1
|
Varisli L, Zoumpourlis P, Spandidos DA, Zoumpourlis V, Vlahopoulos S. ALDH1A1 in breast cancer: A prospective target to overcome therapy resistance (Review). Oncol Lett 2025; 29:213. [PMID: 40093866 PMCID: PMC11905208 DOI: 10.3892/ol.2025.14959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
The expression of cytosolic aldehyde dehydrogenases (ALDHs), which mediate the last step in the pathway of the synthesis of all-trans retinoic acid, is dysregulated in various types of human cancer, and has been associated with the development of cancer stem cells (CSCs) in solid tumors and hematological malignancies. CSCs are considered a minor fraction of cancer cells with the capacity to initiate neoplastic tumors. ALDH1A1 serves a crucial role in the emergence of the CSC phenotype, induces the malignant behavior of cancer cells and promotes treatment resistance. Notably, ALDH1A1-induced therapy resistance is not exclusive to just one group of drugs, but affects diverse types of drugs that use different mechanisms to kill cells. This diversity of drug resistance-inducing effects is associated with the stemness-supporting functions of ALDH1A1. The inhibition of ALDH1A1 activity using chemicals or the depletion of ALDH1A1 via genetic approaches, such as the use of small interfering RNA, can overcome diverse pathways of therapy resistance. In the context of breast cancer, it is critical that only a fraction of malignant cells are expected to manifest stem-like features, which include increased expression of ALDH1A1. From the angle of disease prognosis, the extent of the association of ALDH1A1 with increased malignant behavior and drug resistance remains to be determined through the application of cutting-edge methods that detect the expression of tracked biomarkers within tumors.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
2
|
Vlahopoulos SA, Varisli L, Zoumpourlis P, Spandidos DA, Zoumpourlis V. Investigating the biology of microRNA links to ALDH1A1 reveals candidates for preclinical testing in acute myeloid leukemia. Int J Oncol 2024; 65:115. [PMID: 39513593 PMCID: PMC11575927 DOI: 10.3892/ijo.2024.5703] [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: 07/29/2024] [Accepted: 10/23/2024] [Indexed: 11/15/2024] Open
Abstract
Aldehyde dehydrogenase 1 family member A1 (ALDH1A1) is a member of the aldehyde dehydrogenase gene subfamily that encode enzymes with the ability to oxidize retinaldehyde. It was recently shown that high ALDH1A1 RNA abundance correlates with a poor prognosis in acute myeloid leukemia (AML). AML is a hematopoietic malignancy associated with high morbidity and mortality rates. Although there are a number of agents that inhibit ALDH activity, it would be crucial to develop methodologies for adjustable genetic interference, which would permit interventions on several oncogenic pathways in parallel. Intervention in multiple oncogenic pathways is theoretically possible with microRNAs (miRNAs or miRs), a class of small non‑coding RNAs that have emerged as key regulators of gene expression in AML. A number of miRNAs have shown the ability to interfere with ALDH1A1 gene expression directly in solid tumor cells, and these miRNAs can be evaluated in AML model systems. There are indications that a few of these miRNAs actually do have an association with AML disease course, rendering them a promising target for genetic intervention in AML cells.
Collapse
Affiliation(s)
- Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| |
Collapse
|
3
|
Vlahopoulos SA. Divergent Processing of Cell Stress Signals as the Basis of Cancer Progression: Licensing NFκB on Chromatin. Int J Mol Sci 2024; 25:8621. [PMID: 39201306 PMCID: PMC11354898 DOI: 10.3390/ijms25168621] [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: 07/10/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Inflammation is activated by diverse triggers that induce the expression of cytokines and adhesion molecules, which permit a succession of molecules and cells to deliver stimuli and functions that help the immune system clear the primary cause of tissue damage, whether this is an infection, a tumor, or a trauma. During inflammation, short-term changes in the expression and secretion of strong mediators of inflammation occur, while long-term changes occur to specific groups of cells. Long-term changes include cellular transdifferentiation for some types of cells that need to regenerate damaged tissue, as well as death for specific immune cells that can be detrimental to tissue integrity if they remain active beyond the boundaries of essential function. The transcriptional regulator NFκB enables some of the fundamental gene expression changes during inflammation, as well as during tissue development. During recurrence of malignant disease, cell stress-induced alterations enable the growth of cancer cell clones that are substantially resistant to therapeutic intervention and to the immune system. A number of those alterations occur due to significant defects in feedback signal cascades that control the activity of NFκB. Specifically, cell stress contributes to feedback defects as it overrides modules that otherwise control inflammation to protect host tissue. NFκB is involved in both the suppression and promotion of cancer, and the key distinctive feature that determines its net effect remains unclear. This paper aims to provide a clear answer to at least one aspect of this question, namely the mechanism that enables a divergent response of cancer cells to critical inflammatory stimuli and to cell stress in general.
Collapse
|
4
|
Kontogiannis S, Markantes G, Stamou M, Tsagkarakis M, Mamali I, Giannitsas K, Perimenis P, Georgopoulos N, Athanasopoulos A. Anti-Müllerian hormone: a novel biomarker for aggressive prostate cancer? Emerging evidence from a prospective study of radical prostatectomies. Hormones (Athens) 2024; 23:297-304. [PMID: 38127275 PMCID: PMC11190032 DOI: 10.1007/s42000-023-00520-z] [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: 06/04/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE Prostate cancer patients are a heterogeneous group as regards the aggressiveness of the disease. The relationship of steroid hormones with the aggressiveness of prostate cancer is unclear. It is known that the anti-Müllerian hormone (AMH) inhibits prostate cancer cell lines in vitro. The aim of this study is to investigate the relationship of AMH and steroid hormones with the aggressiveness of prostate cancer. METHODS This was a prospective study of consecutive radical prostatectomy patients. We measured the following hormones: total testosterone, sex hormone-binding globulin, albumin, luteinizing hormone, follicle-stimulating hormone, estradiol, dehydroepiandrosterone sulfate, androstenedione, and AMH. The minimum follow-up after radical prostatectomy was 5 years. For the aggressiveness of prostate cancer, we considered the following three variables: post-operative Gleason score (GS) ≥ 8, TNM pΤ3 disease, and prostate-specific antigen (PSA) biochemical recurrence (BCR). RESULTS In total, 91 patients were enrolled. The mean age and PSA were 64.8 years and 9.3 ng/dl, respectively. The median post-operative GS was 7. Low AMH blood levels were correlated with higher post-operative GS (p = 0.001), as well as with PSA BCR (p = 0.043). With pT3 disease, only albumin was (negatively) correlated (p = 0.008). ROC analysis showed that AMH is a good predictor of BCR (AUC 0.646, 95% CI 0.510-0.782, p = 0.043); a cutoff value of 3.06 ng/dl had a positive prognostic value of 71.4% and a negative prognostic value of 63.3% for BCR. Cox regression analysis showed that AMH is a statistically significant and independent prognostic marker for BCR (p = 0.013). More precisely, for every 1 ng/ml of AMH rise, the probability for PSA BCR decreases by 20.8% (HR = 0.792). Moreover, in Kaplan-Meier analysis, disease-free survival is more probable in patients with AMΗ ≥ 3.06 ng/ml (p = 0.004). CONCLUSIONS Low AMH blood levels were correlated with aggressive prostate cancer in this radical prostatectomy cohort of patients. Therefore, AMH could be a prognostic biomarker for the aggressiveness of the disease.
Collapse
Affiliation(s)
| | | | - Maria Stamou
- Harvard Reproductive Sciences Center, Massachusetts General Hospital, Boston, MA, USA
| | | | - Irini Mamali
- Endocrinology Department, Patras University Hospital, Patras, Greece
| | | | | | | | | |
Collapse
|
5
|
Vlahopoulos S, Pan L, Varisli L, Dancik GM, Karantanos T, Boldogh I. OGG1 as an Epigenetic Reader Affects NFκB: What This Means for Cancer. Cancers (Basel) 2023; 16:148. [PMID: 38201575 PMCID: PMC10778025 DOI: 10.3390/cancers16010148] [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: 11/22/2023] [Revised: 12/16/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
8-oxoguanine glycosylase 1 (OGG1), which was initially identified as the enzyme that catalyzes the first step in the DNA base excision repair pathway, is now also recognized as a modulator of gene expression. What is important for cancer is that OGG1 acts as a modulator of NFκB-driven gene expression. Specifically, oxidant stress in the cell transiently halts enzymatic activity of substrate-bound OGG1. The stalled OGG1 facilitates DNA binding of transactivators, such as NFκB to their cognate sites, enabling the expression of cytokines and chemokines, with ensuing recruitment of inflammatory cells. Recently, we highlighted chief aspects of OGG1 involvement in regulation of gene expression, which hold significance in lung cancer development. However, OGG1 has also been implicated in the molecular underpinning of acute myeloid leukemia. This review analyzes and discusses how these cells adapt through redox-modulated intricate connections, via interaction of OGG1 with NFκB, which provides malignant cells with alternative molecular pathways to transform their microenvironment, enabling adjustment, promoting cell proliferation, metastasis, and evading killing by therapeutic agents.
Collapse
Affiliation(s)
- Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| | - Lang Pan
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA;
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA;
| | - Theodoros Karantanos
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA;
| | - Istvan Boldogh
- Department of Microbiology and Immunology, School of Medicine, University of Texas Medical Branch at Galveston, 301 University Blvd., Galveston, TX 77555, USA;
| |
Collapse
|
6
|
Yu X, Mukwaya V, Mann S, Dou H. Signal Transduction in Artificial Cells. SMALL METHODS 2023; 7:e2300231. [PMID: 37116092 DOI: 10.1002/smtd.202300231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/06/2023] [Indexed: 06/19/2023]
Abstract
In recent years, significant progress has been made in the emerging field of constructing biomimetic soft compartments with life-like behaviors. Given that biological activities occur under a flux of energy and matter exchange, the implementation of rudimentary signaling pathways in artificial cells (protocells) is a prerequisite for the development of adaptive sense-response phenotypes in cytomimetic models. Herein, recent approaches to the integration of signal transduction modules in model protocells prepared by bottom-up construction are discussed. The approaches are classified into two categories involving invasive biochemical signals or non-invasive physical stimuli. In the former mechanism, transducers with intrinsic recognition capability respond with high specificity, while in the latter, artificial cells respond through intra-protocellular energy transduction. Although major challenges remain in the pursuit of a sophisticated artificial signaling network for the orchestration of higher-order cytomimetic models, significant advances have been made in establishing rudimentary protocell communication networks, providing novel organizational models for the development of life-like microsystems and new avenues in protoliving technologies.
Collapse
Affiliation(s)
- Xiaolei Yu
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Zhangjiang Institute for Advanced Study (ZIAS), Shanghai Jiao Tong University, Shanghai, 201203, China
| | - Vincent Mukwaya
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Zhangjiang Institute for Advanced Study (ZIAS), Shanghai Jiao Tong University, Shanghai, 201203, China
| | - Stephen Mann
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Zhangjiang Institute for Advanced Study (ZIAS), Shanghai Jiao Tong University, Shanghai, 201203, China
- Max Planck Bristol Centre for Minimal Biology and Centre for Protolife Research, School of Chemistry, University of Bristol, Bristol, BS8 1TS, UK
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
- Zhangjiang Institute for Advanced Study (ZIAS), Shanghai Jiao Tong University, Shanghai, 201203, China
| |
Collapse
|
7
|
Dancik GM, Varisli L, Tolan V, Vlahopoulos S. Aldehyde Dehydrogenase Genes as Prospective Actionable Targets in Acute Myeloid Leukemia. Genes (Basel) 2023; 14:1807. [PMID: 37761947 PMCID: PMC10531322 DOI: 10.3390/genes14091807] [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: 07/13/2023] [Revised: 08/29/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
It has been previously shown that the aldehyde dehydrogenase (ALDH) family member ALDH1A1 has a significant association with acute myeloid leukemia (AML) patient risk group classification and that AML cells lacking ALDH1A1 expression can be readily killed via chemotherapy. In the past, however, a redundancy between the activities of subgroup members of the ALDH family has hampered the search for conclusive evidence to address the role of specific ALDH genes. Here, we describe the bioinformatics evaluation of all nineteen member genes of the ALDH family as prospective actionable targets for the development of methods aimed to improve AML treatment. We implicate ALDH1A1 in the development of recurrent AML, and we show that from the nineteen members of the ALDH family, ALDH1A1 and ALDH2 have the strongest association with AML patient risk group classification. Furthermore, we discover that the sum of the expression values for RNA from the genes, ALDH1A1 and ALDH2, has a stronger association with AML patient risk group classification and survival than either one gene alone does. In conclusion, we identify ALDH1A1 and ALDH2 as prospective actionable targets for the treatment of AML in high-risk patients. Substances that inhibit both enzymatic activities constitute potentially effective pharmaceutics.
Collapse
Affiliation(s)
- Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey; (L.V.); (V.T.)
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey; (L.V.); (V.T.)
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|
8
|
Dancik GM, Varisli L, Vlahopoulos SA. The Molecular Context of Oxidant Stress Response in Cancer Establishes ALDH1A1 as a Critical Target: What This Means for Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:ijms24119372. [PMID: 37298333 DOI: 10.3390/ijms24119372] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/18/2023] [Accepted: 05/25/2023] [Indexed: 06/12/2023] Open
Abstract
The protein family of aldehyde dehydrogenases (ALDH) encompasses nineteen members. The ALDH1 subfamily consists of enzymes with similar activity, having the capacity to neutralize lipid peroxidation products and to generate retinoic acid; however, only ALDH1A1 emerges as a significant risk factor in acute myeloid leukemia. Not only is the gene ALDH1A1 on average significantly overexpressed in the poor prognosis group at the RNA level, but its protein product, ALDH1A1 protects acute myeloid leukemia cells from lipid peroxidation byproducts. This capacity to protect cells can be ascribed to the stability of the enzyme under conditions of oxidant stress. The capacity to protect cells is evident both in vitro, as well as in mouse xenografts of those cells, shielding cells effectively from a number of potent antineoplastic agents. However, the role of ALDH1A1 in acute myeloid leukemia has been unclear in the past due to evidence that normal cells often have higher aldehyde dehydrogenase activity than leukemic cells. This being true, ALDH1A1 RNA expression is significantly associated with poor prognosis. It is hence imperative that ALDH1A1 is methodically targeted, particularly for the acute myeloid leukemia patients of the poor prognosis risk group that overexpress ALDH1A1 RNA.
Collapse
Affiliation(s)
- Garrett M Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA
| | - Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey
| | - Spiros A Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| |
Collapse
|
9
|
Vezakis IA, Lambrou GI, Matsopoulos GK. Deep Learning Approaches to Osteosarcoma Diagnosis and Classification: A Comparative Methodological Approach. Cancers (Basel) 2023; 15:cancers15082290. [PMID: 37190217 DOI: 10.3390/cancers15082290] [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: 02/01/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
BACKGROUND Osteosarcoma is the most common primary malignancy of the bone, being most prevalent in childhood and adolescence. Despite recent progress in diagnostic methods, histopathology remains the gold standard for disease staging and therapy decisions. Machine learning and deep learning methods have shown potential for evaluating and classifying histopathological cross-sections. METHODS This study used publicly available images of osteosarcoma cross-sections to analyze and compare the performance of state-of-the-art deep neural networks for histopathological evaluation of osteosarcomas. RESULTS The classification performance did not necessarily improve when using larger networks on our dataset. In fact, the smallest network combined with the smallest image input size achieved the best overall performance. When trained using 5-fold cross-validation, the MobileNetV2 network achieved 91% overall accuracy. CONCLUSIONS The present study highlights the importance of careful selection of network and input image size. Our results indicate that a larger number of parameters is not always better, and the best results can be achieved on smaller and more efficient networks. The identification of an optimal network and training configuration could greatly improve the accuracy of osteosarcoma diagnoses and ultimately lead to better disease outcomes for patients.
Collapse
Affiliation(s)
- Ioannis A Vezakis
- Biomedical Engineering Laboratory, School of Electrical & Computer Engineering, National Technical University of Athens, 9 Iroon Polytechniou St., 15780 Athens, Greece
| | - George I Lambrou
- Biomedical Engineering Laboratory, School of Electrical & Computer Engineering, National Technical University of Athens, 9 Iroon Polytechniou St., 15780 Athens, Greece
- Choremeio Research Laboratory, First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527 Athens, Greece
| | - George K Matsopoulos
- Biomedical Engineering Laboratory, School of Electrical & Computer Engineering, National Technical University of Athens, 9 Iroon Polytechniou St., 15780 Athens, Greece
| |
Collapse
|
10
|
Barua A, Hatzikirou H. Cell Decision Making through the Lens of Bayesian Learning. ENTROPY (BASEL, SWITZERLAND) 2023; 25:e25040609. [PMID: 37190396 PMCID: PMC10137733 DOI: 10.3390/e25040609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/20/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
Cell decision making refers to the process by which cells gather information from their local microenvironment and regulate their internal states to create appropriate responses. Microenvironmental cell sensing plays a key role in this process. Our hypothesis is that cell decision-making regulation is dictated by Bayesian learning. In this article, we explore the implications of this hypothesis for internal state temporal evolution. By using a timescale separation between internal and external variables on the mesoscopic scale, we derive a hierarchical Fokker-Planck equation for cell-microenvironment dynamics. By combining this with the Bayesian learning hypothesis, we find that changes in microenvironmental entropy dominate the cell state probability distribution. Finally, we use these ideas to understand how cell sensing impacts cell decision making. Notably, our formalism allows us to understand cell state dynamics even without exact biochemical information about cell sensing processes by considering a few key parameters.
Collapse
Affiliation(s)
- Arnab Barua
- Departement de Biochimie, Université de Montréal, Montréal, QC H3T 1C5, Canada
- Centre Robert-Cedergren en Bio-Informatique et Génomique, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Haralampos Hatzikirou
- Center for Information Services and High Performance Computing, Technische Univesität Dresden, 01062 Dresden, Germany
- Mathematics Department, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
11
|
Son SY, Choi JH, Kim EB, Yin J, Seonu SY, Jin SY, Oh JY, Lee MW. Chemopreventive Activity of Ellagitannins from Acer pseudosieboldianum (Pax) Komarov Leaves on Prostate Cancer Cells. PLANTS (BASEL, SWITZERLAND) 2023; 12:1047. [PMID: 36903908 PMCID: PMC10005130 DOI: 10.3390/plants12051047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Several studies have shown that compounds from Acer pseudosieboldianum (Pax) Komarov leaves (APL) display potent anti-oxidative, anti-inflammatory, and anti-proliferative activities. Prostate cancer (PCa) is the most common cancer among older men, and DNA methylation is associated with PCa progression. This study aimed to investigate the chemopreventive activities of the compounds which were isolated from APL on prostate cancer cells and elucidate the mechanisms of these compounds in relation to DNA methylation. One novel ellagitannin [komaniin (14)] and thirteen other known compounds, including glucose derivatives [ethyl-β-D-glucopyranose (3) and (4R)-p-menth-1-ene-7,8-diol 7-O-β-D-glucopyranoside (4)], one phenylpropanoid [junipetrioloside A (5)], three phenolic acid derivatives [ellagic acid-4-β-D-xylopyranoside (1), 4-O-galloyl-quinic acid (2), and gallic acid (8)], two flavonoids [quercetin (11) and kaempferol (12)], and five hydrolysable tannins [geraniin (6), punicafolin (7), granatin B (9), 1,2,3,4,6-penta-galloyl-β-D-glucopyranoside (10), and mallotusinic acid (13)] were isolated from APL. The hydrolyzable tannins (6, 7, 9, 10, 13, and 14) showed potent anti-PCa proliferative and apoptosis-promoting activities. Among the compounds, the ellagitannins in the dehydrohexahydroxydiphenoyl (DHHDP) group (6, 9, 13, and 14), the novel compound 14 showed the most potent inhibitory activity on DNA methyltransferase (DNMT1, 3a and 3b) and glutathione S-transferase P1 methyl removing and re-expression activities. Thus, our results suggested that the ellagitannins (6, 9, 13, and 14) isolated from APL could be a promising treatment option for PCa.
Collapse
|
12
|
OPN Promotes Cell Proliferation and Invasion through NF- κB in Human Esophageal Squamous Cell Carcinoma. Genet Res (Camb) 2022; 2022:3154827. [PMID: 36619897 PMCID: PMC9779994 DOI: 10.1155/2022/3154827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
Background Osteopontin (OPN) is a phosphorylated glycoprotein. There is increasing evidence that the OPN gene played a major role in the progression of solid organ tumors. However, few studies have clarified how OPN regulated the functional role of human esophageal squamous cell carcinoma (ESCC). This study was designed to investigate the effect of OPN in esophageal squamous cell carcinoma. Methods First, we screened Eca-109 and KYSE-510 cells to construct OPN silencing and overexpression models. Endogenous OPN of Eca-109 and KYSE-510 were knocked down or overexpressed using small interfering RNAs. QRT-PCR, Western blot, flow cytometry, and CCK-8 were used to detect the function of Eca-109 and KYSE-510 cells. Tumor formation in nude mice was used to measure tumor growth after OPN inhibition. Results Eca-109 and KYSE-510 cells contain the si-OPN arrest cell cycle in the S-phase and increase apoptosis. These changes were OPN downregulation of the NF-κB pathway that significantly reduced the protein levels of TNF-α, IL-1β, and p-p65. However, the activity of Eca-109 and KYSE-510 cells was enhanced in OPN overexpressing cells. Then, the in vivo tumor formation experiment in nude mice showed that the tumor volume and weight of nude mice after silencing OPN were significantly reduced. Conclusion This study contributed to understanding the vital role of OPN in ESCC development and progression. This could be a promising molecular target for developing new ESCC diagnostic and therapeutic strategies.
Collapse
|
13
|
Xun K, Sun Y, Zhang Q, Wen N, Wang Z, Qiu L, Tan W. Aptamer-Based Analysis and Manipulation of the Protein Activity in Living Cells. Anal Chem 2022; 94:4352-4358. [PMID: 35230816 DOI: 10.1021/acs.analchem.1c05104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Directly analyzing and precisely manipulating the activity of target proteins without altering their natural structure and expression would be essential to decoding many protein-dominant cellular processes. To meet this goal, we used streptavidin as the carrier to develop an aptamer-based nanoplatform for monitoring the activation process of specific proteins in living cells. Our results showed that this nanoplatform could efficiently enter the cellular cytoplasm and specifically report the presence of RelA in the activated state. Meanwhile, with incorporation of a photoresponsive module, this aptamer-based nanoplatform was able to manipulate the nuclear translocation behavior of active RelA, enabling control over related downstream signaling events.
Collapse
Affiliation(s)
- Kanyu Xun
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China
| | - Yue Sun
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China
| | - Qiang Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China
| | - Nachuan Wen
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China
| | - Zhimin Wang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China
| | - Liping Qiu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China.,NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha 410000, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, Hunan, China
| |
Collapse
|
14
|
Endothelial Dysfunction through Oxidatively Generated Epigenetic Mark in Respiratory Viral Infections. Cells 2021; 10:cells10113067. [PMID: 34831290 PMCID: PMC8623825 DOI: 10.3390/cells10113067] [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: 09/13/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
The bronchial vascular endothelial network plays important roles in pulmonary pathology during respiratory viral infections, including respiratory syncytial virus (RSV), influenza A(H1N1) and importantly SARS-Cov-2. All of these infections can be severe and even lethal in patients with underlying risk factors.A major obstacle in disease prevention is the lack of appropriate efficacious vaccine(s) due to continuous changes in the encoding capacity of the viral genome, exuberant responsiveness of the host immune system and lack of effective antiviral drugs. Current management of these severe respiratory viral infections is limited to supportive clinical care. The primary cause of morbidity and mortality is respiratory failure, partially due to endothelial pulmonary complications, including edema. The latter is induced by the loss of alveolar epithelium integrity and by pathological changes in the endothelial vascular network that regulates blood flow, blood fluidity, exchange of fluids, electrolytes, various macromolecules and responses to signals triggered by oxygenation, and controls trafficking of leukocyte immune cells. This overview outlines the latest understanding of the implications of pulmonary vascular endothelium involvement in respiratory distress syndrome secondary to viral infections. In addition, the roles of infection-induced cytokines, growth factors, and epigenetic reprogramming in endothelial permeability, as well as emerging treatment options to decrease disease burden, are discussed.
Collapse
|
15
|
Poursoltani F, Nejati V, Pazhang Y, Rezaie J. Sulindac and vitamin D3 synergically inhibit proliferation of MCF-7 breast cancer cell through AMPK/Akt/β-catenin axis in vitro. Cell Biochem Funct 2021; 39:991-997. [PMID: 34472641 DOI: 10.1002/cbf.3668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 08/15/2021] [Accepted: 08/21/2021] [Indexed: 12/29/2022]
Abstract
Breast cancer is associated with a high rate of recurrence, resistance therapy and mortality worldwide. We aimed at investigating the inhibitory effects of Sulindac and vitamin D3 (VD) on MCF-7 human breast cancer cells. MCF-7 cells were cultured with different concentrations of Sulindac and VD over a period of 24, 48 and 72 hours for cell viability and IC50 experiments. Hochst staining was used to evaluate apoptosis, whereas quantitative PCR (qPCR) was performed to measure mRNA levels of BCL-2 and BAX genes. Immunofluorescence staining was used to monitor intracellular β-catenin expression. The protein levels of AKT, AMPK and P65 were measured by western blotting. The result showed that cell viability decreased in treated cells dose/time dependently (P < .05). Hochst staining showed an increase in fragmented nuclei in treated cells. The expression of BCL-2 and BAX genes decreased and increased in treated cells, respectively (P < .05). Immunofluorescence staining indicated that the expression of β-catenin significantly reduced in treated cells. The AKT-1/p-Akt-1 and AMPK/p-AMPK ratio increased in treated cells (P < .05), but the P65/p-P65 ratio did not change significantly (P > .05). Our results indicated that the combination of Sulindac and VD has a growth-inhibiting effect on MCF-7 cells through AMPK/Akt/β-catenin axis.
Collapse
Affiliation(s)
| | - Vahid Nejati
- Biology Department, Faculty of Sciences, Urmia University, Urmia, Iran
| | - Yaghub Pazhang
- Biology Department, Faculty of Sciences, Urmia University, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
16
|
Papatsirou M, Artemaki PI, Karousi P, Scorilas A, Kontos CK. Circular RNAs: Emerging Regulators of the Major Signaling Pathways Involved in Cancer Progression. Cancers (Basel) 2021; 13:cancers13112744. [PMID: 34205978 PMCID: PMC8198587 DOI: 10.3390/cancers13112744] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/28/2021] [Accepted: 05/29/2021] [Indexed: 12/11/2022] Open
Abstract
Signal transduction is an essential process that regulates and coordinates fundamental cellular processes, such as development, immunity, energy metabolism, and apoptosis. Through signaling, cells are capable of perceiving their environment and adjusting to changes, and most signaling cascades ultimately lead to alterations in gene expression. Circular RNAs (circRNAs) constitute an emerging type of endogenous transcripts with regulatory roles and unique properties. They are stable and expressed in a tissue-, cell-, and developmental stage-specific manner, while they are involved in the pathogenesis of several diseases, including cancer. Aberrantly expressed circRNAs can mediate cancer progression through regulation of the activity of major signaling cascades, such as the VEGF, WNT/β-catenin, MAPK, PI3K/AKT, and Notch signaling pathways, as well as by interfering with signaling crosstalk. Deregulated signaling can then function to induce angiogenesis, promote invasion, migration, and metastasis, and, generally, modulate the hallmarks of cancer. In this review article, we summarize the most recently described and intriguing cases of circRNA-mediated signaling regulation that are involved in cancer progression, and discuss the biomarker potential of circRNAs, as well as future therapeutic applications.
Collapse
|
17
|
Queralt-Rosinach N, Stupp GS, Li TS, Mayers M, Hoatlin ME, Might M, Good BM, Su AI. Structured reviews for data and knowledge-driven research. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2021; 2020:5818923. [PMID: 32283553 PMCID: PMC7153956 DOI: 10.1093/database/baaa015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 01/21/2020] [Accepted: 02/07/2020] [Indexed: 12/25/2022]
Abstract
Hypothesis generation is a critical step in research and a cornerstone in the rare disease field. Research is most efficient when those hypotheses are based on the entirety of knowledge known to date. Systematic review articles are commonly used in biomedicine to summarize existing knowledge and contextualize experimental data. But the information contained within review articles is typically only expressed as free-text, which is difficult to use computationally. Researchers struggle to navigate, collect and remix prior knowledge as it is scattered in several silos without seamless integration and access. This lack of a structured information framework hinders research by both experimental and computational scientists. To better organize knowledge and data, we built a structured review article that is specifically focused on NGLY1 Deficiency, an ultra-rare genetic disease first reported in 2012. We represented this structured review as a knowledge graph and then stored this knowledge graph in a Neo4j database to simplify dissemination, querying and visualization of the network. Relative to free-text, this structured review better promotes the principles of findability, accessibility, interoperability and reusability (FAIR). In collaboration with domain experts in NGLY1 Deficiency, we demonstrate how this resource can improve the efficiency and comprehensiveness of hypothesis generation. We also developed a read–write interface that allows domain experts to contribute FAIR structured knowledge to this community resource. In contrast to traditional free-text review articles, this structured review exists as a living knowledge graph that is curated by humans and accessible to computational analyses. Finally, we have generalized this workflow into modular and repurposable components that can be applied to other domain areas. This NGLY1 Deficiency-focused network is publicly available at http://ngly1graph.org/. Availability and implementation Database URL: http://ngly1graph.org/. Network data files are at: https://github.com/SuLab/ngly1-graph and source code at: https://github.com/SuLab/bioknowledge-reviewer. Contact asu@scripps.edu
Collapse
Affiliation(s)
- Núria Queralt-Rosinach
- Department of Integrative Structural and Computational Biology, Scripps Research, 10550 N Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Gregory S Stupp
- Department of Integrative Structural and Computational Biology, Scripps Research, 10550 N Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Tong Shu Li
- Department of Integrative Structural and Computational Biology, Scripps Research, 10550 N Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Michael Mayers
- Department of Integrative Structural and Computational Biology, Scripps Research, 10550 N Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Maureen E Hoatlin
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, 3181 SW Sam Jackson Parkway, Portland, OR 97239, USA
| | - Matthew Might
- Department of Medicine, Hugh Kaul Precision Medicine Institute, University of Alabama at Birmingham, 510 20th St S, Birmingham, AL 35210, USA
| | - Benjamin M Good
- Department of Integrative Structural and Computational Biology, Scripps Research, 10550 N Torrey Pines Rd. La Jolla, CA 92037, USA
| | - Andrew I Su
- Department of Integrative Structural and Computational Biology, Scripps Research, 10550 N Torrey Pines Rd. La Jolla, CA 92037, USA
| |
Collapse
|
18
|
Yanagi T, Kaminaga K, Kada W, Hanaizumi O, Igarashi R. Optimization of Wide-Field ODMR Measurements Using Fluorescent Nanodiamonds to Improve Temperature Determination Accuracy. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2282. [PMID: 33217922 PMCID: PMC7698612 DOI: 10.3390/nano10112282] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Fluorescent nanodiamonds containing nitrogen-vacancy centers have attracted attention as nanoprobes for temperature measurements in microenvironments, potentially enabling the measurement of intracellular temperature distributions and temporal changes. However, to date, the time resolution and accuracy of the temperature determinations using fluorescent nanodiamonds have been insufficient for wide-field fluorescence imaging. Here, we describe a method for highly accurate wide-field temperature imaging using fluorescent nanodiamonds for optically detected magnetic resonance (ODMR) measurements. We performed a Monte Carlo simulation to determine the optimal frequency sweep range for ODMR temperature determination. We then applied this sweep range to fluorescent nanodiamonds. As a result, the temperature determination accuracies were improved by a factor ~1.5. Our result paves the way for the contribution of quantum sensors to cell biology for understanding, for example, differentiation in multicellular systems.
Collapse
Affiliation(s)
- Tamami Yanagi
- Division of Electronics and Informatics, Graduate School of Science and Technology, Gunma University, Kiryu, Gunma 376-8515, Japan; (T.Y.); (W.K.)
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Inage-ku, Chiba 263-8555, Japan;
| | - Kiichi Kaminaga
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Inage-ku, Chiba 263-8555, Japan;
- National Institute for Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
| | - Wataru Kada
- Division of Electronics and Informatics, Graduate School of Science and Technology, Gunma University, Kiryu, Gunma 376-8515, Japan; (T.Y.); (W.K.)
| | - Osamu Hanaizumi
- Division of Electronics and Informatics, Graduate School of Science and Technology, Gunma University, Kiryu, Gunma 376-8515, Japan; (T.Y.); (W.K.)
| | - Ryuji Igarashi
- Institute for Quantum Life Science, National Institutes for Quantum and Radiological Science and Technology, Inage-ku, Chiba 263-8555, Japan;
- National Institute for Radiological Sciences, National Institute for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage-ku, Chiba 263-8555, Japan
- JST, PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| |
Collapse
|
19
|
The interplay of the oral microbiome and alcohol consumption in oral squamous cell carcinomas. Oral Oncol 2020; 110:105011. [PMID: 32980528 DOI: 10.1016/j.oraloncology.2020.105011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/11/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022]
Abstract
Oral cancer (OC) is among the top twenty occurring cancers in the world, with a mortality rate of 50%. A shift to a functionally inflammatory or a 'disease state' oral microbiome composition has been observed amongst patients with premalignant disorders and OC, with evidence suggesting alcohol could be exacerbating the inflammatory influence of the oral microorganisms. Alcohol dehydrogenase (ADH, EC 1.1.1.1) converts alcohol into a known carcinogenic metabolite, acetaldehyde and while ADH levels in oral mucosa are low, several oral commensal species possess ADH and could produce genotoxic levels of acetaldehyde. With a direct association between oral microbiome status, alcohol and poor oral health status combining to induce chronic inflammation with increased acetaldehyde levels - this leads to a tumour promoting environment. This new disease state increases the production of reactive oxygen species (ROS), while impairing anti-oxidant systems thus activating the redox signalling required for the promotion and survival of tumours. This review aims to highlight the evidence linking these processes in the progression of oral cancer.
Collapse
|
20
|
Functional variations of NFKB1 and NFKB1A in inflammatory disorders and their implication for therapeutic approaches. ASIAN BIOMED 2020. [DOI: 10.1515/abm-2020-0008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Abstract
Nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) is a sophisticated transcription factor that is particularly important in the inflammatory response, but it regulates more than 400 individual and dependent genes for parts of the apoptotic, angiogenic, and proliferative, differentiative, and cell adhesion pathways. NF-κB function is directly inhibited by the binding of inhibitor of κB (IκB), and the imbalance between NF-κB and IκB has been linked to the development and progression of cancer and a variety of inflammatory disorders. These observations might broaden the horizon of current knowledge, particularly on the pathogenesis of inflammatory diseases considering the roles of NF-κB and IκB. In this context, we focus this narrative review on a comparative discussion of our findings with other literature regarding variations of NFKB1 and NFKB1A and their association with susceptibility to widespread inflammatory disorders (such as atherosclerosis, morbid obesity, Behçet syndrome, Graves disease, Hashimoto disease) and common cancers (such as gliomas).
Collapse
|
21
|
Liubomirski Y, Ben-Baruch A. Notch-Inflammation Networks in Regulation of Breast Cancer Progression. Cells 2020; 9:cells9071576. [PMID: 32605277 PMCID: PMC7407628 DOI: 10.3390/cells9071576] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 12/20/2022] Open
Abstract
Members of the Notch family and chronic inflammation were each separately demonstrated to have prominent malignancy-supporting roles in breast cancer. Recent investigations indicate that bi-directional interactions that exist between these two pathways promote the malignancy phenotype of breast tumor cells and of their tumor microenvironment. In this review article, we demonstrate the importance of Notch-inflammation interplays in malignancy by describing three key networks that act in breast cancer and their impacts on functions that contribute to disease progression: (1) Cross-talks of the Notch pathway with myeloid cells that are important players in cancer-related inflammation, focusing mainly on macrophages; (2) Cross-talks of the Notch pathway with pro-inflammatory factors, exemplified mainly by Notch interactions with interleukin 6 and its downstream pathways (STAT3); (3) Cross-talks of the Notch pathway with typical inflammatory transcription factors, primarily NF-κB. These three networks enhance tumor-promoting functions in different breast tumor subtypes and act in reciprocal manners, whereby Notch family members activate inflammatory elements and vice versa. These characteristics illustrate the fundamental roles played by Notch-inflammation interactions in elevating breast cancer progression and propose that joint targeting of both pathways together may provide more effective and less toxic treatment approaches in this disease.
Collapse
|
22
|
Stagni V, Kaminari A, Sideratou Z, Sakellis E, Vlahopoulos SA, Tsiourvas D. Targeting breast cancer stem-like cells using chloroquine encapsulated by a triphenylphosphonium-functionalized hyperbranched polymer. Int J Pharm 2020; 585:119465. [PMID: 32497731 DOI: 10.1016/j.ijpharm.2020.119465] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 01/07/2023]
Abstract
Cancer stem cells (CSCs) have garnered increasing attention over the past decade, as they are believed to play a crucial role in tumor progression and drug resistance. Accumulating evidence provides insight into the function of autophagy in maintenance and survival of CSCs. Here, we studied the impact of a mitochondriotropic triphenylphosphonium-functionalized dendrimeric nanocarrier on cultured breast cancer cell lines, grown either as adherent cells or as mammospheres that mimic a stem-like phenotype. The nanocarrier manifested a substantial cytotoxicity both alone as well as after encapsulation of chloroquine, a well-known autophagy inhibitor. The cytotoxic effects of the nanocarrier could be ascribed to interference with mitochondrial function. Importantly, mammospheres were selectively sensitive to encapsulated chloroquine and this depends on the expression of the gene encoding ATM kinase. Ataxia-telangiectasia mutated (ATM) kinase is an enzyme that functions as an essential signaling mediator that enables growth of cancer stem cells through the regulation of autophagy. We noted that this ATM-dependent sensitivity of mammospheres to encapsulated chloroquine was independent of the status of the tumor suppressor gene p53. Our study suggests that breast cancer stem cells, as they are modeled by mammospheres, are sensitive to encapsulated chloroquine, depending on the expression of the ATM kinase, which is thereby characterized as a potential biomarker for sensitivity to this type of treatment.
Collapse
Affiliation(s)
- Venturina Stagni
- Laboratory of Cell Signaling, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy; Institute of Molecular Biology and Pathology, National Research Council (CNR), Via DegliApuli 4, 00185 Rome, Italy.
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Zili Sideratou
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Elias Sakellis
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Spiros A Vlahopoulos
- Ηoremeio Research Laboratory, First Department of Paediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitris Tsiourvas
- Institute of Nanoscience and Nanotechnology, NCSR "Demokritos", 15310 Aghia Paraskevi, Greece.
| |
Collapse
|
23
|
Lambrou GI, Hatziagapiou K, Vlahopoulos S. Inflammation and tissue homeostasis: the NF-κB system in physiology and malignant progression. Mol Biol Rep 2020; 47:4047-4063. [PMID: 32239468 DOI: 10.1007/s11033-020-05410-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Accepted: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Disruption of tissue function activates cellular stress which triggers a number of mechanisms that protect the tissue from further damage. These mechanisms involve a number of homeostatic modules, which are regulated at the level of gene expression by the transactivator NF-κB. This transcription factor shifts between activation and repression of discrete, cell-dependent gene expression clusters. Some of its target genes provide feedback to NF-κB itself, thereby strengthening the inflammatory response of the tissue and later terminating inflammation to facilitate restoration of tissue homeostasis. Disruption of key feedback modules for NF-κB in certain cell types facilitates the survival of clones with genomic aberrations, and protects them from being recognized and eliminated by the immune system, to enable thereby carcinogenesis.
Collapse
Affiliation(s)
- George I Lambrou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece
| | - Kyriaki Hatziagapiou
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, 11527, Goudi-Athens, Greece.
| |
Collapse
|
24
|
Varisli L, Cen O, Vlahopoulos S. Dissecting pharmacological effects of chloroquine in cancer treatment: interference with inflammatory signaling pathways. Immunology 2020; 159:257-278. [PMID: 31782148 PMCID: PMC7011648 DOI: 10.1111/imm.13160] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/20/2019] [Accepted: 11/21/2019] [Indexed: 12/11/2022] Open
Abstract
Chloroquines are 4-aminoquinoline-based drugs mainly used to treat malaria. At pharmacological concentrations, they have significant effects on tissue homeostasis, targeting diverse signaling pathways in mammalian cells. A key target pathway is autophagy, which regulates macromolecule turnover in the cell. In addition to affecting cellular metabolism and bioenergetic flow equilibrium, autophagy plays a pivotal role at the interface between inflammation and cancer progression. Chloroquines consequently have critical effects in tissue metabolic activity and importantly, in key functions of the immune system. In this article, we will review the work addressing the role of chloroquines in the homeostasis of mammalian tissue, and the potential strengths and weaknesses concerning their use in cancer therapy.
Collapse
Affiliation(s)
- Lokman Varisli
- Union of Education and Science Workers (EGITIM SEN), Diyarbakir Branch, Diyarbakir, Turkey
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir, Turkey
| | - Osman Cen
- Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Natural Sciences, Joliet Jr College, Joliet, IL, USA
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Purslow JA, Khatiwada B, Bayro MJ, Venditti V. NMR Methods for Structural Characterization of Protein-Protein Complexes. Front Mol Biosci 2020; 7:9. [PMID: 32047754 PMCID: PMC6997237 DOI: 10.3389/fmolb.2020.00009] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/10/2020] [Indexed: 01/21/2023] Open
Abstract
Protein-protein interactions and the complexes thus formed are critical elements in a wide variety of cellular events that require an atomic-level description to understand them in detail. Such complexes typically constitute challenging systems to characterize and drive the development of innovative biophysical methods. NMR spectroscopy techniques can be applied to extract atomic resolution information on the binding interfaces, intermolecular affinity, and binding-induced conformational changes in protein-protein complexes formed in solution, in the cell membrane, and in large macromolecular assemblies. Here we discuss experimental techniques for the characterization of protein-protein complexes in both solution NMR and solid-state NMR spectroscopy. The approaches include solvent paramagnetic relaxation enhancement and chemical shift perturbations (CSPs) for the identification of binding interfaces, and the application of intermolecular nuclear Overhauser effect spectroscopy and residual dipolar couplings to obtain structural constraints of protein-protein complexes in solution. Complementary methods in solid-state NMR are described, with emphasis on the versatility provided by heteronuclear dipolar recoupling to extract intermolecular constraints in differentially labeled protein complexes. The methods described are of particular relevance to the analysis of membrane proteins, such as those involved in signal transduction pathways, since they can potentially be characterized by both solution and solid-state NMR techniques, and thus outline key developments in this frontier of structural biology.
Collapse
Affiliation(s)
- Jeffrey A Purslow
- Department of Chemistry, Iowa State University, Ames, IA, United States
| | | | - Marvin J Bayro
- Department of Chemistry and Molecular Sciences Research Center, University of Puerto Rico, San Juan, Puerto Rico
| | - Vincenzo Venditti
- Department of Chemistry, Iowa State University, Ames, IA, United States.,Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, United States
| |
Collapse
|
26
|
Xu H, Zhao F, Hou Q, Huang W, Liu Y, Zhang H, Sun Z. Metagenomic analysis revealed beneficial effects of probiotics in improving the composition and function of the gut microbiota in dogs with diarrhoea. Food Funct 2020; 10:2618-2629. [PMID: 31021333 DOI: 10.1039/c9fo00087a] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The aim of the present study was to evaluate the effects of probiotics on the composition and function of the gut microbiota in dogs with diarrhoea. Forty dogs with diarrhoea were randomly allocated to the treatment group or control group. Probiotics, containing Lactobacillus casei Zhang, Lactobacillus plantarum P-8, and Bifidobacterium animalis subsp. lactis V9, were only fed to 20 treated dogs for 60 days. The faecal samples of all dogs at day 0 and day 60 were analyzed using a metagenomic approach. The results showed a significantly higher microbial diversity and an obvious change in the structure of the gut microbiota in the treatment group. There was also an increase in the abundance of some beneficial bacteria in differently aged dogs, such as Lactobacillus johnsonii (P < 0.05), Lactobacillus reuteri (P < 0.01), Lactobacillus acidophilus (P < 0.05) and Butyricicoccus pullicaecorum (P < 0.05), and a reduction in the abundance of many opportunistic pathogenic bacteria such as Clostridium perfringens (P < 0.05) and Stenotrophomonas maltophilia (P < 0.05) with the supplementation of probiotics. Intriguingly, the correlated networks among some pathogenic bacteria decreased following the administration of probiotics. Additionally, metagenomic analysis revealed the upregulation of pathways involved in the metabolism of amino acids and biosynthesis of secondary metabolites, accompanied by the downregulation of pathways associated with virulence of pathogenic bacteria and cell signaling, suggesting that probiotics could improve the health of dogs with diarrhoea through regulation of the gut microbiota. Our research provides new information relevant to the treatment of diarrhoea in animals and humans.
Collapse
Affiliation(s)
- Haiyan Xu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot 010018, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
27
|
Wang Z, Yang Y, Cui Y, Wang C, Lai Z, Li Y, Zhang W, Mustonen H, Puolakkainen P, Ye Y, Jiang K, Shen Z, Wang S. Tumor-associated macrophages regulate gastric cancer cell invasion and metastasis through TGFβ2/NF-κB/Kindlin-2 axis. Chin J Cancer Res 2020; 32:72-88. [PMID: 32194307 PMCID: PMC7072013 DOI: 10.21147/j.issn.1000-9604.2020.01.09] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Objective Recent studies have shown that tumor-associated macrophages (TAMs) play an important role in cancer invasion and metastasis. Our previous studies have reported that TAMs promote the invasion and metastasis of gastric cancer (GC) cells through the Kindlin-2 pathway. However, the mechanism needs to be clarified. Methods THP-1 monocytes were induced by PMA/interleukin (IL)-4/IL-13 to establish an efficient TAM model in vitro and M2 macrophages were isolated via flow cytometry. A dual luciferase reporter system and chromatin immunoprecipitation (ChIP) assay were used to investigate the mechanism of transforming growth factor β2 (TGFβ2) regulating Kindlin-2 expression. Immunohistochemistry was used to study the relationships among TAM infiltration in human GC tissues, Kindlin-2 protein expression, clinicopathological parameters and prognosis in human GC tissues. A nude mouse oncogenesis model was used to verify the invasion and metastasis mechanisms in vivo.
Results We found that Kindlin-2 expression was upregulated at both mRNA and protein levels in GC cells cocultured with TAMs, associated with higher invasion rate. Kindlin-2 knockdown reduced the invasion rate of GC cells under coculture condition. TGFβ2 secreted by TAMs regulated the expression of Kindlin-2 through the transcription factor NF-кB. TAMs thus participated in the progression of GC through the TGFβ2/NF-κB/Kindlin-2 axis. Kindlin-2 expression and TAM infiltration were significantly positively correlated with TNM stage, and patients with high Kindlin-2 expression had significantly poorer overall survival than patients with low Kindlin-2 expression. Furthermore, Kindlin-2 promoted the invasion of GC cells in vivo.
Conclusions This study elucidates the mechanism of TAMs participating in GC cell invasion and metastasis through the TGFβ2/NF-κB/Kindlin-2 axis, providing a possibility for new treatment options and approaches.
Collapse
Affiliation(s)
- Zhu Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Yang Yang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Yancheng Cui
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China.,Department of Surgery, Helsinki University Central Hospital, and University of Helsinki, Helsinki 00290, Finland
| | - Chao Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Zhiyong Lai
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Yansen Li
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Wei Zhang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Harri Mustonen
- Department of Surgery, Helsinki University Central Hospital, and University of Helsinki, Helsinki 00290, Finland
| | - Pauli Puolakkainen
- Department of Surgery, Helsinki University Central Hospital, and University of Helsinki, Helsinki 00290, Finland
| | - Yingjiang Ye
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Kewei Jiang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Zhanlong Shen
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| | - Shan Wang
- Department of Gastroenterological Surgery, Peking University People's Hospital, Beijing 100044, China.,Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
28
|
Koo BW, Lim DJ, Oh AY, Na HS. Retrospective Comparison between the Effects of Propofol and Inhalation Anesthetics on Postoperative Recurrence of Early- and Intermediate-Stage Hepatocellular Carcinoma. Med Princ Pract 2020; 29:422-428. [PMID: 32074612 PMCID: PMC7511682 DOI: 10.1159/000506637] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 02/17/2020] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Previous studies have reported that propofol has antitumor, anti-inflammatory, and antioxidant effects in addition to its anesthetic properties. To confirm this, a retrospective investigation was conducted to determine whether different anesthetic agents, particularly propofol and inhalation anesthetics, have an effect on the recurrence of hepatocellular carcinoma (HCC) in patients who were diagnosed with primary HCC and underwent laparoscopic hepatectomy. SUBJECTS AND METHODS Patients with Barcelona Clinic Liver Cancer stages 0, A, and B HCC, who underwent laparoscopic hepatic resection, were enrolled in this study. Post-operative HCC recurrence, which was determined from postoperative liver CT, was evaluated 24 months postoperatively with respect to the main anesthetic agents. The characteristics of HCC and other patient-related or surgery-related variables were evaluated together. RESULTS AND CONCLUSION During the 24-month period after hepatic resection, less HCC patients in the propofol group than in the inhalation group recurred (p = 0.046). The mean time to recurrence was 20.8 months (95% CI, 19.7-22.0) and 19.1 months (95% CI, 17.8-20.4) in the propofol group and the inhalation group, respectively. In addition, multivariable Cox proportional regression analysis revealed that the propofol group showed significantly decreased recurrence versus the inhalation group (hazard ratio, 0.57; 95% CI, 0.47-0.69; p = 0.029). When propofol was used as the main general anesthetic agent for laparoscopic hepatic resection, the postoperative 2-year recurrence rate decreased in early- and intermediate-stage HCC.
Collapse
Affiliation(s)
- Bon-Wook Koo
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Dae-Jin Lim
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ah-Young Oh
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hyo-Seok Na
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea,
| |
Collapse
|
29
|
SIRT5 inhibits bovine preadipocyte differentiation and lipid deposition by activating AMPK and repressing MAPK signal pathways. Genomics 2019; 112:1065-1076. [PMID: 31816429 DOI: 10.1016/j.ygeno.2019.12.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/30/2022]
Abstract
SIRT5 (sirtuin 5) is located in the mitochondria and plays an important role in biological processes such as maintaining the balance of lipid metabolism and promoting fatty acid oxidation mobilization. In this study, the bovine preadipocyte differentiation and obese mouse models were constructed; combined with transcriptome sequencing (RNA-seq) and morphological identification, the regulatory and molecular mechanisms underlying the effects of SIRT5 on bovine preadipocyte differentiation and lipid metabolism were studied. The results reveal that during the differentiation of preadipocytes, SIRT5 inhibited the expression of key genes that promote lipid formation and differentiation in fatty acid biosynthesis and PPAR pathways. SIRT5 significantly activated the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway and repressed the mitogen-activated protein kinase (MAPK) pathway. Eventually, SIRT5 significantly inhibited the differentiation of bovine preadipocytes and simultaneously inhibited lipid synthesis and lipid deposition in adipocytes. The verification experiments performed using obese mice also yielded consistent results in vivo.
Collapse
|
30
|
The Combination of Astragalus membranaceus and Angelica sinensis Inhibits Lung Cancer and Cachexia through Its Immunomodulatory Function. JOURNAL OF ONCOLOGY 2019; 2019:9206951. [PMID: 31781219 PMCID: PMC6875282 DOI: 10.1155/2019/9206951] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 08/11/2019] [Indexed: 12/24/2022]
Abstract
Lung cancer and its related cachexia are the leading cause of cancer death in the world. In this study, we report the inhibitory effect of the combined therapy of Astragalus membranaceus and Angelica sinensis, on tumor growth and cachexia in tumor-bearing mice. Lewis lung carcinoma cells were inoculated into male C57BL/6 and CAnN.Cg-Foxn1nu nude mice. After tumor inoculation, mice were fed orally by the combination of AM and AS in different doses. In C57BL/6 mice, the combination of AM and AS significantly inhibited the growth of cancer tumor and prevented the loss of body weight and skeletal muscle. It also diminished the formation of free radicals and cytokines, stimulated the differentiation of NK and Tc cells, and rebalanced the ratios of Th/Tc cells, Th1/Th2 cytokines, and M1/M2 tumor-associated macrophages. The herbal combination also downregulated the expression of NFκΒ, STAT3, HIF-1α, and VEGF in tumors. In contrast, the findings were not observed in the nude mice. Therefore, the combination of AM and AS is confirmed to inhibit the progression of lung cancer, cancer cachexia, and cancer inflammation through the immunomodulatory function.
Collapse
|
31
|
Radstake FDW, Raaijmakers EAL, Luttge R, Zinger S, Frimat JP. CALIMA: The semi-automated open-source calcium imaging analyzer. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2019; 179:104991. [PMID: 31443860 PMCID: PMC6718774 DOI: 10.1016/j.cmpb.2019.104991] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 07/09/2019] [Accepted: 07/19/2019] [Indexed: 05/05/2023]
Abstract
BACKGROUND AND OBJECTIVE Ever since its discovery, calcium imaging has proven its worth in discovering new insights into the mechanisms of cellular communication. Yet, the analysis of the data generated by calcium imaging experiments demands a large amount of time from researchers. Tools enabling automated and semi-automated analysis are available, but often they allow automating only a part of the data analysis process. Therefore, we developed CALIMA (https://aethelraed.nl/calima), a free and open-source standalone software tool that provides an opportunity to quickly detect cells, to obtain the calcium spikes, and to determine the underlying network structure of neuronal cell cultures. METHODS Owing to the difference of Gaussians algorithm applied for the cell detection, CALIMA is able to detect regions of interest (ROIs) quickly. The z-scoring algorithm provides a means to set the requirements for spike detection, and the neuronal connections can be reconstructed by analyzing the cross-correlation between the cellular activity. We evaluated CALIMA's reliability, speed, and functionality with a special focus on neuronal cell detection and network reconstruction. The evaluation was performed by using real-life data such as a known example dataset (cultured primary rat cortical neurons, University of Pennsylvania) and by analyzing video graphic footage of in vitro brain cell samples (SH-SY5Y neuroblastoma cultures, one sample with synchronous neuron firing). The obtained results were compared to the corresponding outcomes observed on same datasets for other similar software solutions. Moreover, we compared the results of segmentation and peak detection analysis, the ones obtained using CALIMA and those acquired manually. RESULTS CALIMA was able to detect the cells in the cultures within seconds. The average sensitivity was 82% across the datasets checked, comparing favorably with the alternative software solutions. Using the correct parameters, CALIMA's Ca-spikes detection sensitivity reached 96%. Lastly, neuronal networks were reconstructed by combining the data on the ROI's activity and the cell's positions, finding the most likely inter-cell connections. CONCLUSIONS We found that CALIMA proved to be a robust and fast tool to analyze the data of experiments for the digital reconstruction of the neuronal cellular network while being able to process the analysis steps with minimal user input required and in a time efficient manner.
Collapse
Affiliation(s)
- F D W Radstake
- Department of Electrical Engineering, Signal Processing Systems Group, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - E A L Raaijmakers
- Department of Electrical Engineering, Electromagnetics Group, Eindhoven University of Technology, Eindhoven, the Netherlands.
| | - R Luttge
- Department of Mechanical Engineering, Neuro-Nanoscale Engineering Group, Microsystems Section & ICMS Institute for Complex Molecular Systems, Eindhoven University of Technology, the Netherlands
| | - S Zinger
- Department of Electrical Engineering, Signal Processing Systems Group, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - J P Frimat
- Department of Mechanical Engineering, Neuro-Nanoscale Engineering Group, Microsystems Section & ICMS Institute for Complex Molecular Systems, Eindhoven University of Technology, the Netherlands
| |
Collapse
|
32
|
Microfluidic models of physiological or pathological flow shear stress for cell biology, disease modeling and drug development. Trends Analyt Chem 2019. [DOI: 10.1016/j.trac.2019.06.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
33
|
Rezazadeh F, Salehi S, Rezaee M. Salivary Level of Trace Element in Oral Lichen Planus, A Premalignant Condition. Asian Pac J Cancer Prev 2019; 20:2009-2013. [PMID: 31350958 PMCID: PMC6745238 DOI: 10.31557/apjcp.2019.20.7.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Indexed: 12/15/2022] Open
Abstract
Background: Oral Lichen Planus (OLP) is a common inflammatory disease of unknown cause that knows also as a premalignant condition. Recent studies contributed nutritional factors to the pathogenesis of many autoimmune disorders. Objective: The aim of present study was to investigate the salivary levels of Magnesium (Mg), Calcium (Ca), Iron (Fe), Zinc (Zn) and Copper (Cu) in this disorder. Materials and Methods: In this observational case-control study, the unstimulated saliva of 40 patients with OLP and 40 age and sex matched healthy control subjects were collected. The salivary levels of Mg, Ca, Fe, Zn and Cu were determined using ICP-AES. Results: Mg levels were significantly lower in OLP patients than in healthy controls, although it didn’t differ significantly between erosive and non-erosive types of Lichen Planus. Also there was no meaningful relationship between the levels of Ca, Fe, Zn and Cu in case and control groups. Conclusion: Trace elements such as Mg may have a role in ethiopathogenesis of OLP.
Collapse
Affiliation(s)
- Fahimeh Rezazadeh
- Oral and Dental Disease Research Center, Oral and Maxillofacial Medicine Department, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Sadaf Salehi
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mostafa Rezaee
- Oral and Dental Disease Research Center, Oral and Maxillofacial Medicine Department, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
34
|
MiR-940 inhibits migration and invasion of tongue squamous cell carcinoma via regulatingCXCR2/NF-κB system-mediated epithelial-mesenchymal transition. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1359-1369. [PMID: 31214736 DOI: 10.1007/s00210-019-01671-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 05/28/2019] [Indexed: 02/06/2023]
Abstract
Tongue squamous cell carcinoma (TSCC) is one of the most common incident oral cancers, which is accompanied by high rate of metastasis and recurrence. It has been demonstrated that elevated interleukin-8 (IL-8) promoted metastasis of various cancers via regulating epithelial-mesenchymal transition (EMT) process, whereas the accurate mechanism is left to be elucidated. The present work was aimed to investigate the role of microRNA-940 (miR-940)/C-X-C chemokine receptor type 2 (CXCR2) system in the metastasis ability and EMT process of IL-8-treated TSCC cells and further explore the underlying mechanisms. We found that miR-940 up-regulation inhibited IL-8-induced migration and invasion, which could be deprived by CXCR2 silence. We also observed that miR-940 suppressed epithelial marker E-cadherin expression while increased mesenchymal markers N-cadherin and Twist levels in IL-8-stimulated TSCC cells. Besides, IL-8-induced invasion and EMT process of TSCC cells were impeded in the present of the NF-κB inhibitor, PDTC or BAY117082. In conclusion, our data demonstrated that miR-940/CXCR2 system regulated the metastasis of TSCC cells via NF-κB-induced EMT process.
Collapse
|
35
|
Wang Q, Pan F, Li S, Huang R, Wang X, Wang S, Liao X, Li D, Zhang L. The prognostic value of the proteasome activator subunit gene family in skin cutaneous melanoma. J Cancer 2019; 10:2205-2219. [PMID: 31258724 PMCID: PMC6584401 DOI: 10.7150/jca.30612] [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: 10/12/2018] [Accepted: 04/25/2019] [Indexed: 12/25/2022] Open
Abstract
Background: The functional significance of the proteasome activator subunit (PSME) gene family in the pathogenesis of skin cutaneous melanoma (SKCM) remains to be elucidated. Materials and methods: Clinical data for patients with SKCM, including expression levels of PSME genes, were extracted from TCGA. GO term and KEGG pathway enrichment analyses were performed. Correlations between the expression levels of PSME genes in SKCM were evaluated with the Pearson correlation coefficient. Functional and enrichment analyses were conducted using DAVID. Univariate and multivariate survival analyses adjusted by Cox regression were used to construct a prognostic signature. The mechanisms underlying the association between PSME gene expression and overall survival (OS) were explored with gene set enrichment analysis. Joint-effects survival analysis was performed to evaluate the clinical value of the prognostic signature. Results: The median expression levels of PSME1, PSME2 and PSME3 were significantly higher in SKCM than in normal skin. PSME1, PSME2, and PSME3 were significantly enriched in several biological processes and pathways including cell adhesion, adherens junction organization, regulation of autophagy, cellular protein localization, the cell cycle, apoptosis, and the Wnt and NF-κB pathways. High expression levels of PSME1 and PSME2 combined with a low expression level of PSME3 was associated with favorable OS. Conclusion: Knowledge of the expression levels of the PSME gene family could provide a sensitive strategy for predicting prognosis in SKCM.
Collapse
Affiliation(s)
- Qiaoqi Wang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Fuqiang Pan
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Sizhu Li
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Rui Huang
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiangkun Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shijun Wang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, People's Republic of China
| | - Xiwen Liao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Dong Li
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Liming Zhang
- Department of Medical Cosmetology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| |
Collapse
|
36
|
Ahmed M, Lorence E, Wang J, Jung D, Zhang L, Nomie K, Wang M. Interrogating B cell signaling pathways: A quest for novel therapies for mantle cell lymphoma. Sci Signal 2019; 12:12/567/eaat4105. [PMID: 30723172 DOI: 10.1126/scisignal.aat4105] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mantle cell lymphoma (MCL) is an aggressive B cell lymphoma that is largely chemoresistant. Ibrutinib, a drug that inhibits Bruton's tyrosine kinase (BTK), has improved the overall survival of patients with MCL; however, resistance to ibrutinib has emerged as a decisive, negative factor in the prognosis of MCL. Adopting a more patient-centric therapeutic approach that incorporates applied genomics and interrogation of B cell signaling pathways may offer an alternative route to reach durable remission in patients with MCL. Although targeting genetic variants in MCL is not yet feasible in the clinical setting, the identification and targeting of increasingly active B cell signaling pathways may be a viable therapeutic strategy that may improve patient outcomes. Genome-editing tools and sequencing platforms could play dominant roles in patient-centric approaches of treatment in the future, potentially improving clinical outcomes for patients with MCL.
Collapse
Affiliation(s)
- Makhdum Ahmed
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA.
| | - Elizabeth Lorence
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA.
| | - Jeffrey Wang
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA
| | - Dayoung Jung
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA
| | - Liang Zhang
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA
| | - Krystle Nomie
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA
| | - Michael Wang
- Department of Lymphoma and Myeloma, the University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard Unit 0429, Houston, TX 77030-4009, USA.
| |
Collapse
|
37
|
Schillaci O, Scimeca M, Trivigno D, Chiaravalloti A, Facchetti S, Anemona L, Bonfiglio R, Santeusanio G, Tancredi V, Bonanno E, Urbano N, Mauriello A. Prostate cancer and inflammation: A new molecular imaging challenge in the era of personalized medicine. Nucl Med Biol 2019; 68-69:66-79. [PMID: 30770226 DOI: 10.1016/j.nucmedbio.2019.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/23/2018] [Accepted: 01/14/2019] [Indexed: 12/21/2022]
Abstract
The relationship between cancer and inflammation is one of the most important fields for both clinical and translational research. Despite numerous studies reported interesting and solid data about the prognostic value of the presence of inflammatory infiltrate in cancers, the biological role of inflammation in prostate cancer development is not yet fully clarified. The characterization of molecular pathways that connect altered inflammatory response and prostate cancer progression can provide the scientific rationale for the identification of new prognostic and predictive biomarkers. Specifically, the detection of infiltrating immune cells or related-cytokines by histology and/or by molecular imaging techniques could profoundly change the management of prostate cancer patients. In this context, the anatomic pathology and imaging diagnostic teamwork can provide a valuable support for the validation of new targets for diagnosis and therapy of prostate cancer lesions associated to the inflammatory infiltrate. The aim of this review is to summarize the current literature about the role of molecular imaging technique and anatomic pathology in the study of the mutual interaction occurring between prostate cancer and inflammation. Specifically, we reported the more recent advances in molecular imaging and histological methods for the early detection of prostate lesions associated to the inflammatory infiltrate.
Collapse
Affiliation(s)
- Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Manuel Scimeca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; University of San Raffaele, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Donata Trivigno
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Via Montpellier 1, Rome 00133, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Simone Facchetti
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Lucia Anemona
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Rita Bonfiglio
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Giuseppe Santeusanio
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Virginia Tancredi
- University of San Raffaele, Via di Val Cannuta 247, 00166 Rome, Italy; Department of Systems Medicine, School of Sport and Exercise Sciences, University of Rome "Tor Vergata", Rome, Italy
| | - Elena Bonanno
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| | - Nicoletta Urbano
- Nuclear Medicine, Policlinico "Tor Vergata", Viale Oxford 81, 00133 Rome, Italy
| | - Alessandro Mauriello
- Department of Experimental Medicine and Surgery, University "Tor Vergata", Via Montpellier 1, Rome 00133, Italy
| |
Collapse
|
38
|
Wang R, Hao W, Pan L, Boldogh I, Ba X. The roles of base excision repair enzyme OGG1 in gene expression. Cell Mol Life Sci 2018; 75:3741-3750. [PMID: 30043138 PMCID: PMC6154017 DOI: 10.1007/s00018-018-2887-8] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/11/2018] [Accepted: 07/19/2018] [Indexed: 12/13/2022]
Abstract
Modifications of DNA strands and nucleobases-both induced and accidental-are associated with unfavorable consequences including loss or gain in genetic information and mutations. Therefore, DNA repair proteins have essential roles in keeping genome fidelity. Recently, mounting evidence supports that 8-oxoguanine (8-oxoG), one of the most abundant genomic base modifications generated by reactive oxygen and nitrogen species, along with its cognate repair protein 8-oxoguanine DNA glycosylase1 (OGG1), has distinct roles in gene expression through transcription modulation or signal transduction. Binding to 8-oxoG located in gene regulatory regions, OGG1 acts as a transcription modulator, which can control transcription factor homing, induce allosteric transition of G-quadruplex structure, or recruit chromatin remodelers. In addition, post-repair complex formed between OGG1 and its repair product-free 8-oxoG increases the levels of active small GTPases and induces downstream signaling cascades to trigger gene expressions. The present review discusses how cells exploit damaged guanine base(s) and the authentic repair protein to orchestrate a profile of various transcriptomes in redox-regulated biological processes.
Collapse
Affiliation(s)
- Ruoxi Wang
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, Jilin, China
- School of Life Science, Northeast Normal University, Changchun, 130024, Jilin, China
| | - Wenjing Hao
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, Jilin, China
- School of Life Science, Northeast Normal University, Changchun, 130024, Jilin, China
| | - Lang Pan
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, Jilin, China
- Department of Physiology, Xiangya Medicine School in Central South University, Changsha, 410078, Hunan, China
| | - Istvan Boldogh
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
- Sealy Center for Molecular Medicine, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Xueqing Ba
- Key Laboratory of Molecular Epigenetics of Ministry of Education, Institute of Genetics and Cytology, Northeast Normal University, 5268 Renmin Street, Changchun, 130024, Jilin, China.
- School of Life Science, Northeast Normal University, Changchun, 130024, Jilin, China.
| |
Collapse
|
39
|
Vlahopoulos S, Adamaki M, Khoury N, Zoumpourlis V, Boldogh I. Roles of DNA repair enzyme OGG1 in innate immunity and its significance for lung cancer. Pharmacol Ther 2018; 194:59-72. [PMID: 30240635 DOI: 10.1016/j.pharmthera.2018.09.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cytokines are pivotal mediators of the immune response, and their coordinated expression protects host tissue from excessive damage and oxidant stress. Nevertheless, the development of lung pathology, including asthma, chronic obstructive pulmonary disease, and ozone-induced lung injury, is associated with oxidant stress; as evidence, there is a significant increase in levels of the modified guanine base 7,8-dihydro-8-oxoguanine (8-oxoG) in the genome. 8-OxoG is primarily recognized by 8-oxoguanine glycosylase 1 (OGG1), which catalyzes the first step in the DNA base excision repair pathway. However, oxidant stress in the cell transiently halts enzymatic activity of substrate-bound OGG1. The stalled OGG1 facilitates DNA binding of transactivators, including NF-κB, to their cognate sites to enable expression of cytokines and chemokines, with ensuing recruitments of inflammatory cells. Hence, defective OGG1 will modulate the coordination between innate and adaptive immunity through excessive oxidant stress and cytokine dysregulation. Both oxidant stress and cytokine dysregulation constitute key elements of oncogenesis by KRAS, which is mechanistically coupled to OGG1. Thus, analysis of the mechanism by which OGG1 modulates gene expression helps discern between beneficial and detrimental effects of oxidant stress, exposes a missing functional link as a marker, and yields a novel target for lung cancer.
Collapse
Affiliation(s)
- Spiros Vlahopoulos
- Ηoremeio Research Laboratory, First Department of Paediatrics, National and Kapodistrian University of Athens, 11527 Athens, Greece.
| | - Maria Adamaki
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Nikolas Khoury
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Biology, Medicinal Chemistry and Biotechnology, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Istvan Boldogh
- Departments of Microbiology and Immunology and the Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
40
|
Zhong L, Xu F, Chen F. Arsenic trioxide induces the apoptosis and decreases NF-κB expression in lymphoma cell lines. Oncol Lett 2018; 16:6267-6274. [PMID: 30333888 PMCID: PMC6176401 DOI: 10.3892/ol.2018.9424] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 04/16/2018] [Indexed: 12/11/2022] Open
Abstract
Lymphoma is a type of cancer that develops from certain immune system cells. Arsenic trioxide (ATO) has attracted wide attention owing to its antitumor activities. However, the role of ATO in tumorigenesis and progression remains to be investigated. In the present study, the antitumor function of ATO was investigated in in lymphoma Raji and Jurkat cell lines and the effect of ATO on nuclear factor (NF)-κB expression levels. A Cell Counting kit-8 assay was used to assess cellular proliferation and the degree of cell apoptosis was measured by flow cytometric analysis; these assays demonstrated that ATO inhibited proliferation and promoted the apoptosis of Raji and Jurkat cells in a dose- and time-dependent manner. Western blot analysis revealed that ATO treatment affected the expression of apoptosis-associated proteins by downregulating the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2) and upregulating the pro-apoptotic protein Bcl-2-associatedX and the degree of caspase-3 cleavage. In addition, reverse transcription-quantitative polymerase chain reaction and western blot analysis showed that the mRNA and protein expression levels of NF-κB were downregulated significantly following treatment with 2 µM ATO for 24, 48 and 72 h in the two cell lines. Additionally, immunofluorescence staining indicated that NF-κB expression diminished following ATO treatment in a time-dependent manner. These data indicated that ATO inhibited the proliferation of lymphoma cells by inducing cell apoptosis, which may be associated with the inhibition of the NF-κB signaling pathway. The findings of the present study may lay the foundation for developing a personalized medicine strategy using ATO via targeting of the NF-κB signaling pathway in lymphoma.
Collapse
Affiliation(s)
- Lu Zhong
- Department of Hematology, Renji Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200001, P.R. China
| | - Fei Xu
- Department of Ultrasound, The Affiliated Shuhuang Hospital of University of Shanghai Chinese Medicine, Shanghai 201111, P.R. China
| | - Fangyuan Chen
- Department of Hematology, Renji Hospital Affiliated to Shanghai Jiaotong University, Shanghai 200001, P.R. China
| |
Collapse
|
41
|
Al-Tamimi J, Semlali A, Hassan I, Ebaid H, Alhazza IM, Mehdi SH, Al-Khalifa M, Alanazi MS. Samsum Ant Venom Exerts Anticancer Activity Through Immunomodulation In Vitro and In Vivo. Cancer Biother Radiopharm 2018; 33:65-73. [PMID: 29634416 DOI: 10.1089/cbr.2017.2400] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Samsum ant venom (SAV) is a rich repertoire of natural compounds with tremendous pharmacological properties. The present work explores its antineoplastic activity in different cell lines followed by its confirmation in vivo. The cell lines, HepG2, MCF-7, and LoVo showed the differential dose-dependent antineoplastic effect with an increased level of significant cytokines, including Interleukin (IL)-1β, IL-6, and IL-8 and transcription factor, Nuclear factor-kappa B (NF-κB). However, the venom was more effective on HepG2 and MCF-7 cells than LoVo cells. Furthermore, the extract was administered to four groups (n = 8) of rats. Group I was taken as a control without any treatment, whereas group II received CCl4 (1 mL/kg) for induction of mild hepatoma. Group III was given 100 μg/kg of SAV twice a week for 1 month. Group IV was pretreated with the CCl4 (like group II) followed by dosing with SAV (100 μg/kg) for 2 months as per the authors' prestandardized dosing schedule. Intriguingly, the rats of group IV demonstrated significant decrease in key cytokines, IL-1β and IL-6, as well as the transcription factors, including Tumor Necrosis Factor-alpha (TNF-α), NF-κB, and Inhibitor-kappa B (I-κB) as compared with group II. Furthermore, increase in IL-10 and First apoptosis signal (FAS) in the same group confirmed that SAV induces apoptosis at the given dose through immunomodulation leading to enhanced tumor killing in vivo. Hence, SAV has an excellent antineoplastic activity that can be directly used to treat certain types of cancer. Moreover, study of its ingredients can pave ways to design novel anticancer drugs. However, further in-depth investigation is required before its clinical trials.
Collapse
Affiliation(s)
- Jameel Al-Tamimi
- 1 Department of Zoology, College of Science, King Saud University , Riyadh, Saudi Arabia
| | - Abdelhabib Semlali
- 2 Genome Research Chair, Department of Biochemistry, College of Science, King Saud University , Riyadh, Saudi Arabia
| | - Iftekhar Hassan
- 1 Department of Zoology, College of Science, King Saud University , Riyadh, Saudi Arabia
| | - Hossam Ebaid
- 1 Department of Zoology, College of Science, King Saud University , Riyadh, Saudi Arabia
| | - Ibrahim M Alhazza
- 1 Department of Zoology, College of Science, King Saud University , Riyadh, Saudi Arabia
| | - Syed H Mehdi
- 3 Department of Geriatrics, Donald W Reynolds Institute of Aging , UAMS Little Rock, Little Rock, Arkansas
| | - Mohammed Al-Khalifa
- 1 Department of Zoology, College of Science, King Saud University , Riyadh, Saudi Arabia
| | - Mohammad S Alanazi
- 2 Genome Research Chair, Department of Biochemistry, College of Science, King Saud University , Riyadh, Saudi Arabia
| |
Collapse
|
42
|
Jiang W, Huang RZ, Zhang J, Guo T, Zhang MT, Huang XC, Zhang B, Liao ZX, Sun J, Wang HS. Discovery of antitumor ursolic acid long-chain diamine derivatives as potent inhibitors of NF-κB. Bioorg Chem 2018; 79:265-276. [PMID: 29778798 DOI: 10.1016/j.bioorg.2018.05.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 02/09/2023]
Abstract
A series of inhibitors of NF-κB based on ursolic acid (UA) derivatives containing long-chain diamine moieties were designed and synthesized as well as evaluated the antitumor effects. These compounds exhibited significant inhibitory activity to the NF-κB with IC50 values at micromolar concentrations in A549 lung cancer cell line. Among them, compound 8c exerted potent activity against the test tumor cell lines including multidrug resistant human cancer lines, with the IC50 values ranged from 5.22 to 8.95 μM. Moreover, compound 8c successfully suppressed the migration of A549 cells. Related mechanism study indicated compound 8c caused cell cycle arrest at G1 phase and triggered apoptosis in A549 cells through blockage of NF-κB signalling pathway. Molecular docking study revealed that key interactions between 8c and the active site of NF-κB in which the bulky and strongly electrophilic group of long-chain diamine moieties were important for improving activity.
Collapse
Affiliation(s)
- Wei Jiang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Ri-Zhen Huang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Jing Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Tong Guo
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Meng-Ting Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Xiao-Chao Huang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Bin Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China
| | - Zhi-Xin Liao
- Jiangsu Province Hi-Tech Key Laboratory for Biomedical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing 211189, PR China.
| | - Jing Sun
- Chinese Acad Sci, Northwest Inst Plateau Biol, Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources, Xining 810000, PR China.
| | - Heng-Shan Wang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences of Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
43
|
Kan S, Zhang W, Mao J, Wang M, Ni L, Zhang M, Zhang Q, Chen J. NF-κB activation contributes to parathyroid cell proliferation in chronic kidney disease. J Nephrol 2018; 31:941-951. [DOI: 10.1007/s40620-018-0530-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 05/26/2018] [Indexed: 12/20/2022]
|
44
|
Liu C, Zhang F, Liu H, Wei F. NF-kB mediated CX3CL1 activation in the dorsal root ganglion contributes to the maintenance of neuropathic pain induced in adult male Sprague Dawley rats. Acta Cir Bras 2018; 33:619-628. [DOI: 10.1590/s0102-865020180070000007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/22/2018] [Indexed: 01/23/2023] Open
|
45
|
Long L, Pang XX, Lei F, Zhang JS, Wang W, Liao LD, Xu XE, He JZ, Wu JY, Wu ZY, Wang LD, Lin DC, Li EM, Xu LY. SLC52A3 expression is activated by NF-κB p65/Rel-B and serves as a prognostic biomarker in esophageal cancer. Cell Mol Life Sci 2018; 75:2643-2661. [PMID: 29428966 PMCID: PMC6003972 DOI: 10.1007/s00018-018-2757-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/16/2018] [Accepted: 01/18/2018] [Indexed: 02/05/2023]
Abstract
The human riboflavin transporter-3 (encoded by SLC52A3) plays a prominent role in riboflavin absorption. Interestingly, abnormal expression patterns of SLC52A3 in multiple types of human cancers have been recently noted. However, the molecular mechanisms underlying its dysregulation remain unclear. In this study, we find that SLC52A3 has two transcript variants that differ in the transcriptional start site, and encode different proteins: SLC52A3a and SLC52A3b. Importantly, aberrant expressions of SLC52A3 are associated with stepwise development of esophageal squamous cell carcinoma (ESCC) as well as the survival rates of ESCC patients. Functionally, SLC52A3a, but not SLC52A3b, strongly promotes the proliferation and colony formation of ESCC cells. Furthermore, SLC52A3 5'-flanking regions contain NF-κB p65/Rel-B-binding sites, which are crucial for mediating SLC52A3 transcriptional activity in ESCC cells. Chromatin immunoprecipitation and electrophoretic mobility shift assay reveal that p65/Rel-B bind to 5'-flanking regions of SLC52A3. Accordingly, NF-κB signaling upregulates SLC52A3 transcription upon TNFα stimulation. Taken together, these results elucidate the mechanisms underlying SLC52A3 overexpression in ESCC. More importantly, our findings identify SLC52A3 as both a predictive and prognostic biomarker for this deadly cancer.
Collapse
Affiliation(s)
- Lin Long
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xiao-Xiao Pang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Fei Lei
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Jia-Sheng Zhang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Wei Wang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Xiu-E Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jian-Zhong He
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Jian-Yi Wu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Zhi-Yong Wu
- Department of Oncology Surgery, Shantou Central Hospital, Affiliated Shantou Hospital of Sun Yat-Sen University, Shantou, China
| | - Li-Dong Wang
- Henan Key Laboratory for Esophageal Cancer Research, Department of Basic Oncology and Pathology at College of Medicine, The First and The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - De-Chen Lin
- Guangdong Province Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Research Center of Medicine, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China.
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China.
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China.
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| |
Collapse
|
46
|
Collignon E, Canale A, Al Wardi C, Bizet M, Calonne E, Dedeurwaerder S, Garaud S, Naveaux C, Barham W, Wilson A, Bouchat S, Hubert P, Van Lint C, Yull F, Sotiriou C, Willard-Gallo K, Noel A, Fuks F. Immunity drives TET1 regulation in cancer through NF-κB. SCIENCE ADVANCES 2018; 4:eaap7309. [PMID: 29938218 PMCID: PMC6010319 DOI: 10.1126/sciadv.aap7309] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 05/09/2018] [Indexed: 05/27/2023]
Abstract
Ten-eleven translocation enzymes (TET1, TET2, and TET3), which induce DNA demethylation and gene regulation by converting 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC), are often down-regulated in cancer. We uncover, in basal-like breast cancer (BLBC), genome-wide 5hmC changes related to TET1 regulation. We further demonstrate that TET1 repression is associated with high expression of immune markers and high infiltration by immune cells. We identify in BLBC tissues an anticorrelation between TET1 expression and the major immunoregulator family nuclear factor κB (NF-κB). In vitro and in mice, TET1 is down-regulated in breast cancer cells upon NF-κB activation through binding of p65 to its consensus sequence in the TET1 promoter. We lastly show that these findings extend to other cancer types, including melanoma, lung, and thyroid cancers. Together, our data suggest a novel mode of regulation for TET1 in cancer and highlight a new paradigm in which the immune system can influence cancer cell epigenetics.
Collapse
Affiliation(s)
- Evelyne Collignon
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB (Université libre de Bruxelles)–Cancer Research Center (U-CRC), ULB, Brussels, Belgium
| | - Annalisa Canale
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)–Cancer, University of Liège, Liège, Belgium
| | - Clémence Al Wardi
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB (Université libre de Bruxelles)–Cancer Research Center (U-CRC), ULB, Brussels, Belgium
| | - Martin Bizet
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB (Université libre de Bruxelles)–Cancer Research Center (U-CRC), ULB, Brussels, Belgium
| | - Emilie Calonne
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB (Université libre de Bruxelles)–Cancer Research Center (U-CRC), ULB, Brussels, Belgium
| | - Sarah Dedeurwaerder
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB (Université libre de Bruxelles)–Cancer Research Center (U-CRC), ULB, Brussels, Belgium
| | - Soizic Garaud
- Molecular Immunology Unit, Institut Jules Bordet, ULB, Brussels, Belgium
| | - Céline Naveaux
- Molecular Immunology Unit, Institut Jules Bordet, ULB, Brussels, Belgium
| | - Whitney Barham
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Andrew Wilson
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Sophie Bouchat
- Service of Molecular Virology, Department of Molecular Biology, U-CRC, ULB, Gosselies, Belgium
| | - Pascale Hubert
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Carine Van Lint
- Service of Molecular Virology, Department of Molecular Biology, U-CRC, ULB, Gosselies, Belgium
| | - Fiona Yull
- Department of Cancer Biology, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, TN 37232, USA
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Jules Bordet Institute, ULB, Brussels, Belgium
| | | | - Agnès Noel
- Laboratory of Tumor and Development Biology, Groupe Interdisciplinaire de Génoprotéomique Appliquée (GIGA)–Cancer, University of Liège, Liège, Belgium
| | - François Fuks
- Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB (Université libre de Bruxelles)–Cancer Research Center (U-CRC), ULB, Brussels, Belgium
| |
Collapse
|
47
|
Gulic T, Laskarin G, Dominovic M, Glavan Gacanin L, Babarović E, Rubesa Z, Haller H, Rukavina D. Granulysin-mediated apoptosis of trophoblasts in blighted ovum and missed abortion. Am J Reprod Immunol 2018; 80:e12978. [PMID: 29774968 DOI: 10.1111/aji.12978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/18/2018] [Indexed: 01/09/2023] Open
Abstract
PROBLEM Granulysin (GNLY) is a cytotoxic molecule mostly present in decidual natural killer (NK) cells. Blighted ovum (BO) and missed abortion (MA) represent the early pathological pregnancies with hindered development of the embryoblast or a dead embryo. We investigated the GNLY-mediated apoptotic mechanism potentially responsible for delayed termination of pregnancy. METHOD OF STUDY We performed immunohistological and immunofluorescence labeling of decidual tissues (GNLY, Apaf-1, NF-κB). NKG2A expression was analyzed by flow cytometry and GNLY mRNA by RT-qPCR. RESULTS The GNLY labeling intensity (H score) was lower in the nuclei of trophoblast cells in BO and MA. GNLY gene levels were inversely detected in BO and MA. A decreased decidual NK cell percentage was found in MA. NK cells from pathological pregnancies expressed lower NKG2A levels. The highest frequency of Apaf-1 was found in trophoblast cells of MA. NF-kB was highly expressed in decidual cells of BO. CONCLUSION The reduced activation of GNLY-mediated killing might be implicated in the slower rejection of trophoblast cells in BO and MA. A decreased authentic decidual NK cell number could be responsible for low cytotoxicity against trophoblast cells in MA. In BO, trophoblast cells have a higher survival potential due to increased NF-kB expression.
Collapse
Affiliation(s)
- Tamara Gulic
- Medical Faculty, Department of Physiology and Immunology, University of Rijeka, Rijeka, Croatia
| | - Gordana Laskarin
- Medical Faculty, Department of Physiology and Immunology, University of Rijeka, Rijeka, Croatia.,Division of Rheumatology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, Opatija, Croatia
| | - Marin Dominovic
- Medical Faculty, Department of Physiology and Immunology, University of Rijeka, Rijeka, Croatia
| | - Lana Glavan Gacanin
- Department of Obstetrics and Gynecology, Clinical Hospital, University of Rijeka, Rijeka, Croatia
| | - Emina Babarović
- Department of Pathology, Medical Faculty, University of Rijeka, Rijeka, Croatia
| | - Zeljka Rubesa
- Department of Cardiology, Clinical Hospital Center Rijeka, Rijeka, Croatia
| | - Herman Haller
- Department of Obstetrics and Gynecology, Clinical Hospital, University of Rijeka, Rijeka, Croatia
| | - Daniel Rukavina
- Medical Faculty, Department of Physiology and Immunology, University of Rijeka, Rijeka, Croatia.,Department of Biomedical Sciences in Rijeka, Croatian Academy of Sciences and Arts, Rijeka, Croatia
| |
Collapse
|
48
|
Pratt R, Stapelberg NJC. Early warning biomarkers in major depressive disorder: a strategic approach to a testing question. Biomarkers 2018; 23:563-572. [DOI: 10.1080/1354750x.2018.1463563] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- R. Pratt
- Consultation Liaison Psychiatry, Nepean Hospital, Penrith, Australia
| | - N. J. C. Stapelberg
- Faculty of Health Sciences and Medicine, Bond University and Gold Coast Hospital and Health Service, Southport, Australia
| |
Collapse
|
49
|
Rig-G is a growth inhibitory factor of lung cancer cells that suppresses STAT3 and NF-κB. Oncotarget 2018; 7:66032-66050. [PMID: 27602766 PMCID: PMC5323212 DOI: 10.18632/oncotarget.11797] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 08/24/2016] [Indexed: 12/19/2022] Open
Abstract
The expression of the retinoic acid-induced G (Rig-G) gene, an all trans retinoic acid (ATRA)-inducible gene, was observed in multiple cancer cells, including lung cancer cells. However, whether Rig-G is a tumor suppressor in lung cancer is unknown. Here, we found that ectopic expression of Rig-G can lead to a significant decrease in proliferation of lung cancer cells, resulting in an inhibition of tumor growth. Rig-G knockdown results in a modest increase in cell proliferation, as well as confers an increase in colony formation. Furthermore, transcriptome and pathway analyses of cancer cells revealed a fundamental impact of Rig-G on various growth signaling pathways, including the NF-κB pathway. Rig-G inhibits NF-κB activity by suppressing STAT3 in lung cancer cells. The downregulation of miR21 and miR181b-1 and subsequent activation of PTEN/Akt and CYLD/IκB signaling axis leading to decreased NF-κB activity required to maintain the tumor-inhibiting effect of Rig-G.. Our findings contribute to a better understanding of the antitumor effect mechanism of Rig-G, as well as offer a novel strategy for lung cancer therapy.
Collapse
|
50
|
De Silva E, Kim H. Drug-induced thrombocytopenia: Focus on platelet apoptosis. Chem Biol Interact 2018; 284:1-11. [PMID: 29410286 DOI: 10.1016/j.cbi.2018.01.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 12/23/2017] [Accepted: 01/18/2018] [Indexed: 12/16/2022]
Abstract
Thrombocytopenia is a serious and potentially fatal complication of drug therapy that results either from a decrease in bone marrow platelet production or the excessive destruction of circulating platelets. Although multiple mechanisms are responsible for deregulated platelet clearance, the role of programmed platelet death (apoptosis) in drug-induced thrombocytopenia has been relatively under-investigated until recently. Here we review apoptotic signaling pathways in platelets, with a focus on current data that provide mechanistic insights into drug-induced apoptosis and thrombocytopenia.
Collapse
Affiliation(s)
- Enoli De Silva
- Centre for Blood Research, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Hugh Kim
- Centre for Blood Research, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada; Faculty of Dentistry, University of British Columbia, Vancouver, Canada.
| |
Collapse
|