due to cancer cell resistance. As one of the dietary flavonoids, kaempferol, has been shown to be antiproliferative and might have a protective effect against TRAIL resistance, particularly for hematologic malignancies. METHODS AND RESULTS Here, we studied the potential of kaempferol to enhance the TRAIL-induced cytotoxicity and apoptosis in human chronic myelogenous leukemia (CML) cell line K-562, as well as the expression of specific genes with impact on TRAIL signal regulation. Analysis of flowcytometry data showed that treatment with kaempferol did enhance sensitivity of CML cells to pro-apoptotic effects of anti-TRAIL antibody. Although the gene expression levels were heterogeneous, cFLIP, cIAP1 and cIAP2 expression were generally downregulated where co-treatment of kaempferol and TRAIL was employed and these effects appeared to be dose-dependent. We further demonstrated that the expression of death receptors 4 and 5 tended to increase subsequent to the combination treatment. CONCLUSIONS Consequently, it is reasonable to conclude that sensitization of chronic leukemia cells to TRAIL by kaempferol in vitro should be considered as a way of focusing clinical attention on leukemia therapy.
Collapse
|
6
|
Romine KA, Nechiporuk T, Bottomly D, Jeng S, McWeeney SK, Kaempf A, Corces MR, Majeti R, Tyner JW. Monocytic differentiation and AHR signaling as Primary Nodes of BET Inhibitor Response in Acute Myeloid Leukemia. Blood Cancer Discov 2021; 2:518-531. [PMID: 34568834 DOI: 10.1158/2643-3230.bcd-21-0012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
To understand mechanisms of response to BET inhibitors (BETi), we mined the Beat AML functional genomic dataset and performed genome-wide CRISPR screens on BETi- sensitive and BETi- resistant AML cells. Both strategies revealed regulators of monocytic differentiation, SPI1, JUNB, FOS, and aryl-hydrocarbon receptor signaling (AHR/ARNT), as determinants of BETi response. AHR activation synergized with BETi while inhibition antagonized BETi-mediated cytotoxicity. Consistent with BETi sensitivity dependence on monocytic differentiation, ex vivo sensitivity to BETi in primary AML patient samples correlated with higher expression of monocytic markers CSF1R, LILRs, and VCAN. In addition, HL-60 cell line differentiation enhanced its sensitivity to BETi. Further, screens to rescue BETi sensitivity identified BCL2 and CDK6 as druggable vulnerabilities. Finally, monocytic AML patient samples refractory to venetoclax ex vivo were significantly more sensitive to combined BETi + venetoclax. Together, our work highlights mechanisms that could predict BETi response and identifies combination strategies to overcome resistance.
Collapse
Affiliation(s)
- Kyle A Romine
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Tamilla Nechiporuk
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Daniel Bottomly
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Portland, OR, USA
| | - Sophia Jeng
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Oregon Clinical and Translational Research Institute, Portland, OR, USA
| | - Shannon K McWeeney
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Portland, OR, USA.,Oregon Clinical and Translational Research Institute, Portland, OR, USA
| | - Andy Kaempf
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Biostatistics Shared Resource, Portland, OR, USA
| | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA.,Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Ravindra Majeti
- Department of Medicine, Division of Hematology, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey W Tyner
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA.,Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA.,Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
7
|
Naimi A, Safaei S, Entezari A, Solali S, Hassanzadeh A. Knockdown of Enhancer of Zeste Homolog 2 Affects mRNA Expression of Genes Involved in the Induction of Resistance to Apoptosis in MOLT-4 Cells. Anticancer Agents Med Chem 2021; 20:571-579. [PMID: 32000648 DOI: 10.2174/1871520620666200130091955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 11/06/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND The Enhancer of Zeste Homolog 2 (EZH2) is a subunit of the polycomb repressive complex 2 that silences the gene transcription via H3K27me3. Previous studies have shown that EZH2 has an important role in the induction of the resistance against the Tumor necrosis factor-Related Apoptosis-Inducing Ligand (TRAIL)-Induced Apoptosis (TIA) in some leukemia cells. OBJECTIVE The aim of this study was to determine the effect of silencing EZH2 gene expression using RNA interference on the expression of death receptors 4 and 5 (DR4/5), Preferentially expressed Antigen in Melanoma (PRAME), and TRAIL human lymphoid leukemia MOLT-4 cells. METHODS Quantitative RT-PCR was used to detect the EZH2 expression and other candidate genes following the siRNA knockdown in MOLT-4 cells. The toxicity of the EZH2 siRNA was evaluated using Annexin V/PI assay following the transfection of the cells by 80 pM EZH2 siRNA at 48 hours. RESULTS Based on the flow-cytometry results, the EZH2 siRNA had no toxic effects on MOLT-4 cells. Also, the EZH2 inhibition increased the expression of DR4/5 but reduced the PRAME gene expression at the mRNA levels. Moreover, the EZH2 silencing could not change the TRAIL mRNA in the transfected cells. CONCLUSION Our results revealed that the down-regulation of EZH2 in MOLT-4 cells was able to affect the expression of important genes involved in the induction of resistance against TIA. Hence, we suggest that the silencing of EZH2 using RNA interference can be an effective and safe approach to help defeat the MOLT-4 cell resistance against TIA.
Collapse
Affiliation(s)
- Adel Naimi
- Cellular and Molecular Research Center, Sabzevar University of Medical Science, Sabzevar, Iran.,Department of Medical Laboratory Sciences, Faculty of Paramedicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Atefeh Entezari
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hassanzadeh
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Continuous NF-κB pathway inhibition promotes expansion of human phenotypical hematopoietic stem/progenitor cells through metabolism regulation. Exp Cell Res 2021; 399:112468. [PMID: 33428903 DOI: 10.1016/j.yexcr.2020.112468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 12/24/2020] [Accepted: 12/27/2020] [Indexed: 01/24/2023]
Abstract
Hematopoietic stem/progenitor cells (HSPCs) ex vivo expansion is critical in facilitating their widespread clinical application. NF-κB pathway is implicated in the energy homeostasis and metabolic adaptation. To explore the effect of NF-κB pathway on the ex vivo HSPC expansion and metabolism, the 50 nM-1 μM inhibitor of NF-κB pathway TPCA-1 was used to expand cord blood derived CD34+ cells in serum-free culture. The expansion folds, function, mitochondrial profile and metabolism of HSPCs were determined. After 10 days of culture with 100 nM TPCA-1, the expansion of total cells, CD34+CD38- cells, and CD34+CD38-CD45RA-CD90+CD49f+ cells were significantly increased compared to the cytokine priming alone. Notably, TPCA-1 treatment generated ~ 2-fold greater percentage of CD34+EPCR+ and CD34+CD38-CD45RA-CD90+CD49f+ cells compared to cytokine only conditions. Moreover, TPCA-1 expanded CD34+ cells displayed enhanced serial colonies forming potential and secondary expansion capability. NF-κB inhibition increased the expression of self-renewal related genes, while downregulated the expression of mitochondrial biogenesis regulator (Pgc1α) and mitochondrial chaperones and proteases (ClpP, Hsp10, Hsp60). Mitochondrial mass and membrane potential were markedly decreased with TPCA-1 treatment, leading to the reduced mitochondrial reactive oxygen species (ROS) level in HSPCs. NF-κB inhibition displayed augmented glycolysis rate with compromising mitochondrial metabolism. This study demonstrated that NF-κB pathway inhibition improved glycolysis and limited ROS production that promoted the ex vivo expansion and maintenance of functional HSPCs.
Collapse
|
9
|
Khan I, Eklund EE, Gartel AL. Therapeutic Vulnerabilities of Transcription Factors in AML. Mol Cancer Ther 2020; 20:229-237. [PMID: 33158995 DOI: 10.1158/1535-7163.mct-20-0115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/13/2020] [Accepted: 10/19/2020] [Indexed: 11/16/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by impaired myeloid lineage differentiation, uncontrolled proliferation, and inhibition of proapoptotic pathways. In spite of a relatively homogeneous clinical disease presentation, risk of long-term survival in AML varies from 20% to 80% depending on molecular disease characteristics. In recognition of the molecular heterogeneity of AML, the European Leukemia Net (ELN) and WHO classification systems now incorporate cytogenetics and increasing numbers of gene mutations into AML prognostication. Several of the genomic AML subsets are characterized by unique transcription factor alterations that are highlighted in this review. There are many mechanisms of transcriptional deregulation in leukemia. We broadly classify transcription factors based on mechanisms of transcriptional deregulation including direct involvement of transcription factors in recurrent translocations, loss-of-function mutations, and intracellular relocalization. Transcription factors, due to their pleiotropic effects, have been attractive but elusive targets. Indirect targeting approaches include inhibition of upstream kinases such as TAK1 for suppression of NFκB signaling and downstream effectors such as FGF signaling in HOXA-upregulated leukemia. Other strategies include targeting scaffolding proteins like BrD4 in the case of MYC or coactivators such as menin to suppress HOX expression; disrupting critical protein interactions in the case of β-catenin:TCF/LEF, and preventing transcription factor binding to DNA as in the case of PU.1 or FOXM1. We comprehensively describe the mechanism of deregulation of transcription factors in genomic subsets of AML, consequent pathway addictions, and potential therapeutic strategies.
Collapse
Affiliation(s)
- Irum Khan
- Department of Medicine, University of Illinois, Chicago, Illinois
| | - Elizabeth E Eklund
- Feinberg School of Medicine and Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois.,Jesse Brown VA Medical Center, Chicago, Illinois
| | - Andrei L Gartel
- Department of Medicine, University of Illinois, Chicago, Illinois.
| |
Collapse
|
10
|
Meydan C, Madrer N, Soreq H. The Neat Dance of COVID-19: NEAT1, DANCR, and Co-Modulated Cholinergic RNAs Link to Inflammation. Front Immunol 2020; 11:590870. [PMID: 33163005 PMCID: PMC7581732 DOI: 10.3389/fimmu.2020.590870] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022] Open
Abstract
The COVID-19 pandemic exerts inflammation-related parasympathetic complications and post-infection manifestations with major inter-individual variability. To seek the corresponding transcriptomic origins for the impact of COVID-19 infection and its aftermath consequences, we sought the relevance of long and short non-coding RNAs (ncRNAs) for susceptibility to COVID-19 infection. We selected inflammation-prone men and women of diverse ages among the cohort of Genome Tissue expression (GTEx) by mining RNA-seq datasets from their lung, and blood tissues, followed by quantitative qRT-PCR, bioinformatics-based network analyses and thorough statistics compared to brain cell culture and infection tests with COVID-19 and H1N1 viruses. In lung tissues from 57 inflammation-prone, but not other GTEx donors, we discovered sharp declines of the lung pathology-associated ncRNA DANCR and the nuclear paraspeckles forming neuroprotective ncRNA NEAT1. Accompanying increases in the acetylcholine-regulating transcripts capable of controlling inflammation co-appeared in SARS-CoV-2 infected but not H1N1 influenza infected lung cells. The lung cells-characteristic DANCR and NEAT1 association with inflammation-controlling transcripts could not be observed in blood cells, weakened with age and presented sex-dependent links in GTEx lung RNA-seq dataset. Supporting active involvement in the inflammatory risks accompanying COVID-19, DANCR's decline associated with decrease of the COVID-19-related cellular transcript ACE2 and with sex-related increases in coding transcripts potentiating acetylcholine signaling. Furthermore, transcription factors (TFs) in lung, brain and cultured infected cells created networks with the candidate transcripts, indicating tissue-specific expression patterns. Supporting links of post-infection inflammatory and cognitive damages with cholinergic mal-functioning, man and woman-originated cultured cholinergic neurons presented differentiation-related increases of DANCR and NEAT1 targeting microRNAs. Briefly, changes in ncRNAs and TFs from inflammation-prone human lung tissues, SARS-CoV-2-infected lung cells and man and woman-derived differentiated cholinergic neurons reflected the inflammatory pathobiology related to COVID-19. By shifting ncRNA differences into comparative diagnostic and therapeutic profiles, our RNA-sequencing based Resource can identify ncRNA regulating candidates for COVID-19 and its associated immediate and predicted long-term inflammation and neurological complications, and sex-related therapeutics thereof. Our findings encourage diagnostics of involved tissue, and further investigation of NEAT1-inducing statins and anti-cholinergic medications in the COVID-19 context.
Collapse
Affiliation(s)
- Chanan Meydan
- Department of Internal Medicine, Mayanei Hayeshua Medical Center, Bnei Brak, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Central District, Leumit Health Services, Tel Aviv, Israel
| | - Nimrod Madrer
- The Department of Biological Chemistry and The Edmond and Lilly Safra Center for Brain Sciences, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hermona Soreq
- The Department of Biological Chemistry and The Edmond and Lilly Safra Center for Brain Sciences, The Alexander Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
11
|
Hotta M, Sakatani T, Ishino K, Wada R, Kudo M, Yokoyama Y, Yamada T, Yoshida H, Naito Z. Farnesoid X receptor induces cell death and sensitizes to TRAIL-induced inhibition of growth in colorectal cancer cells through the up-regulation of death receptor 5. Biochem Biophys Res Commun 2019; 519:824-831. [DOI: 10.1016/j.bbrc.2019.09.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/09/2019] [Indexed: 01/22/2023]
|
12
|
Fei HR, Yuan C, Wang GL, Zhao Y, Li ZJ, Du X, Wang FZ. Caudatin potentiates the anti-tumor effects of TRAIL against human breast cancer by upregulating DR5. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152950. [PMID: 31102888 DOI: 10.1016/j.phymed.2019.152950] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/04/2019] [Accepted: 05/05/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND The ability of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to preferentially induce apoptosis in transformed cells while sparing most normal cells is well established. However, the intrinsic and acquired resistance of tumors to TRAIL-induced apoptosis limits its therapeutic applicability. PURPOSE We investigated the effect of caudatin, a species of C-21 steroidal glycosides isolated from the roots of Cynanchum auriculatum, on TRAIL-induced apoptosis in human breast cancer cells. METHODS Cell growth inhibition was evaluated by the CCK-8 assay. The cell cycle distribution was assessed by propidium iodide flow cytometry. Apoptosis was determined by TUNEL staining. Protein expression was detected by western blotting analysis. RESULTS Caudatin enhanced TRAIL-induced apoptosis in human breast cancer cells. This sensitization was achieved by upregulating death receptor 5 (DR5). Knockdown of DR5 abolished the enhancing effect of caudatin on TRAIL responses. The caudatin-induced upregulation of DR5 was accompanied by increased expression of CHOP and phosphorylation of p38 MAPK and JNK. CHOP knockdown blocked caudatin-upregulated DR5 expression. Moreover, cotreatment of breast cancer cells with p38 MAPK and JNK inhibitors significantly counteracted the caudatin-induced expression of DR5. CONCLUSION Our results showed that caudatin sensitized breast cancer cells to TRAIL-induced apoptosis through activation of CHOP, p38 MAPK and JNK-mediated upregulation of DR5 expression. The combination of TRAIL and caudatin may be a promising therapeutic approach for the treatment of breast cancer.
Collapse
Affiliation(s)
- Hong-Rong Fei
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Chuang Yuan
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Gui-Ling Wang
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Ying Zhao
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Zhao-Jun Li
- School of Pharmacy, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China; School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Xin Du
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China
| | - Feng-Ze Wang
- School of Life Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, PR China.
| |
Collapse
|
13
|
The Role of Forkhead Box Proteins in Acute Myeloid Leukemia. Cancers (Basel) 2019; 11:cancers11060865. [PMID: 31234353 PMCID: PMC6627614 DOI: 10.3390/cancers11060865] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/29/2019] [Accepted: 06/18/2019] [Indexed: 12/20/2022] Open
Abstract
Forkhead box (FOX) proteins are a group of transcriptional factors implicated in different cellular functions such as differentiation, proliferation and senescence. A growing number of studies have focused on the relationship between FOX proteins and cancers, particularly hematological neoplasms such as acute myeloid leukemia (AML). FOX proteins are widely involved in AML biology, including leukemogenesis, relapse and drug sensitivity. Here we explore the role of FOX transcription factors in the major AML entities, according to "The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia", and in the context of the most recurrent gene mutations identified in this heterogeneous disease. Moreover, we report the new evidences about the role of FOX proteins in drug sensitivity, mechanisms of chemoresistance, and possible targeting for personalized therapies.
Collapse
|
14
|
Jo EB, Lee YS, Lee H, Park JB, Park H, Choi YL, Hong D, Kim SJ. Combination therapy with c-met inhibitor and TRAIL enhances apoptosis in dedifferentiated liposarcoma patient-derived cells. BMC Cancer 2019; 19:496. [PMID: 31126284 PMCID: PMC6534902 DOI: 10.1186/s12885-019-5713-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 05/14/2019] [Indexed: 12/12/2022] Open
Abstract
Background Liposarcoma (LPS) is a tumor derived from adipose tissue, and has the highest incidence among soft tissue sarcomas. Dedifferentiated liposarcoma (DDLPS) is a malignant tumor with poor prognosis. Recurrence and metastasis rates in LPS remain high even after chemotherapy and radiotherapy following complete resection. Therefore, the development of advanced treatment strategies for LPS is required. In the present study, we investigated the effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment, and of combination treatment using TRAIL and a c-Met inhibitor on cell viability and apoptosis in LPS and DDLPS cell lines of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment, and of combination treatment using TRAIL and a c-Met inhibitor. Methods We analyzed cell viability after treatment with TRAIL and a c-Met inhibitor by measuring CCK8 and death receptor 5 (DR5) expression levels via fluorescence activated cell sorting (FACS) in both sarcoma cell lines and DDLPS patient-derived cells (PDCs). Moreover, we validated the effects of TRAIL alone and in combination with c-Met inhibitor on apoptosis in LPS cell lines and DDLPS PDCs via FACS. Results Our results revealed that combination treatment with a c-Met inhibitor and human recombinant TRAIL (rhTRAIL) suppressed cell viability and induced cell death in both sarcoma cell lines and DDLPS PDCs, which showed varying sensitivities to rhTRAIL alone. Also, we confirmed that treatment with a c-Met inhibitor upregulated DR5 levels in sarcoma cell lines and DDLPS PDCs. In both TRAIL-susceptible and TRAIL-resistant cells subjected to combination treatment, promotion of apoptosis was dependent on DR5 upregulation. Conclusion From these results, our findings validated that DR5 up-regulation caused by combination therapy with a c-Met inhibitor and rhTRAIL enhanced TRAIL sensitization and promoted apoptosis. We propose the use of this approach to overcome TRAIL resistance and serve as a novel treatment strategy for clinical trials. Electronic supplementary material The online version of this article (10.1186/s12885-019-5713-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Eun Byeol Jo
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Samsung Advanced Institute for Health Sciences and Technology, SKKU, Seoul, Republic of Korea
| | - Young Sang Lee
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Samsung Advanced Institute for Health Sciences and Technology, SKKU, Seoul, Republic of Korea
| | - Hyunjoo Lee
- Personalized Medicine, Children's Cancer Institute Australia, Sydney, NSW, Australia
| | - Jae Berm Park
- Department of Surgery, Samsung Medical Center, SungKyunKwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Hyojun Park
- Department of Surgery, Samsung Medical Center, SungKyunKwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea
| | - Yoon-La Choi
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.,Department of Pathology, Samsung Medical Center, Seoul, Republic of Korea
| | - Doopyo Hong
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
| | - Sung Joo Kim
- Sarcoma Research Center, Samsung Biomedical Research Institute, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea. .,Samsung Advanced Institute for Health Sciences and Technology, SKKU, Seoul, Republic of Korea. .,Department of Surgery, Samsung Medical Center, SungKyunKwan University School of Medicine, 81 Irwon-ro, Gangnam-gu, Seoul, 06351, South Korea.
| |
Collapse
|
15
|
Staniek J, Lorenzetti R, Heller B, Janowska I, Schneider P, Unger S, Warnatz K, Seidl M, Venhoff N, Thiel J, Smulski CR, Rizzi M. TRAIL-R1 and TRAIL-R2 Mediate TRAIL-Dependent Apoptosis in Activated Primary Human B Lymphocytes. Front Immunol 2019; 10:951. [PMID: 31114586 PMCID: PMC6503035 DOI: 10.3389/fimmu.2019.00951] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 04/12/2019] [Indexed: 11/28/2022] Open
Abstract
The maintenance of B cell homeostasis requires a tight control of B cell generation, survival, activation, and maturation. In lymphocytes upon activation, increased sensitivity to apoptotic signals helps controlling differentiation and proliferation. The death receptor Fas is important in this context because genetic Fas mutations in humans lead to an autoimmune lymphoproliferative syndrome that is similar to lymphoproliferation observed in Fas-deficient mice. In contrast, the physiological role of TNF-related apoptosis-inducing ligand receptors (TRAIL-Rs) in humans has been poorly studied so far. Indeed, most studies have focused on tumor cell lines and on mouse models whose results are difficult to transpose to primary human B cells. In the present work, the expression of apoptosis-inducing TRAIL-R1 and TRAIL-R2 and of the decoy receptors TRAIL-R3 and TRAIL-R4 was systematically studied in all developmental stages of peripheral B cells isolated from the blood and secondary lymphoid organs. Expression of TRAIL-Rs is modulated along development, with highest levels observed in germinal center B cells. In addition, T-dependent and T-independent signals elicited induction of TRAIL-Rs with distinct kinetics, which differed among B cell subpopulations: switched memory cells rapidly upregulated TRAIL-R1 and -2 upon activation while naïve B cells only reached similar expression levels at later time points in culture. Increased expression of TRAIL-R1 and -2 coincided with a caspase-3-dependent sensitivity to TRAIL-induced apoptosis in activated B cells but not in freshly isolated resting B cells. Finally, both TRAIL-R1 and TRAIL-R2 could signal actively and both contributed to TRAIL-induced apoptosis. In conclusion, this study provides a systematic analysis of the expression of TRAIL-Rs in human primary B cells and of their capacity to signal and induce apoptosis. This dataset forms a basis to further study and understand the dysregulation of TRAIL-Rs and TRAIL expression observed in autoimmune diseases. Additionally, it will be important to foresee potential bystander immunomodulation when TRAIL-R agonists are used in cancer treatment.
Collapse
Affiliation(s)
- Julian Staniek
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Raquel Lorenzetti
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Bianca Heller
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Iga Janowska
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Pascal Schneider
- Department of Biochemistry, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Susanne Unger
- Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Maximilian Seidl
- Department of Pathology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Nils Venhoff
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Jens Thiel
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Cristian Roberto Smulski
- Medical Physics Department, Centro Atómico Bariloche, Comisión Nacional de Energía Atómica (CNEA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Carlos de Bariloche, Argentina
| | - Marta Rizzi
- Clinic for Rheumatology and Clinical Immunology, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, Freiburg, Germany
| |
Collapse
|
16
|
Castelli G, Pelosi E, Testa U. Emerging Therapies for Acute Myelogenus Leukemia Patients Targeting Apoptosis and Mitochondrial Metabolism. Cancers (Basel) 2019; 11:E260. [PMID: 30813354 PMCID: PMC6406361 DOI: 10.3390/cancers11020260] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 02/14/2019] [Indexed: 02/06/2023] Open
Abstract
Acute Myelogenous Leukemia (AML) is a malignant disease of the hematopoietic cells, characterized by impaired differentiation and uncontrolled clonal expansion of myeloid progenitors/precursors, resulting in bone marrow failure and impaired normal hematopoiesis. AML comprises a heterogeneous group of malignancies, characterized by a combination of different somatic genetic abnormalities, some of which act as events driving leukemic development. Studies carried out in the last years have shown that AML cells invariably have abnormalities in one or more apoptotic pathways and have identified some components of the apoptotic pathway that can be targeted by specific drugs. Clinical results deriving from studies using B-cell lymphoma 2 (BCL-2) inhibitors in combination with standard AML agents, such as azacytidine, decitabine, low-dose cytarabine, provided promising results and strongly support the use of these agents in the treatment of AML patients, particularly of elderly patients. TNF-related apoptosis-inducing ligand (TRAIL) and its receptors are frequently deregulated in AML patients and their targeting may represent a promising strategy for development of new treatments. Altered mitochondrial metabolism is a common feature of AML cells, as supported through the discovery of mutations in the isocitrate dehydrogenase gene and in mitochondrial electron transport chain and of numerous abnormalities of oxidative metabolism existing in AML subgroups. Overall, these observations strongly support the view that the targeting of mitochondrial apoptotic or metabolic machinery is an appealing new therapeutic perspective in AML.
Collapse
Affiliation(s)
- Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
17
|
Nakhost B, Nasiri M, Karimi M, Montazeri S. Down-Regulation of PU.1 Gene in Pediatric Acute Lymphoblastic Leukemia Patients from South of Iran. Int J Hematol Oncol Stem Cell Res 2019; 13:20-24. [PMID: 31205624 PMCID: PMC6557971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Acute lymphoblastic leukemia (ALL) is resulted from the infiltration of high amount of non-differentiated cells in bone marrow. Differentiation of the hematopoietic stem cells into specific cell lineage occurs through a highly regulated pathway which is mainly monitored during transcription step. Expression level and pattern of transcription factors e.g. PU.1 determine fate and developmental phases in this pathway. This study was performed to evaluate the expression level of the PU.1 gene in a group of children suffering from ALL. Materials and Methods: The mRNA expression level of the PU.1 gene was compared between 30 children diagnosed as new cases of ALL and 30 sex- and gender-matched healthy children in the present case-control study. The quantitative real time PCR (qRT-PCR) was used to determine the level of PU.1 gene expression. The data were analyzed using Graph Pad Prism statistical software. Results: The mRNA level of the PU.1 gene was significantly lower in the blood samples of the ALL patients compared to the controls (p= 0.002). Conclusion: The results of the study indicated that the PU.1 gene seemed to have key roles in the differentiation pathway of blood cells.
Collapse
Affiliation(s)
- Bita Nakhost
- Department of Biology, Islamic Azad University, Arsanjan Branch, Arsanjan, Iran
| | - Mahboobeh Nasiri
- Department of Biology, Islamic Azad University, Arsanjan Branch, Arsanjan, Iran
| | - Mehran Karimi
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Somayeh Montazeri
- Thalassemia and Hemophilia Genetic, PND Research Center, Dastgheib Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
18
|
You C, Zhang S, Sun Y, Zhang S, Tang G, Tang F, Liu X, Xiao Y, Zhang J, Gong Y, Xie C. β-catenin decreases acquired TRAIL resistance in non-small-cell lung cancer cells by regulating the redistribution of death receptors. Int J Oncol 2018; 53:2258-2268. [PMID: 30132512 DOI: 10.3892/ijo.2018.4529] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/25/2018] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor‑related apoptosis‑inducing ligand (TRAIL) exhibits antitumor activity in various types of tumor cell and tumor‑bearing animals. However, acquired TRAIL resistance is a common issue that restricts its clinical application. Previous studies have revealed that β‑catenin is associated with TRAIL resistance in melanoma and colorectal tumors. In the present study, an acquired‑resistance non‑small‑cell lung cancer (NSCLC) cell line (H460‑TR) was established from parental TRAIL‑sensitive H460 cells using a gradient ascent model (8‑256 ng/ml TRAIL). Cellular FADD‑like interleukin‑1β converting enzyme inhibitory protein and Mcl‑1 were upregulated and the cell surface distribution of death receptor (DR)4 and DR5 was downregulated in H460‑TR cells compared with the parental H460 cells. The results of reverse transcription‑quantitative polymerase chain reaction and western blot analysis indicated that H460 cells expressed increased levels of β‑catenin and were more sensitive to TRAIL compared with H460‑TR cells. β‑catenin‑knockdown in H460 cells decreased their sensitivity to TRAIL, while upregulation of β‑catenin expression in H460‑TR cells increased their sensitivity to TRAIL, increased the cell surface distribution of DRs and activated caspase‑3/8. Taken together, the results of the present study suggest that β‑catenin impairs acquired TRAIL resistance in NSCLC cells by promoting the redistribution of DR4 and DR5 to the cytomembrane, and inducing TRAIL‑mediated cell apoptosis via caspase‑3/8 activation.
Collapse
Affiliation(s)
- Chengcheng You
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Shimin Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Yingming Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Shiyu Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Guiliang Tang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Fang Tang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Xuefeng Liu
- Department of Pathology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington, DC, USA
| | - Yu Xiao
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Junhong Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
19
|
Ntari L, Sakkou M, Chouvardas P, Mourouzis I, Prados A, Denis MC, Karagianni N, Pantos C, Kollias G. Comorbid TNF-mediated heart valve disease and chronic polyarthritis share common mesenchymal cell-mediated aetiopathogenesis. Ann Rheum Dis 2018; 77:926-934. [PMID: 29475857 PMCID: PMC5965351 DOI: 10.1136/annrheumdis-2017-212597] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 12/29/2017] [Accepted: 01/25/2018] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Patients with rheumatoid arthritis and spondyloarthritisshow higher mortality rates, mainly caused by cardiac comorbidities. The TghuTNF (Tg197) arthritis model develops tumour necrosis factor (TNF)-driven and mesenchymalsynovial fibroblast (SF)-dependent polyarthritis. Here, we investigate whether this model develops, similarly to human patients, comorbid heart pathology and explore cellular and molecular mechanisms linking arthritis to cardiac comorbidities. METHODS Histopathological analysis and echocardiographic evaluation of cardiac function were performed in the Tg197 model. Valve interstitial cells (VICs) were targeted by mice carrying the ColVI-Cretransgene. Tg197 ColVI-Cre Tnfr1fl/fl and Tg197 ColVI-Cre Tnfr1cneo/cneo mutant mice were used to explore the role of mesenchymal TNF signalling in the development of heart valve disease. Pathogenic VICs and SFs were further analysed by comparative RNA-sequencing analysis. RESULTS Tg197 mice develop left-sided heart valve disease, characterised by valvular fibrosis with minimal signs of inflammation. Thickened valve areas consist almost entirely of hyperproliferative ColVI-expressing mesenchymal VICs. Development of pathology results in valve stenosis and left ventricular dysfunction, accompanied by arrhythmic episodes and, occasionally, valvular regurgitation. TNF dependency of the pathology was indicated by disease modulation following pharmacological inhibition or mesenchymal-specific genetic ablation or activation of TNF/TNFR1 signalling. Tg197-derived VICs exhibited an activated phenotype ex vivo, reminiscent of the activated pathogenic phenotype of Tg197-derived SFs. Significant functional similarities between SFs and VICs were revealed by RNA-seq analysis, demonstrating common cellular mechanisms underlying TNF-mediated arthritides and cardiac comorbidities. CONCLUSIONS Comorbidheart valve disease and chronic polyarthritis are efficiently modelled in the Tg197 arthritis model and share common TNF/TNFR1-mediated, mesenchymal cell-specific aetiopathogenic mechanisms.
Collapse
Affiliation(s)
- Lydia Ntari
- Institute of Immunology, Biomedical Sciences Research Center (BSRC), ‘Alexander Fleming’, Vari, Greece
- Faculty of Medicine, University of Crete, Heraklion, Greece
| | - Maria Sakkou
- Institute of Immunology, Biomedical Sciences Research Center (BSRC), ‘Alexander Fleming’, Vari, Greece
| | - Panagiotis Chouvardas
- Institute of Immunology, Biomedical Sciences Research Center (BSRC), ‘Alexander Fleming’, Vari, Greece
- Department of Physiology, School of Medicine, National Kapodistrian University, Athens, Greece
| | - Iordanis Mourouzis
- Department of Pharmacology, School of Medicine, National Kapodistrian University, Athens, Greece
| | - Alejandro Prados
- Institute of Immunology, Biomedical Sciences Research Center (BSRC), ‘Alexander Fleming’, Vari, Greece
| | | | | | - Constantinos Pantos
- Department of Pharmacology, School of Medicine, National Kapodistrian University, Athens, Greece
| | - George Kollias
- Institute of Immunology, Biomedical Sciences Research Center (BSRC), ‘Alexander Fleming’, Vari, Greece
- Department of Physiology, School of Medicine, National Kapodistrian University, Athens, Greece
| |
Collapse
|
20
|
Yang Y, Wang X, Zhang Y, Yuan W. Hepatitis B virus X protein and proinflammatory cytokines synergize to enhance TRAIL-induced apoptosis of renal tubular cells by upregulation of DR4. Int J Biochem Cell Biol 2018; 97:62-72. [PMID: 29432906 DOI: 10.1016/j.biocel.2018.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/14/2018] [Accepted: 02/06/2018] [Indexed: 01/07/2023]
Abstract
Persistent infection with hepatitis B virus (HBV) may lead to HBV-associated glomerulonephritis (HBV-GN). Presence of HBV-DNA and -RNA in renal tubular epithelial cells (RTECs) suggests direct virus-induced injury. Increase in proinflammatory cytokines is also observed under these conditions. Apoptosis by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) plays a significant role in the pathogenesis of HBV-infections. However, the effects of HBV X protein (HBx) on TRAIL-induced apoptosis of RTECs especially under certain inflammatory conditions remain obscure. Here, we show that HBx synergizes with proinflammatory cytokines to significantly increase TRAIL-induced apoptosis of RTECs. HBx markedly up-regulates death receptor-4 (DR4) expression by enhancing the activation of nuclear factor-kappa B (NF-κB) in the presence of proinflammatory cytokines. Dramatic increase in DR4 expression leads to the sensitization of RTECs to TRAIL-induced apoptosis. Furthermore, in patients with HBV-GN, DR4 expression in the kidneys is significantly elevated and is positively correlated with the HBx and proinflammatory cytokines expression. These findings provide a novel insight into the underlying mechanisms of renal tubule lesions induced by HBx in HBV-GN.
Collapse
Affiliation(s)
- Yitong Yang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xuan Wang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yueyue Zhang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Weijie Yuan
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|