1
|
Qian C, Zhang X, Tian YS, Yuan L, Wei Q, Yang Y, Xu M, Wang X, Sun M. Coptisine inhibits esophageal carcinoma growth by modulating pyroptosis via inhibition of HGF/c-Met signaling. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03765-6. [PMID: 39792166 DOI: 10.1007/s00210-024-03765-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Esophageal carcinoma is a highly prevalent malignancy worldwide. The present study aimed to investigate the mechanism by which the natural compound coptisine affects pyroptosis in esophageal squamous cell carcinoma (ESCC). The expression of c-Met in ESCC patients was assessed by immunohistochemical analysis of tissue microarrays. Natural drugs that bind to c-Met were identified by screening and molecular docking. The effect of coptisine on the proliferation of ESCC cells was detected by CCK-8 and colony formation assays. Cell cycle progression and cell apoptosis were detected by flow cytometry. The levels of mRNAs related to pyroptosis and miR-21 after coptisine treatment were assessed via real-time quantitative PCR. The effect of pyroptosis was evaluated by reactive oxygen species level detection and transmission electron microscopy (TEM) analysis. The expression of proteins related to pyroptosis and the HGF/c-Met pathway was detected by western blotting. A xenograft tumor model was established, and the inhibitory effect of coptisine was evaluated by observing tumor growth. The results showed that the highly expressed protein c-Met in esophageal cancer could bind with coptisine. Coptisine inhibited c-Met phosphorylation and proliferation in ESCC cells. Furthermore, coptisine inhibited the expression of downstream proteins of the HGF/c-Met signaling pathway and induced ROS generation. Tumor xenograft experiments demonstrated that coptisine effectively inhibited tumor growth by reducing the levels of pyroptosis-associated proteins. In conclusion, these findings indicate that inhibition of the HGF/c-Met signaling pathway suppresses pyroptosis to enhance the antitumor effect of coptisine in ESCC and support the potential use of coptisine for EC treatment.
Collapse
Affiliation(s)
- Chunmei Qian
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xing Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yu-Shi Tian
- Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Lin Yuan
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Qiao Wei
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yifu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
- Institute of Pathology, Fudan University, Shanghai, 200032, China.
| | - Xiaoyu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Menghong Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Shi XC, Zhang T, Li C, Guo CJ, Yang Q, Feng Y, Wang J, Qu CX. Impact of Nuclear Peripheral Chromatin Lamin LMNB1 Gene in the Proliferation and Migration of Glioma Cells. Neurochem Res 2024; 50:46. [PMID: 39636549 DOI: 10.1007/s11064-024-04298-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
The goal of this study is to explore the role of the LMNB1 gene in glioma. A cohort of 160 patients who underwent glioma surgery were randomly selected of this study. The LMNB1 expression was assessed employing immunohistochemical and real-time quantitative polymerase chain reaction methods. Initially, RNA interference technology was applied to suppress gene expression, followed by the evaluation of tumor cell proliferation, apoptosis, cell cycle dynamics, and migration. The underlying molecular mechanisms of LMNB1 function were examined by a human phospho-kinase array and immunoblotting. And we established the xenograft models to determine the effect of tumor growth as well as the degree of invasion in shLMNB1 mice. Elevated LMNB1 expression correlated with unfavorable overall survival and disease-free survival. A substantial inhibition in cell growth was observed subsequent to LMNB1 knockdown in SHG-44 and U251 glioma cells. SHG-44-shLMNB1 cells exhibited a reduction in the S phase population, along with an increase in cells in G1 and G2 phases. Similarly, shLMNB1 U251 cells showed fewer cells in the S phase and an elevation in cells in G1 phase. Notably, increased apoptosis was observed in U251-shLMNB1 cells and SHG-44-shLMNB1 cells. Wound healing and Transwell migration assays demonstrated a significant decrease in the migration rate of both SHG-44-shLMNB1 and U251-shLMNB1 cells. The phosphorylation levels of Akt1/2/3, as well as the expressions of PI3K, AKT, and p-AKT proteins, were reduced in the shLMNB1 group. Downregulation of LMNB1 repressed tumor progress in vivo. The silencing of LMNB1 was found to significantly reduce the proliferation of human glioma cells, induce apoptosis in tumor cells, impede the progression of the cell cycle, and inhibit the migration of tumor cells. Consequently, we hypothesize that LMNB1 promotes glioma cell proliferation through mechanisms involving the inhibition of tumor cell apoptosis, acceleration of the cell cycle, and enhancement of tumor cell migration. We found that LMNB1 exert critical roles in glioma progression may via regulation of PI3K/Akt signaling pathway. These observations suggest that LMNB1 holds clinical potential for diagnostic and prognostic applications in glioma, presenting novel targets for drug development.
Collapse
Affiliation(s)
- Xiang-Cheng Shi
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Ting Zhang
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Cheng Li
- The Pathology Department of Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, 030012, China
| | - Chen-Jia Guo
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Qin Yang
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Yao Feng
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Jie Wang
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Chong-Xiao Qu
- The Pathology Department of Shanxi Provincial People's Hospital, Taiyuan, 030012, China.
- Department of Pathology, Shanxi Provincial People's Hospital, No. 29 of Shuangtasi Road, Yingze District, Taiyuan, 030012, China.
| |
Collapse
|
3
|
Kan LK, Drill M, Jayakrishnan PC, Sequeira RP, Sanfilippo PG, McLean C, Hunn M, Williams DA, O'Brien TJ, Drummond KJ, Monif M. P2X7 receptor antagonism by AZ10606120 significantly depletes glioblastoma cancer stem cells in vitro. Brain Res Bull 2024; 215:110996. [PMID: 38857832 DOI: 10.1016/j.brainresbull.2024.110996] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma is the most aggressive and lethal primary brain malignancy with limited treatment options and poor prognosis. Self-renewing glioblastoma cancer stem cells (GSCs) facilitate tumour progression, resistance to conventional treatment and tumour recurrence. GSCs are resistant to standard treatments. There is a need for novel treatment alternatives that effectively target GSCs. The purinergic P2X receptor 7 (P2X7R) is expressed in glioblastomas and has been implicated in disease pathogenesis. However, the roles of P2X7R have not been comprehensively elucidated in conventional treatment-resistant GSCs. This study characterised P2X7R channel and pore function and investigated the effect of pharmacological P2X7R inhibition in GSCs. Immunofluorescence and live cell fluorescent dye uptake experiments revealed P2X7R expression, and channel and pore function in GSCs. Treatment of GSCs with the P2X7R antagonist, AZ10606120 (AZ), for 72 hours significantly reduced GSC numbers, compared to untreated cells. When compared with the effect of the first-line conventional chemotherapy, temozolomide (TMZ), GSCs treated with AZ had significantly lower cell numbers than TMZ-treated cultures, while TMZ treatment alone did not significantly deplete GSC numbers compared to the control. AZ treatment also induced significant lactate dehydrogenase release by GSCs, indicative of treatment-induced cytotoxic cell death. There were no significant differences in the expression of apoptotic markers, Annexin V and cleaved caspase-3, between AZ-treated cells and the control. Collectively, this study reveals for the first time functional P2X7R channel and pore in GSCs and significant GSC depletion following P2X7R inhibition by AZ. These results indicate that P2X7R inhibition may be a novel therapeutic alternative for glioblastoma, with effectiveness against GSCs resistant to conventional chemotherapy.
Collapse
Affiliation(s)
- Liyen K Kan
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Matthew Drill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | | | - Richard P Sequeira
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Paul G Sanfilippo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Catriona McLean
- Department of Pathology, The Alfred, Melbourne, Victoria, Australia
| | - Martin Hunn
- Department of Neurosurgery, The Alfred, Melbourne, Victoria, Australia
| | - David A Williams
- Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Neurology, The Alfred, Melbourne, Victoria, Australia
| | - Katharine J Drummond
- Department of Neurosurgery, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Mastura Monif
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Department of Physiology, The University of Melbourne, Melbourne, Victoria, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Victoria, Australia; Department of Neurology, The Alfred, Melbourne, Victoria, Australia.
| |
Collapse
|
4
|
Mendanha D, Casanova MR, Gimondi S, Ferreira H, Neves NM. Microfluidic-Derived Docosahexaenoic Acid Liposomes for Targeting Glioblastoma and Its Inflammatory Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2024; 16:40543-40554. [PMID: 39042828 PMCID: PMC11310905 DOI: 10.1021/acsami.4c01368] [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: 04/17/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024]
Abstract
Glioblastoma (GBM) is the most common malignant primary brain tumor, characterized by limited treatment options and a poor prognosis. Its aggressiveness is attributed not only to the uncontrolled proliferation and invasion of tumor cells but also to the complex interplay between these cells and the surrounding microenvironment. Within the tumor microenvironment, an intricate network of immune cells, stromal cells, and various signaling molecules creates a pro-inflammatory milieu that supports tumor growth and progression. Docosahexaenoic acid (DHA), an essential ω3 polyunsaturated fatty acid for brain function, is associated with anti-inflammatory and anticarcinogenic properties. Therefore, in this work, DHA liposomes were synthesized using a microfluidic platform to target and reduce the inflammatory environment of GBM. The liposomes were rapidly taken up by macrophages in a time-dependent manner without causing cytotoxicity. Moreover, DHA liposomes successfully downregulated the expression of inflammatory-associated genes (IL-6; IL-1β; TNFα; NF-κB, and STAT-1) and the secretion of key cytokines (IL-6 and TNFα) in stimulated macrophages and GBM cells. Conversely, no significant differences were observed in the expression of IL-10, an anti-inflammatory gene expressed in alternatively activated macrophages. Additionally, DHA liposomes were found to be more efficient in regulating the inflammatory profile of these cells compared with a free formulation of DHA. The nanomedicine platform established in this work opens new opportunities for developing liposomes incorporating DHA to target GBM and its inflammatory milieu.
Collapse
Affiliation(s)
- Daniel Mendanha
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Marta R. Casanova
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Sara Gimondi
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Helena Ferreira
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| | - Nuno M. Neves
- 3B’s
Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables
and Biomimetics, University of Minho, Headquarters of the European Institute
of Excellence on Tissue Engineering and Regenerative Medicine, AvePark,
Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal
- ICVS/3B’s-PT
Government Associate Laboratory, 4805-017 Barco, Braga/Guimarães, Portugal
| |
Collapse
|
5
|
Meybodi SM, Ejlalidiz M, Manshadi MR, Raeisi M, Zarin M, Kalhor Z, Saberiyan M, Hamblin MR. Crosstalk between hypoxia-induced pyroptosis and immune escape in cancer: From mechanisms to therapy. Crit Rev Oncol Hematol 2024; 197:104340. [PMID: 38570176 DOI: 10.1016/j.critrevonc.2024.104340] [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: 02/08/2024] [Revised: 03/12/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Pyroptosis can be triggered through both canonical and non-canonical inflammasome pathways, involving the cleavage of gasdermin (GSDM) protein family members, like GSDMD and GSDME. The impact of pyroptosis on tumors is nuanced, because its role in regulating cancer progression and anti-tumor immunity may vary depending on the tumor type, stage, location, and immune status. However, pyroptosis cannot be simply categorized as promoting or inhibiting tumors based solely on whether it is acute or chronic in nature. The interplay between pyroptosis and cancer is intricate, with some evidence suggesting that chronic pyroptosis may facilitate tumor growth, while the acute induction of pyroptosis could stimulate anti-cancer immune responses. Tumor hypoxia activates hypoxia inducible factor (HIF) signaling to modulate pyroptosis and immune checkpoint expression. Targeting this hypoxia-pyroptosis-immune escape axis could be a promising therapeutic strategy. This review highlights the complex crosstalk between hypoxia, pyroptosis, and immune evasion in the TME.
Collapse
Affiliation(s)
| | - Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadsadegh Rezaeian Manshadi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Raeisi
- Clinical Research Developmental Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Zarin
- Department of Medical Genetics, Semnan University of Medical Sciences, Semnan, Iran
| | - Zahra Kalhor
- Department of Anatomical Sciences, Factulty of Medicine, Kurdistan University of Medical Scidnces, Sanandaj, Iran
| | - Mohammadreza Saberiyan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran; Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa.
| |
Collapse
|
6
|
Yi L, Lin X, She X, Gao W, Wu M. Chronic stress as an emerging risk factor for the development and progression of glioma. Chin Med J (Engl) 2024; 137:394-407. [PMID: 38238191 PMCID: PMC10876262 DOI: 10.1097/cm9.0000000000002976] [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: 05/13/2023] [Indexed: 02/21/2024] Open
Abstract
ABSTRACT Gliomas tend to have a poor prognosis and are the most common primary malignant tumors of the central nervous system. Compared with patients with other cancers, glioma patients often suffer from increased levels of psychological stress, such as anxiety and fear. Chronic stress (CS) is thought to impact glioma profoundly. However, because of the complex mechanisms underlying CS and variability in individual tolerance, the role of CS in glioma remains unclear. This review suggests a new proposal to redivide the stress system into two parts. Neuronal activity is dominant upstream. Stress-signaling molecules produced by the neuroendocrine system are dominant downstream. We discuss the underlying molecular mechanisms by which CS impacts glioma. Potential pharmacological treatments are also summarized from the therapeutic perspective of CS.
Collapse
Affiliation(s)
- Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
| | - Xiaoling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wei Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Minghua Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
7
|
Chen Q, Sun Y, Wang S, Xu J. New prospects of cancer therapy based on pyroptosis and pyroptosis inducers. Apoptosis 2024; 29:66-85. [PMID: 37943371 DOI: 10.1007/s10495-023-01906-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2023] [Indexed: 11/10/2023]
Abstract
Pyroptosis is a gasdermin-mediated programmed cell death (PCD) pathway. It differs from apoptosis because of the secretion of inflammatory molecules. Pyroptosis is closely associated with various malignant tumors. Recent studies have demonstrated that pyroptosis can either inhibit or promote the development of malignant tumors, depending on the cell type (immune or cancer cells) and duration and severity of the process. This review summarizes the molecular mechanisms of pyroptosis, its relationship with malignancies, and focuses on current pyroptosis inducers and their significance in cancer treatment. The molecules involved in the pyroptosis signaling pathway could serve as therapeutic targets for the development of novel drugs for cancer therapy. In addition, we analyzed the potential of combining pyroptosis with conventional anticancer techniques as a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Qiaoyun Chen
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuxiang Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225000, China
| | - Siliang Wang
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China.
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| | - Jingyan Xu
- China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, 210008, China.
- Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China.
| |
Collapse
|
8
|
Lu J, Gu B, Lu W, Liu J, Lu J. Lnc-ANRIL modulates the immune response associated with NF-κB pathway in LPS-stimulated bovine mammary epithelial cells. Immun Inflamm Dis 2023; 11:e1125. [PMID: 38156382 PMCID: PMC10740337 DOI: 10.1002/iid3.1125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 11/18/2023] [Accepted: 12/05/2023] [Indexed: 12/30/2023] Open
Abstract
BACKGROUND The antisense noncoding RNA in the INK4 locus (ANRIL) has been confirmed related to multiple disease progression, but the role and exact mechanisms of lnc-ANRIL in lipopolysaccharide (LPS)-induced inflammation of bovine mammary epithelial cells (MAC-T) remain unclear. AIMS This manuscript focused on expounding the functional role of lnc-ANRIL through experiments performed in MAC-T. METHODS At the in vitro level, we established a Bovine mammary epithelial cell (BMEC) cell model of mastitis by LPS treatment. Transfection of siRNA was examined by immunofluorescence localization and RT-qPCR. CCK8, clonogenic assay and EdU were used to detect the proliferation ability of the cells. Cell cycle and apoptosis were detected by flow cytometry and Western blot. The levels of inflammatory factors and oxidative stress markers were detected by ELISA kits. RESULTS Cell Counting Kit-8, colony formation, and 5-ethynyl-20-deoxyuridine were adopted and the data illustrated that LPS could significantly suppress the cell proliferation, while knockdown of lnc-ANRIL expression obviously promoted MAC-T cell proliferation compared with LPS or LPS + si-NC group. Flow cytometry analysis demonstrated that lnc-ANRIL could induce MAC-T cell apoptosis. In addition, downregulation of lnc-ANRIL affected LPS-induced immune response by regulating inflammatory factor expressions and modulating the nuclear factor kappa B (NF-κB) axis in MAC-T cells. CONCLUSION Our results suggest that lnc-ANRIL is involved in the regulation of cell proliferation, cell cycle, and cell apoptosis of MAC-T cells, and plays an important role in the inflammatory and immune response of MAC-T cells through the regulation of the NF-κB pathway, proposing new therapeutic strategies for the treatment of innate immune response-related disease such as bovine mastitis.
Collapse
Affiliation(s)
- Jinye Lu
- College of Pet TechnologyJiangsu Agri‐Animal Husbandry Vocational CollegeTaizhouChina
| | - Beibei Gu
- Integrated Technical Service CenterTaizhou CustomsTaizhouChina
| | - Wei Lu
- College of Pet TechnologyJiangsu Agri‐Animal Husbandry Vocational CollegeTaizhouChina
| | - Jing Liu
- College of Pet TechnologyJiangsu Agri‐Animal Husbandry Vocational CollegeTaizhouChina
| | - Jiang Lu
- College of Pet TechnologyJiangsu Agri‐Animal Husbandry Vocational CollegeTaizhouChina
| |
Collapse
|
9
|
Chen Z, Giotti B, Kaluzova M, Vallcorba MP, Rawat K, Price G, Herting CJ, Pinero G, Cristea S, Ross JL, Ackley J, Maximov V, Szulzewsky F, Thomason W, Marquez-Ropero M, Angione A, Nichols N, Tsankova NM, Michor F, Shayakhmetov DM, Gutmann DH, Tsankov AM, Hambardzumyan D. A paracrine circuit of IL-1β/IL-1R1 between myeloid and tumor cells drives genotype-dependent glioblastoma progression. J Clin Invest 2023; 133:e163802. [PMID: 37733448 PMCID: PMC10645395 DOI: 10.1172/jci163802] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/19/2023] [Indexed: 09/23/2023] Open
Abstract
Monocytes and monocyte-derived macrophages (MDMs) from blood circulation infiltrate glioblastoma (GBM) and promote growth. Here, we show that PDGFB-driven GBM cells induce the expression of the potent proinflammatory cytokine IL-1β in MDM, which engages IL-1R1 in tumor cells, activates the NF-κB pathway, and subsequently leads to induction of monocyte chemoattractant proteins (MCPs). Thus, a feedforward paracrine circuit of IL-1β/IL-1R1 between tumors and MDM creates an interdependence driving PDGFB-driven GBM progression. Genetic loss or locally antagonizing IL-1β/IL-1R1 leads to reduced MDM infiltration, diminished tumor growth, and reduced exhausted CD8+ T cells and thereby extends the survival of tumor-bearing mice. In contrast to IL-1β, IL-1α exhibits antitumor effects. Genetic deletion of Il1a/b is associated with decreased recruitment of lymphoid cells and loss-of-interferon signaling in various immune populations and subsets of malignant cells and is associated with decreased survival time of PDGFB-driven tumor-bearing mice. In contrast to PDGFB-driven GBM, Nf1-silenced tumors have a constitutively active NF-κB pathway, which drives the expression of MCPs to recruit monocytes into tumors. These results indicate local antagonism of IL-1β could be considered as an effective therapy specifically for proneural GBM.
Collapse
Affiliation(s)
- Zhihong Chen
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Milota Kaluzova
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Department of Neurology, Rutgers University, New Brunswick, New Jersey, USA
| | - Montse Puigdelloses Vallcorba
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Kavita Rawat
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Gabrielle Price
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Cameron J. Herting
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Gonzalo Pinero
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
| | - James L. Ross
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Emory University Department of Microbiology and Immunology, Emory Vaccine Center, Atlanta, Georgia, USA
| | - James Ackley
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Victor Maximov
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
| | - Frank Szulzewsky
- Department of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Wes Thomason
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Mar Marquez-Ropero
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Angelo Angione
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | | | - Nadejda M. Tsankova
- Department of Pathology and Molecular and Cell-Based Medicine, Mount Sinai Icahn School of Medicine, New York, New York, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts, USA
- The Ludwig Center at Harvard, Boston, Massachusetts, USA
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Dmitry M. Shayakhmetov
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Lowance Center for Human Immunology and Emory Vaccine Center, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alexander M. Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Mount Sinai Icahn School of Medicine, New York, New York, USA
- Department of Pediatrics, AFLAC Cancer and Blood Disorders Center, Children’s Healthcare of Atlanta and Winship Cancer Institute, and
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, USA
- Department of Neurosurgery and
| |
Collapse
|
10
|
Miller CR, Hjelmeland AB. Breaking the feed forward inflammatory cytokine loop in the tumor microenvironment of PDGFB-driven glioblastomas. J Clin Invest 2023; 133:e175127. [PMID: 37966120 PMCID: PMC10645375 DOI: 10.1172/jci175127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023] Open
Abstract
Glioblastoma (GBM) tumor-associated macrophages (TAMs) provide a major immune cell population contributing to growth and immunosuppression via the production of proinflammatory factors, including IL-1. In this issue of the JCI, Chen, Giotti, and colleagues investigated loss of ll1b in the immune tumor microenvironment (TME) in GBM models driven by PDGFB expression and Nf1 knockdown. Survival was only improved in PDGFB-driven GBM models, suggesting that tumor cell genotype influenced the immune TME. IL-1β in the TME increased PDGFB-driven GBM growth by increasing tumor-derived NF-κB, expression of monocyte chemoattractants, and increased infiltration of bone marrow-derived myeloid cells (BMDMs). In contrast, no requirement for IL-1β was evident in Nf1-silenced tumors due to high basal levels of NF-κB and monocyte chemoattractants and increased infiltration of BMDM and TAMs. Notably, treatment of mice bearing PDGFB-driven GBM with anti-IL-1β or an IL1R1 antagonist extended survival. These findings suggest that effective clinical immunotherapy may require differential targeting strategies.
Collapse
Affiliation(s)
- C. Ryan Miller
- Department of Pathology, Division of Neuropathology and O’Neal Comprehensive Cancer Center, and
| | - Anita B. Hjelmeland
- Department of Cell, Developmental, and Integrative Biology and O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
11
|
Caruso A, Gelsomino L, Panza S, Accattatis FM, Naimo GD, Barone I, Giordano C, Catalano S, Andò S. Leptin: A Heavyweight Player in Obesity-Related Cancers. Biomolecules 2023; 13:1084. [PMID: 37509120 PMCID: PMC10377641 DOI: 10.3390/biom13071084] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/04/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Obesity, defined as the abnormal or excessive expansion of white adipose tissue, has reached pandemic proportions and is recognized as an important health concern since it is a common root for several comorbidities, including malignancies. Indeed, the current knowledge of the white adipose tissue, which shifts its role from an energy storage tissue to an important endocrine and metabolic organ, has opened up new avenues for the discovery of obesity's effects on tumor biology. In this review, we will report the epidemiological studies concerning the strong impact of obesity in several types of cancer and describe the mechanisms underlying the heterotypic signals between cancer cell lines and adipocytes, with particular emphasis on inflammation, the insulin/IGF-1 axis, and adipokines. Among the adipokines, we will further describe the in vitro, in vivo, and clinical data concerning the role of leptin, recognized as one of the most important mediators of obesity-associated cancers. In fact, leptin physiologically regulates energy metabolism, appetite, and reproduction, and several studies have also described the role of leptin in affecting cancer development and progression. Finally, we will summarize the newest pharmacological strategies aimed at mitigating the protumorigenic effects of leptin, underlining their mechanisms of action.
Collapse
Affiliation(s)
- Amanda Caruso
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Luca Gelsomino
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Felice Maria Accattatis
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Giuseppina Daniela Naimo
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Cinzia Giordano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, Via P Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
- Centro Sanitario, Via P. Bucci, University of Calabria, Arcavacata di Rende (CS), 87036 Cosenza, Italy
| |
Collapse
|
12
|
Toedebusch RG, Wei NW, Simafranca KT, Furth-Jacobus JA, Brust-Mascher I, Stewart SL, Dickinson PJ, Woolard KD, Li CF, Vernau KM, Meyers FJ, Toedebusch CM. Intra- and Intertumoral Microglia/Macrophage Infiltration and Their Associated Molecular Signature Is Highly Variable in Canine Oligodendroglioma: A Preliminary Evaluation. Vet Sci 2023; 10:403. [PMID: 37368789 PMCID: PMC10303632 DOI: 10.3390/vetsci10060403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The goal of this study was to define the glioma-associated microglia/macrophage (GAM) response and associated molecular landscape in canine oligodendrogliomas. Here, we quantified the intratumoral GAM density of low- and high-grade oligodendrogliomas compared to that of a normal brain, as well as the intratumoral concentration of several known GAM-derived pro-tumorigenic molecules in high-grade oligodendrogliomas compared to that in a normal brain. Our analysis demonstrated marked intra- and intertumoral heterogeneity of GAM infiltration. Correspondingly, we observed significant variability in the intratumoral concentrations of several GAM-associated molecules, unlike what we previously observed in high-grade astrocytomas. However, high-grade oligodendroglioma tumor homogenates (n = 6) exhibited an increase in the pro-tumorigenic molecules hepatocyte growth factor receptor (HGFR) and vascular endothelial growth factor (VEGF), as we observed in high-grade astrocytomas. Moreover, neoplastic oligodendrocytes displayed robust expression of GAL-3, a chimeric galectin implicated in driving immunosuppression in human glioblastoma. While this work identifies shared putative therapeutic targets across canine glioma subtypes (HGFR, GAL-3), it highlights several key differences in the immune landscape. Therefore, a continued effort to develop a comprehensive understanding of the immune microenvironment within each subtype is necessary to inform therapeutic strategies going forward.
Collapse
Affiliation(s)
- Ryan G. Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Ning-Wei Wei
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Kulani T. Simafranca
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Jennie A. Furth-Jacobus
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Ingrid Brust-Mascher
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Susan L. Stewart
- Division of Biostatistics, School of Medicine, University of California, Davis, CA 95616, USA;
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
| | - Peter J. Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
| | - Kevin D. Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616, USA;
| | - Chai-Fei Li
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Karen M. Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
| | - Frederick J. Meyers
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
- Department of Internal Medicine, Division of Hematology and Oncology, Center for Precision Medicine, Microbiology, and Immunology, School of Medicine, University of California, Sacramento, CA 95817, USA
| | - Christine M. Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616, USA; (R.G.T.); (N.-W.W.); (K.T.S.); (J.A.F.-J.); (P.J.D.); (C.-F.L.); (K.M.V.)
- UC Davis Comprehensive Cancer Center, Sacramento, CA 95817, USA;
| |
Collapse
|
13
|
Kan LK, Drill M, Jayakrishnan PC, Sequeira RP, Galea E, Todaro M, Sanfilippo PG, Hunn M, Williams DA, O'Brien TJ, Drummond KJ, Monif M. P2X7 receptor antagonism by AZ10606120 significantly reduced in vitro tumour growth in human glioblastoma. Sci Rep 2023; 13:8435. [PMID: 37225786 DOI: 10.1038/s41598-023-35712-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/22/2023] [Indexed: 05/26/2023] Open
Abstract
Glioblastomas are highly aggressive and deadly brain tumours, with a median survival time of 14-18 months post-diagnosis. Current treatment modalities are limited and only modestly increase survival time. Effective therapeutic alternatives are urgently needed. The purinergic P2X7 receptor (P2X7R) is activated within the glioblastoma microenvironment and evidence suggests it contributes to tumour growth. Studies have implicated P2X7R involvement in a range of neoplasms, including glioblastomas, although the roles of P2X7R in the tumour milieu remain unclear. Here, we report a trophic, tumour-promoting role of P2X7R activation in both patient-derived primary glioblastoma cultures and the U251 human glioblastoma cell line, and demonstrate its inhibition reduces tumour growth in vitro. Primary glioblastoma and U251 cell cultures were treated with the specific P2X7R antagonist, AZ10606120 (AZ), for 72 h. The effects of AZ treatment were also compared to cells treated with the current first-line chemotherapeutic drug, temozolomide (TMZ), and a combination of both AZ and TMZ. P2X7R antagonism by AZ significantly depleted glioblastoma cell numbers compared to untreated cells, in both primary glioblastoma and U251 cultures. Notably, AZ treatment was more effective at tumour cell killing than TMZ. No synergistic effect between AZ and TMZ was observed. AZ treatment also significantly increased lactate dehydrogenase release in primary glioblastoma cultures, suggesting AZ-induced cellular cytotoxicity. Our results reveal a trophic role of P2X7R in glioblastoma. Importantly, these data highlight the potential for P2X7R inhibition as a novel and effective alternative therapeutic approach for patients with lethal glioblastomas.
Collapse
Affiliation(s)
- Liyen K Kan
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Matthew Drill
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | | | - Richard P Sequeira
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Emily Galea
- Department of Neurosurgery, The Alfred, Melbourne, VIC, Australia
| | - Marian Todaro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Neurology, The Alfred, Melbourne, VIC, Australia
| | - Paul G Sanfilippo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Martin Hunn
- Department of Neurosurgery, The Alfred, Melbourne, VIC, Australia
| | - David A Williams
- Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Department of Neurology, The Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Neurology, The Alfred, Melbourne, VIC, Australia
| | - Katharine J Drummond
- Department of Neurosurgery, The Royal Melbourne Hospital, Melbourne, VIC, Australia
| | - Mastura Monif
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Department of Physiology, The University of Melbourne, Melbourne, VIC, Australia.
- Department of Neurology, The Royal Melbourne Hospital, Melbourne, VIC, Australia.
- Department of Neurology, The Alfred, Melbourne, VIC, Australia.
| |
Collapse
|
14
|
Effect of Low-Carbohydrate Diet on Beta-Hydroxybutyrate Ketogenesis Metabolic Stimulation and Regulation of NLRP3 Ubiquitination in Obese Saudi Women. Nutrients 2023; 15:nu15040820. [PMID: 36839178 PMCID: PMC9958539 DOI: 10.3390/nu15040820] [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: 01/12/2023] [Revised: 01/25/2023] [Accepted: 02/03/2023] [Indexed: 02/08/2023] Open
Abstract
The effects of a ketogenic diet (KD) on anthropometric indices, the lipid profile, and the benefits of the ketone body beta-hydroxybutyrate (BHB) as an inhibitor of the NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome in obese women were investigated in this study. From January to March 2021, 23 obese adult women (n = 23) with an average age of 35.30 years and BMI of 33.96 kg/m2 followed a KD. Instructions for the KD were given to eligible participants, with a typical plan and a menu for all the main meals, snacks, and drinks permitted over seven days. They were also free to change meals according to their preferences provided that they followed the plan. The participants attended six times throughout the intervention for measurements of their anthropometric indices, BHB levels, interleukin-1beta (1L-1β) levels, and completion of a questionnaire (pre-intervention, mid-intervention, and post-intervention). Following the KD caused significant weight loss, a reduction in waist circumference and BHB levels, as well as a reduction in BMI and appetite. Cholesterol, triglycerides (TG), and high-density lipoprotein cholesterol (HDL-C) increased slightly. However, low-density lipoprotein cholesterol (LDL-C) in serum increased significantly (p < 0.05), and 1L-1β decreased significantly (p < 0.0001). The results show that the KD effectively encouraged weight loss and NLRP3 inflammasome inhibition. Based on the questionnaire results, it was found that a variety of physical symptoms, including overall energy, physical activity, mood, sleep, focus, skin conditions, and menstruation, had significantly improved.
Collapse
|
15
|
Chen J, Chen S, Li B, Zhou S, Lin H. A pyroptosis-related signature predicts prognosis and indicates immune microenvironment infiltration in glioma. Cancer Med 2023; 12:5071-5087. [PMID: 36161280 PMCID: PMC9972150 DOI: 10.1002/cam4.5247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Glioma, the most common malignant brain tumor, leads to high recurrence rates and disabilities in patients. Pyroptosis is an inflammasomes-induced programmed cell death in response to infection or chemotherapy. However, the role of pyroptosis in glioma has not yet been elucidated. METHODS RNA-seq data and clinical information of 660 gliomas and 847 samples were downloaded from the TCGA and CGGA, respectively. Then, data of 104 normal brain tissues was retrieved from the GTEx for differential expression analysis. Twelve pairs of peritumoral tissue and glioma samples were used for validation. Gene alteration status of differentially expressed pyroptosis-related regulators in gliomas was detected in cBioPortal algorithm. Consensus clustering was employed to classify gliomas based on differentially expressed pyroptosis-related regulators. Subsequently, a PS-signature was constructed using LASSO-congressional analysis for clinical application. The immune infiltration of glioma microenvironment (TME) was explored using ESTIMATE, CIBERSORT, and the other immune signatures. RESULTS cBioPortal algorithm revealed alteration of these regulators was correlated to better prognosis of gliomas. Then, our study showed that pyroptosis-related regulators can be used to sort out patients into two clusters with distinct prognostic outcome and immune status. Moreover, a PS-signature for predicting the prognosis of glioma patients was developed based on the identified subtypes. The high PS-score group showed more abundant inflammatory cell infiltration and stronger immune response, but with poorer prognosis of gliomas. CONCLUSION The findings of this study provide a therapeutic basis for future research on pyroptosis and unravel the relationship between pyroptosis and glioma prognosis. The risk signature can be utilized as a prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Jia Chen
- The Fourth People's Hospital of ChengduChengduChina
- The Clinical Hospital of Chengdu Brain Science InstituteMOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduChina
| | - Shanwei Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
- Shantou University Medical CollegeShantouChina
| | - Bingxian Li
- Department of Neurology, Shantou Central HospitalShantouChina
| | - Shaojiong Zhou
- Department of Neurology, Shantou Central HospitalShantouChina
| | - Han Lin
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
16
|
Koch MS, Zdioruk M, Nowicki MO, Griffith AM, Aguilar-Cordova E, Aguilar LK, Guzik BW, Barone F, Tak PP, Schregel K, Hoetker MS, Lederer JA, Chiocca EA, Tabatabai G, Lawler SE. Perturbing DDR signaling enhances cytotoxic effects of local oncolytic virotherapy and modulates the immune environment in glioma. Mol Ther Oncolytics 2022; 26:275-288. [PMID: 36032633 PMCID: PMC9391522 DOI: 10.1016/j.omto.2022.07.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/22/2022] [Indexed: 11/24/2022] Open
Abstract
CAN-2409 is a replication-deficient adenovirus encoding herpes simplex virus (HSV) thymidine kinase (tk) currently in clinical trials for treatment of glioblastoma. The expression of tk in transduced cancer cells results in conversion of the pro-drug ganciclovir into a toxic metabolite causing DNA damage, inducing immunogenic cell death and immune activation. We hypothesize that CAN-2409 combined with DNA-damage-response inhibitors could amplify tumor cell death, resulting in an improved response. We investigated the effects of ATR inhibitor AZD6738 in combination with CAN-2409 in vitro using cytotoxicity, cytokine, and fluorescence-activated cell sorting (FACS) assays in glioma cell lines and in vivo with an orthotopic syngeneic murine glioma model. Tumor immune infiltrates were analyzed by cytometry by time of flight (CyTOF). In vitro, we observed a significant increase in the DNA-damage marker γH2AX and decreased expression of PD-L1, pro-tumorigenic cytokines (interleukin-1β [IL-1β], IL-4), and ligand NKG2D after combination treatment compared with monotherapy or control. In vivo, long-term survival was increased after combination treatment (66.7%) compared with CAN-2409 (50%) and control. In a tumor re-challenge, long-term immunity after combination treatment was not improved. Our results suggest that ATR inhibition could amplify CAN-2409's efficacy in glioblastoma through increased DNA damage while having complex immunological ramifications, warranting further studies to determine the ideal conditions for maximized therapeutic benefit.
Collapse
Affiliation(s)
- Marilin S. Koch
- Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Mykola Zdioruk
- Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Michal O. Nowicki
- Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Alec M. Griffith
- Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | | | - Laura K. Aguilar
- Candel Therapeutics, 117 Kendrick St, Suite 450, Needham, MA 02494, USA
| | - Brian W. Guzik
- Candel Therapeutics, 117 Kendrick St, Suite 450, Needham, MA 02494, USA
| | - Francesca Barone
- Candel Therapeutics, 117 Kendrick St, Suite 450, Needham, MA 02494, USA
| | - Paul Peter Tak
- Candel Therapeutics, 117 Kendrick St, Suite 450, Needham, MA 02494, USA
| | - Katharina Schregel
- Department of Neuroradiology, University Hospital Heidelberg, Im Neuenheimer Feld 400, 69120 Heidelberg, Germany
| | - Michael S. Hoetker
- Department of Molecular Biology, Massachusetts General Hospital, 185 Cambridge St, Boston, MA 02114, USA
| | - James A. Lederer
- Department of Surgery, Brigham & Women’s Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - E. Antonio Chiocca
- Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| | - Ghazaleh Tabatabai
- Department of Neurology and Interdisciplinary Neuro-Oncology, University Hospital Tübingen, Hertie Institut for Clinical Brain Research, Eberhard Karls University Tübingen, Hoppe-Seyler-Straße 6, 72076 Tübingen, Germany
| | - Sean E. Lawler
- Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, 60 Fenwood Road, Boston, MA 02115, USA
| |
Collapse
|
17
|
Chen R, Wu W, Liu T, Zhao Y, Wang Y, Zhang H, Wang Z, Dai Z, Zhou X, Luo P, Zhang J, Liu Z, Zhang LY, Cheng Q. Large-scale bulk RNA-seq analysis defines immune evasion mechanism related to mast cell in gliomas. Front Immunol 2022; 13:914001. [PMID: 36159780 PMCID: PMC9492887 DOI: 10.3389/fimmu.2022.914001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Accumulating evidence has demonstrated that the immune cells have an emerging role in controlling anti-tumor immune responses and tumor progression. The comprehensive role of mast cell in glioma has not been illustrated yet. In this study, 1,991 diffuse glioma samples were collected from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA). xCell algorithm was employed to define the mast cell-related genes. Based on mast cell-related genes, gliomas were divided into two clusters with distinct clinical and immunological characteristics. The survival probability of cluster 1 was significantly lower than that of cluster 2 in the TCGA dataset, three CGGA datasets, and the Xiangya cohort. Meanwhile, the hypoxic and metabolic pathways were active in cluster 1, which were beneficial to the proliferation of tumor cells. A potent prognostic model based on mast cell was constructed. Via machine learning, DRG2 was screened out as a characteristic gene, which was demonstrated to predict treatment response and predict survival outcome in the Xiangya cohort. In conclusion, mast cells could be used as a potential effective prognostic factor for gliomas.
Collapse
Affiliation(s)
- Rui Chen
- Department of Neurosurgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Liu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Yihan Zhao
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yifan Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hao Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxi Zhou
- Department of Neurosurgery, Affiliated Nanhua Hospital, University of South China, Hengyang, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, China
| | - Li-Yang Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Quan Cheng
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
18
|
Chen N, Peng C, Li D. Epigenetic Underpinnings of Inflammation: A Key to Unlock the Tumor Microenvironment in Glioblastoma. Front Immunol 2022; 13:869307. [PMID: 35572545 PMCID: PMC9100418 DOI: 10.3389/fimmu.2022.869307] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/28/2022] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain tumor in adults, and immunotherapies and genetic therapies for GBM have evolved dramatically over the past decade, but GBM therapy is still facing a dilemma due to the high recurrence rate. The inflammatory microenvironment is a general signature of tumors that accelerates epigenetic changes in GBM and helps tumors avoid immunological surveillance. GBM tumor cells and glioma-associated microglia/macrophages are the primary contributors to the inflammatory condition, meanwhile the modification of epigenetic events including DNA methylation, non-coding RNAs, and histone methylation and deacetylases involved in this pathological process of GBM, finally result in exacerbating the proliferation, invasion, and migration of GBM. On the other hand, histone deacetylase inhibitors, DNA methyltransferases inhibitors, and RNA interference could reverse the inflammatory landscapes and inhibit GBM growth and invasion. Here, we systematically review the inflammatory-associated epigenetic changes and regulations in the microenvironment of GBM, aiming to provide a comprehensive epigenetic profile underlying the recognition of inflammation in GBM.
Collapse
Affiliation(s)
- Nian Chen
- State Key Laboratory of Southwestern Characteristic Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Characteristic Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dan Li
- State Key Laboratory of Southwestern Characteristic Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
19
|
Exogenous interleukin-1 beta promotes the proliferation and migration of HeLa cells via the MEK/ERK signaling pathway. Mol Biol Rep 2022; 49:3765-3772. [PMID: 35301650 DOI: 10.1007/s11033-022-07216-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/25/2022] [Accepted: 01/31/2022] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Interleukin-1 beta (IL-1β) is a crucial cytokine that has been implicated in cancer and metastasis development. However, its possible mechanistic role in cervical cancer remains unclear. This study aimed to investigate the functions of exogenous IL-1β in cervical cancer cell proliferation and migration. METHODS HeLa cell proliferation and migration were measured using MTT and Transwell assays. A lentivirus-mediated packaging system was used to construct an IL-1β overexpressing cell line. MEK/ERK signal transduction was inhibited by pretreatment with the MEK inhibitor PD98059. qRT-PCR and Western blotting were used to test the expression of relevant genes. RESULTS Exogenous IL-1β promoted the proliferation and migration of HeLa cells. In addition, overexpression of IL-1β in HeLa cells promoted cell proliferation. Mechanistically, exogenous IL-1β increased the phosphorylated MEK and ERK levels in HeLa cells and the expression of JUN, RELB, and NF-κB2. Alternatively, blockade of MEK inhibited the promoting proliferation effects of IL-1β and the expression of JUN, RELB, and NF-κB2. CONCLUSIONS Our data suggest that exogenous IL-1β regulates HeLa cell functions by regulating the MEK/ERK signaling pathway and by targeting JUN, RELB, and NF-κB2. Our study uncovered a potential association across IL-1β, cervical tumor development, and cancer progression.
Collapse
|
20
|
Xie J, Zhang Y, Jiang L. Role of Interleukin-1 in the pathogenesis of colorectal cancer: A brief look at anakinra therapy. Int Immunopharmacol 2022; 105:108577. [PMID: 35121226 DOI: 10.1016/j.intimp.2022.108577] [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: 11/13/2021] [Revised: 01/16/2022] [Accepted: 01/23/2022] [Indexed: 12/21/2022]
Abstract
Colorectal cancer (CRC) is known as one of the deadliest and most common cancers globally and causes nearly one million cancer deaths yearly. Like many malignancies, the immune system and its components play a crucial role in the pathogenesis of CRC. As multifunction mediators of the immune system, cytokines are involved in several inflammatory and anti-inflammatory responses. Interleukin-1 (IL-1) belongs to a family of 11 members and is involved in inflammatory responses. Beyond its biological role as a mediator of innate immune responses, it is also seen in chronic stress and inflammation and numerous pathological states. The role of IL-1 in malignancies can also be very significant because it has recently been shown that this cytokine can also be secreted from tumor cells and induce the recruitment of myeloid-derived immunosuppressive cells. As a result, the tumor microenvironment (TME) is affected and, despite being inflammatory, causes the onset and progression of tumor cells. Since surgery and chemotherapy are the first choices to treat patients with cancer, especially CRC, it is usually not well-prognosed, particularly in patients with metastatic lesions CRC. Therefore, targeted therapy may prolong the overall survival of CRC patients. Furthermore, evidence shows that anakinra has had satisfactory results in treating CRC. Therefore, this review summarized the role of IL-1 in the pathogenesis of CRC as well as immunotherapy based on inhibition of this cytokine in this type of cancer.
Collapse
Affiliation(s)
- Jun Xie
- Department of Colorectal Surgery, Affiliated Hospital of Shaoxing University, Shaoxing 312000, Zhejiang Province, China
| | - Yu Zhang
- Cancer Center, Department of Gastroenterology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014,Zhejiang, China
| | - Luxi Jiang
- Laboratory Medicine Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, Zhejiang, China.
| |
Collapse
|
21
|
do Nascimento RP, dos Santos BL, Amparo JAO, Soares JRP, da Silva KC, Santana MR, Almeida ÁMAN, da Silva VDA, Costa MDFD, Ulrich H, Moura-Neto V, Lopes GPDF, Costa SL. Neuroimmunomodulatory Properties of Flavonoids and Derivates: A Potential Action as Adjuvants for the Treatment of Glioblastoma. Pharmaceutics 2022; 14:pharmaceutics14010116. [PMID: 35057010 PMCID: PMC8778519 DOI: 10.3390/pharmaceutics14010116] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 01/27/2023] Open
Abstract
Glioblastomas (GBMs) are tumors that have a high ability to migrate, invade and proliferate in the healthy tissue, what greatly impairs their treatment. These characteristics are associated with the complex microenvironment, formed by the perivascular niche, which is also composed of several stromal cells including astrocytes, microglia, fibroblasts, pericytes and endothelial cells, supporting tumor progression. Further microglia and macrophages associated with GBMs infiltrate the tumor. These innate immune cells are meant to participate in tumor surveillance and eradication, but they become compromised by GBM cells and exploited in the process. In this review we discuss the context of the GBM microenvironment together with the actions of flavonoids, which have attracted scientific attention due to their pharmacological properties as possible anti-tumor agents. Flavonoids act on a variety of signaling pathways, counteracting the invasion process. Luteolin and rutin inhibit NFκB activation, reducing IL-6 production. Fisetin promotes tumor apoptosis, while inhibiting ADAM expression, reducing invasion. Naringenin reduces tumor invasion by down-regulating metalloproteinases expression. Apigenin and rutin induce apoptosis in C6 cells increasing TNFα, while decreasing IL-10 production, denoting a shift from the immunosuppressive Th2 to the Th1 profile. Overall, flavonoids should be further exploited for glioma therapy.
Collapse
Affiliation(s)
- Ravena Pereira do Nascimento
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Balbino Lino dos Santos
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- Academic College of Nurse, Department of Health, Federal University of Vale do São Francisco, Petrolina 56304-205, Pernambuco, Brazil
| | - Jéssika Alves Oliveira Amparo
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Janaina Ribeiro Pereira Soares
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Karina Costa da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Monique Reis Santana
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Áurea Maria Alves Nunes Almeida
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Victor Diógenes Amaral da Silva
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
| | - Maria de Fátima Dias Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
- Correspondence: (H.U.); (S.L.C.)
| | - Vivaldo Moura-Neto
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508-000, São Paulo, Brazil
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Rio de Janeiro, Brazil
- Paulo Niemeyer State Institute of the Brain, Rio de Janeiro 20230-024, Rio de Janeiro, Brazil
| | - Giselle Pinto de Faria Lopes
- Department of Marine Biotechnology, Admiral Paulo Moreira Institute for Sea Studies (IEAPM), Arraial do Cabo 28930-000, Rio de Janeiro, Brazil;
| | - Silvia Lima Costa
- Laboratory of Neurochemistry and Cell Biology, Department of Biochemistry and Biophysics, Institute of Health Sciences, Federal University of Bahia, Salvador 40110-902, Bahia, Brazil; (R.P.d.N.); (B.L.d.S.); (J.A.O.A.); (J.R.P.S.); (K.C.d.S.); (M.R.S.); (Á.M.A.N.A.); (V.D.A.d.S.); (M.d.F.D.C.)
- National Institute for Translational Neurosciences (INCT/CNPq INNT), Rio de Janeiro 21941-902, Rio de Janeiro, Brazil;
- Correspondence: (H.U.); (S.L.C.)
| |
Collapse
|
22
|
Maugeri G, D’Amico AG, Saccone S, Federico C, Rasà DM, Caltabiano R, Broggi G, Giunta S, Musumeci G, D’Agata V. Effect of PACAP on Hypoxia-Induced Angiogenesis and Epithelial-Mesenchymal Transition in Glioblastoma. Biomedicines 2021; 9:biomedicines9080965. [PMID: 34440169 PMCID: PMC8392618 DOI: 10.3390/biomedicines9080965] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 08/03/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) exerts different effects in various human cancer. In glioblastoma (GBM), PACAP has been shown to interfere with the hypoxic micro-environment through the modulation of hypoxia-inducible factors via PI3K/AKT and MAPK/ERK pathways inhibition. Considering that hypoxic tumor micro-environment is strictly linked to angiogenesis and Epithelial–Mesenchymal transition (EMT), in the present study, we have investigated the ability of PACAP to regulate these events. Results have demonstrated that PACAP and its related receptor, PAC1R, are expressed in hypoxic area of human GBM colocalizing either in epithelial or mesenchymal cells. By using an in vitro model of GBM cells, we have observed that PACAP interferes with hypoxic/angiogenic pathway by reducing vascular-endothelial growth factor (VEGF) release and inhibiting formation of vessel-like structures in H5V endothelial cells cultured with GBM-conditioned medium. Moreover, PACAP treatment decreased the expression of mesenchymal markers such as vimentin, matrix metalloproteinase 2 (MMP-2) and matrix metalloproteinase 9 (MMP-9) as well as CD44 in GBM cells by affecting their invasiveness. In conclusion, our study provides new insights regarding the multimodal role of PACAP in GBM malignancy.
Collapse
Affiliation(s)
- Grazia Maugeri
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (D.M.R.); (S.G.); (G.M.)
| | | | - Salvatore Saccone
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology, University of Catania, 95123 Catania, Italy; (S.S.); (C.F.)
| | - Concetta Federico
- Department of Biological, Geological and Environmental Sciences, Section of Animal Biology, University of Catania, 95123 Catania, Italy; (S.S.); (C.F.)
| | - Daniela Maria Rasà
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (D.M.R.); (S.G.); (G.M.)
- Department of Neuroscience Rita Levi Montalcini, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10124 Turin, Italy
| | - Rosario Caltabiano
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (R.C.); (G.B.)
| | - Giuseppe Broggi
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (R.C.); (G.B.)
| | - Salvatore Giunta
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (D.M.R.); (S.G.); (G.M.)
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (D.M.R.); (S.G.); (G.M.)
| | - Velia D’Agata
- Department of Biomedical and Biotechnological Sciences, Section of Anatomy, Histology and Movement Sciences, University of Catania, 95100 Catania, Italy; (G.M.); (D.M.R.); (S.G.); (G.M.)
- Correspondence: ; Tel.: +39-095-3782147; Fax: +39-095-3782046
| |
Collapse
|
23
|
Lu X, Guo T, Zhang X. Pyroptosis in Cancer: Friend or Foe? Cancers (Basel) 2021; 13:cancers13143620. [PMID: 34298833 PMCID: PMC8304688 DOI: 10.3390/cancers13143620] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Pyroptosis is a new form of programmed cell death that differs from apoptosis in terms of its release of inflammatory factors and its characteristic bubble-like morphology. Pyroptosis was first discovered in the process of immune defense against bacterial infection, but the field of research soon spread to other inflammatory diseases and cancer. As cancer constitutes a serious risk for public health, numerous studies investigating pyroptosis in cancer have been carried out during these years. Tumorigenesis and new therapeutic treatments have been the focus of much recent research. This review discusses the role of pyroptosis in tumorigenesis and its influence on tumor immunity. Abstract Pyroptosis is an inflammatory form of programmed cell death that is mediated by pore-forming proteins such as the gasdermin family (GSDMs), including GSDMA-E. Upon cleavage by activated caspases or granzyme proteases, the N-terminal of GSDMs oligomerizes in membranes to form pores, resulting in pyroptosis. Though all the gasdermin proteins have been studied in cancer, the role of pyroptosis in cancer remains mysterious, with conflicting findings. Numerous studies have shown that various stimuli, such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and chemotherapeutic drugs, could trigger pyroptosis when the cells express GSDMs. However, it is not clear whether pyroptosis in cancer induced by chemotherapeutic drugs or CAR T cell therapy is beneficial or harmful for anti-tumor immunity. This review discusses the discovery of pyroptosis as well as its role in inflammatory diseases and cancer, with an emphasis on tumor immunity.
Collapse
|
24
|
Abu Halim NH, Zakaria N, Theva Das K, Lin J, Lim MN, Fakiruddin KS, Yahaya BH. The Effects of Lentivirus-Mediated Gene Silencing of RARβ on the Stemness Capability of Non-Small Cell Lung Cancer. J Cancer 2021; 12:3468-3485. [PMID: 33995625 PMCID: PMC8120186 DOI: 10.7150/jca.50793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 12/01/2020] [Indexed: 11/05/2022] Open
Abstract
Retinoic acid receptor beta is a nuclear receptor protein that binds to retinoic acid (RA) to mediate cellular signalling in embryogenic morphogenesis, cell growth, and differentiation. However, the function of RARβ in cancer stem cells (CSCs) has yet to be determined. This study aimed to understand the role of RARβ in regulating cell growth and differentiation of lung cancer stem cells. Based on the clonogenic assay, spheroid assay, mRNA levels of stem cell transcription factors, and cell cycle being arrested at the G0/G1 phase, the suppression of RARβ resulted in significant inhibition of A549 parental cell growth. This finding was contradictory to the results seen in CSCs, where RARβ inhibition enhanced the cell growth of putative and non-putative CSCs. These results suggest that RARβ suppression may act as an essential regulator in A549 parental cells, but not in the CSCs population. The findings in this study demonstrated that the loss of RARβ promotes tumorigenicity in CSCs. Microarray analysis revealed that various cancer pathways were significantly activated following the suppression of RARβ. The changes seen might compensate for the loss of RARβ function, CSCs population's aggressiveness, which led to the CSCs population's aggressiveness. Thus, understanding the role of RARβ in regulating the stemness of CSCs may lead to targeted therapy for lung CSCs.
Collapse
Affiliation(s)
- Noor Hanis Abu Halim
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Sains@Bertam, Universiti Sains Malaysia, Kepala Batas Penang, 13200, Malaysia
| | - Norashikin Zakaria
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Sains@Bertam, Universiti Sains Malaysia, Kepala Batas Penang, 13200, Malaysia
| | - Kumitaa Theva Das
- Infectomics Cluster, Advanced Medical and Dental Institute (IPPT), Sains@Bertam, Universiti Sains Malaysia, Kepala Batas Penang, 13200, Malaysia
| | - Juntang Lin
- Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University (XXMU), Henan Province 453000, China.,Stem Cell and Biotherapy Technology Research Centre of Henan Province, Xinxiang Medical University (XXMU), Henan Province 453000, China
| | - Moon Nian Lim
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre (CaRC), Institute for Medical Research (IMR), National Institute of Health, Setia Alam, 40170 Shah Alam, Selangor
| | - Kamal Shaik Fakiruddin
- Stem Cell Laboratory, Haematology Unit, Cancer Research Centre (CaRC), Institute for Medical Research (IMR), National Institute of Health, Setia Alam, 40170 Shah Alam, Selangor
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Sains@Bertam, Universiti Sains Malaysia, Kepala Batas Penang, 13200, Malaysia
| |
Collapse
|
25
|
Toedebusch R, Grodzki AC, Dickinson PJ, Woolard K, Vinson N, Sturges B, Snyder J, Li CF, Nagasaka O, Consales B, Vernau K, Knipe M, Murthy V, Lein PJ, Toedebusch CM. Glioma-associated microglia/macrophages augment tumorigenicity in canine astrocytoma, a naturally occurring model of human glioma. Neurooncol Adv 2021; 3:vdab062. [PMID: 34131649 PMCID: PMC8193901 DOI: 10.1093/noajnl/vdab062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Glioma-associated microglia/macrophages (GAMs) markedly influence glioma progression. Under the influence of transforming growth factor beta (TGFB), GAMs are polarized toward a tumor-supportive phenotype. However, neither therapeutic targeting of GAM recruitment nor TGFB signaling demonstrated efficacy in glioma patients despite efficacy in preclinical models, underscoring the need for a comprehensive understanding of the TGFB/GAM axis. Spontaneously occurring canine gliomas share many features with human glioma and provide a complementary translational animal model for further study. Given the importance of GAM and TGFB in human glioma, the aims of this study were to further define the GAM-associated molecular profile and the relevance of TGFB signaling in canine glioma that may serve as the basis for future translational studies. METHODS GAM morphometry, levels of GAM-associated molecules, and the canonical TGFB signaling axis were compared in archived samples of canine astrocytomas versus normal canine brain. Furthermore, the effect of TGFB on the malignant phenotype of canine astrocytoma cells was evaluated. RESULTS GAMs diffusely infiltrated canine astrocytomas. GAM density was increased in high-grade tumors that correlated with a pro-tumorigenic molecular signature and upregulation of the canonical TGFB signaling axis. Moreover, TGFB1 enhanced the migration of canine astrocytoma cells in vitro. CONCLUSIONS Canine astrocytomas share a similar GAM-associated immune landscape with human adult glioma. Our data also support a contributing role for TGFB1 signaling in the malignant phenotype of canine astrocytoma. These data further support naturally occurring canine glioma as a valid model for the investigation of GAM-associated therapeutic strategies for human malignant glioma.
Collapse
Affiliation(s)
- Ryan Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Ana Cristina Grodzki
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Peter J Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Kevin Woolard
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Nicole Vinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Beverly Sturges
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - John Snyder
- Riemann Computing, LLC, St. Louis, Missouri, USA
| | - Chai-Fei Li
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Ori Nagasaka
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Blaire Consales
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Karen Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Marguerite Knipe
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Vishal Murthy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| | - Christine M Toedebusch
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, California, USA
| |
Collapse
|
26
|
Novel pyrano 1,3 oxazine based ligand inhibits the epigenetic reader hBRD2 in glioblastoma. Biochem J 2020; 477:2263-2279. [PMID: 32484211 DOI: 10.1042/bcj20200339] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/14/2022]
Abstract
Glioblastoma (GBM) is the most common primary brain malignancy, rarely amenable to treatment with a high recurrence rate. GBM are prone to develop resistance to the current repertoire of drugs, including the first-line chemotherapeutic agents with frequent recurrence, limiting therapeutic success. Recent clinical data has evidenced the BRD2 and BRD4 of the BET family proteins as the new druggable targets against GBM. In this relevance, we have discovered a compound (pyrano 1,3 oxazine derivative; NSC 328111; NS5) as an inhibitor of hBRD2 by the rational structure-based approach. The crystal structure of the complex, refined to 1.5 Å resolution, revealed that the NS5 ligand significantly binds to the N-terminal bromodomain (BD1) of BRD2 at the acetylated (Kac) histone binding site. The quantitative binding studies, by SPR and MST assay, indicate that NS5 binds to BD1 of BRD2 with a KD value of ∼1.3 µM. The cell-based assay, in the U87MG glioma cells, confirmed that the discovered compound NS5 significantly attenuated proliferation and migration. Furthermore, evaluation at the translational level established significant inhibition of BRD2 upon treatment with NS5. Hence, we propose that the novel lead compound NS5 has an inhibitory effect on BRD2 in glioblastoma.
Collapse
|
27
|
Lee JE, Yoon SS, Lee JW, Moon EY. Curcumin-induced cell death depends on the level of autophagic flux in A172 and U87MG human glioblastoma cells. Chin J Nat Med 2020; 18:114-122. [PMID: 32172947 DOI: 10.1016/s1875-5364(20)30012-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Indexed: 12/14/2022]
Abstract
Glioblastoma is the deadliest neoplasm with the worst 5-year survival rate among all human cancers. Autophagy promotes autophagic cell death or blocks the induction of apoptosis in eukaryotic cells. Here, we investigated whether varying levels of autophagic flux in glioblastoma lead to different efficacies of curcumin treatment using U87MG and A172 human glioblastoma cells. The number of LC3 puncta, the number of cells with LC3 puncta and the level of LC3 II, Atg5 and Atg7 protein were higher in U87MG cells compared with A172 cells. When the cells were incubated with curcumin for 24 or 48 h, the percentage of cell death was higher in A172 cells compared with U87MG cells. Although the level of LC3 was lower, that of curcumin-induced LC3 was higher, in A172 cells than in U87MG cells. The relative increases in cell death and LC3-mediated autophagy were greater under serum starvation in A172 cells compared with U87MG cells. Curcumin-induced A172 cell death was reduced by serum starvation. When both types of cells were transfected with LC3-GFP, the percentage of cell death was higher in A172 cells than that in U87MG cells. Taken together, the data demonstrate that curcumin-mediated tumor cell death is regulated by the basal level of autophagic flux in different glioblastoma cells. This suggests that prior to the use of various curcumin therapeutics, the level of basal or induced autophagic flux should be carefully examined in tumor cells for the best efficacy.
Collapse
Affiliation(s)
- Jong-Eun Lee
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Sung Sik Yoon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Jae-Wook Lee
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Eun-Yi Moon
- Department of Bioscience and Biotechnology, Sejong University, Seoul 05006, Republic of Korea.
| |
Collapse
|
28
|
Fractalkine/CX3CL1 in Neoplastic Processes. Int J Mol Sci 2020; 21:ijms21103723. [PMID: 32466280 PMCID: PMC7279446 DOI: 10.3390/ijms21103723] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 02/06/2023] Open
Abstract
Fractalkine/CX3C chemokine ligand 1 (CX3CL1) is a chemokine involved in the anticancer function of lymphocytes-mainly NK cells, T cells and dendritic cells. Its increased levels in tumors improve the prognosis for cancer patients, although it is also associated with a poorer prognosis in some types of cancers, such as pancreatic ductal adenocarcinoma. This work focuses on the 'hallmarks of cancer' involving CX3CL1 and its receptor CX3CR1. First, we describe signal transduction from CX3CR1 and the role of epidermal growth factor receptor (EGFR) in this process. Next, we present the role of CX3CL1 in the context of cancer, with the focus on angiogenesis, apoptosis resistance and migration and invasion of cancer cells. In particular, we discuss perineural invasion, spinal metastasis and bone metastasis of cancers such as breast cancer, pancreatic cancer and prostate cancer. We extensively discuss the importance of CX3CL1 in the interaction with different cells in the tumor niche: tumor-associated macrophages (TAM), myeloid-derived suppressor cells (MDSC) and microglia. We present the role of CX3CL1 in the development of active human cytomegalovirus (HCMV) infection in glioblastoma multiforme (GBM) brain tumors. Finally, we discuss the possible use of CX3CL1 in immunotherapy.
Collapse
|
29
|
Hübner M, Moellhoff N, Effinger D, Hinske CL, Hirschberger S, Wu T, Müller MB, Strauß G, Kreth FW, Kreth S. MicroRNA-93 acts as an "anti-inflammatory tumor suppressor" in glioblastoma. Neurooncol Adv 2020; 2:vdaa047. [PMID: 32642700 PMCID: PMC7282490 DOI: 10.1093/noajnl/vdaa047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Inflammation is an important driver of malignant glioma disease. Inflammatory mediators are not only produced by immune cells in the tumor microenvironment, but also by glioblastoma (GBM) cells themselves creating a mutually reinforcing loop. We here aimed at identifying an “anti-inflammatory switch” that allows to dampen inflammation in GBM. Methods We used human GBM specimens, primary cultures, and cell lines. The response of GBM cells toward inflammatory stimuli was tested by incubation with supernatant of stimulated human immune cells. Expression levels were measured by whole transcriptome microarrays and qRT-PCR, and protein was quantified by LUMINEX and SDS-PAGE. MicroRNA binding to 3′UTRs was analyzed by luciferase assays. Proliferation rates were determined by flow cytometry, and invasion and angiogenesis were studied using migration and endothelial tube formation assays. Results We demonstrated GBM cells to secrete high amounts of proinflammatory mediators in an inflammatory microenvironment. We found miR-93 as a potential “anti-inflammatory tumor suppressor” dramatically downregulated in GBM. Concordantly, cytokine secretion dropped after miR-93 re-expression. Transfection of miR-93 in GBM cells led to down-regulation of hubs of the inflammatory networks, namely, HIF-1α and MAP3K2 as well as IL-6, G-CSF, IL-8, LIF, IL-1β, COX2, and CXCL5. We showed only COX2 and CXCL5 to be indirectly regulated by miR-93 while all other genes are true targets. Phenotypically, re-expression of miR-93 in GBM cells substantially suppressed proliferation, migration, and angiogenesis. Conclusions Alleviating GBM-derived inflammation by re-expression of miR-93 may be a powerful tool to mitigate these tumors’ aggressiveness and holds promise for new clinical approaches.
Collapse
Affiliation(s)
- Max Hübner
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Nicholas Moellhoff
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - David Effinger
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Simon Hirschberger
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Tingting Wu
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Martin Bernhard Müller
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Gabriele Strauß
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Simone Kreth
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
30
|
Hübner M, Effinger D, Wu T, Strauß G, Pogoda K, Kreth FW, Kreth S. The IL-1 Antagonist Anakinra Attenuates Glioblastoma Aggressiveness by Dampening Tumor-Associated Inflammation. Cancers (Basel) 2020; 12:E433. [PMID: 32069807 PMCID: PMC7072290 DOI: 10.3390/cancers12020433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The recombinant IL-1 receptor antagonist anakinra-currently approved for the treatment of autoinflammatory diseases-blocks IL-1β-mediated inflammatory signaling. As inflammation is a major driver of cancer, we hypothesized that anakinra might be able to mitigate glioblastoma (GBM) aggressiveness. METHODS Primary GBM or T98G cells were incubated alone or with peripheral blood mononuclear cells (PBMCs) and were subsequently treated with IL-1β and/or anakinra. T cells were obtained by magnetic bead isolation. Protein and mRNA expression were quantified by SDS-PAGE, qRT-PCR, and ELISA, respectively. Cell proliferation and apoptosis were analyzed via flow cytometry. Chemotaxis was studied via time-lapse microscopy. RESULTS Upon IL-1β stimulation, anakinra attenuated proinflammatory gene expression in both GBM cells and PBMCs, and mitigated tumor migration and proliferation. In a more lifelike model replacing IL-1β stimulation by GBM-PBMC co-culture, sole presence of PBMCs proved sufficient to induce a proinflammatory phenotype in GBM cells with enhanced proliferation and migration rates and attenuated apoptosis. Anakinra antagonized these pro-tumorigenic effects and, moreover, reduced inflammatory signaling in T cells without compromising anti-tumor effector molecules. CONCLUSION By dampening the inflammatory crosstalk between GBM and immune cells, anakinra mitigated GBM aggressiveness. Hence, counteracting IL-1β-mediated inflammation might be a promising strategy to pursue.
Collapse
Affiliation(s)
- Max Hübner
- Department of Anesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.H.); (D.E.); (T.W.); (G.S.)
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany;
| | - David Effinger
- Department of Anesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.H.); (D.E.); (T.W.); (G.S.)
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany;
| | - Tingting Wu
- Department of Anesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.H.); (D.E.); (T.W.); (G.S.)
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany;
| | - Gabriele Strauß
- Department of Anesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.H.); (D.E.); (T.W.); (G.S.)
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany;
| | - Kristin Pogoda
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany;
- Biomedical Center, Ludwig-Maximilians-University, 82152 Planegg, Germany
| | | | - Simone Kreth
- Department of Anesthesiology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.H.); (D.E.); (T.W.); (G.S.)
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, 81377 Munich, Germany;
| |
Collapse
|
31
|
Ramaswamy P, Dalavaikodihalli Nanjaiah N, Prasad C, Goswami K. Transcriptional modulation of calcium-permeable AMPA receptor subunits in glioblastoma by MEK-ERK1/2 inhibitors and their role in invasion. Cell Biol Int 2019; 44:830-837. [PMID: 31814223 DOI: 10.1002/cbin.11279] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 12/05/2019] [Indexed: 11/09/2022]
Abstract
Glioblastoma is the most common primary brain tumor. Glioblastoma cells secrete a significant amount of glutamate, which serve as a potential growth factor in glioma pathobiology through their specific receptor subtypes including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR). Glioblastoma express AMPAR subunits; however, its regulation and activation with downstream intracellular signaling are not well-understood. Phosphorylated-extracellular signaling-regulated kinase (ERK)1/2 is known to regulate the ionotropic glutamate receptors in cortical neurons. The mitogen-activated protein kinase cascade is frequently activated in several tumors, including glioma. Nonetheless, the association of ERK signaling with AMPAR subunits in glioblastoma is undetermined. Here, we demonstrated potential role of AMPAR in invasion, and the modulation of AMPAR subunits at transcript level by ERK signaling in glioblastoma cells. Inhibition of ERK signaling specifically downregulated the expression of calcium-permeable AMPAR subunits, GluA1 and GluA4, and upregulated calcium-impermeable AMPAR subunit GluA2 implying differential regulation of the expression of calcium-permeable AMPAR subunits of glioblastoma. Concomitantly, it significantly decreased the invasion of U87MG cells. Taken together, these findings suggest that the AMPAR enhances invasion of glioblastoma, and ERK signaling modulates the differential expression of calcium-permeable AMPAR phenotype that might play a crucial role in the invasive propensity of glioblastoma cells.
Collapse
Affiliation(s)
- Palaniswamy Ramaswamy
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, 560029, India
| | | | - Chandrajit Prasad
- Department of Neuroimaging and Intervention Radiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, 560029, India
| | - Kalyan Goswami
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raipur, 492099, India
| |
Collapse
|
32
|
Iguratimod encapsulated PLGA-NPs improves therapeutic outcome in glioma, glioma stem-like cells and temozolomide resistant glioma cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 22:102101. [PMID: 31654739 DOI: 10.1016/j.nano.2019.102101] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 09/17/2019] [Accepted: 09/25/2019] [Indexed: 01/27/2023]
Abstract
Glioma is the most common neoplasm of the central nervous system, with the highest mortality rate. The present study was designed to examine the therapeutic effect of Iguratimod (IGU) encapsulated-poly (lactic-co-glycolic acid) PLGA nanoparticles (IGU-PLGA-NPs), which showed inhibition of glioma cells proliferation both in vitro and in vivo. IGU encapsulated in PLGA nanoparticles with an average size of 100-200 nm was prepared using modified double-emulsion (W1/O/W2) method. Cell Counting Kit-8 (CCK-8) analysis of Glioma cancer cells and glioma stem-like cells (GSCs) demonstrated significant inhibition of their growth treated with IGU-PLGA-NPs. IGU-PLGA-NPs inhibit migration in glioma cells as well as tumor sphere formation in GSCs. Treatment with IGU-PLGA-NPs showed a significant decrease in tumor growth through the apoptotic pathway in mice model without any visible organ toxicity and it can successfully cross the blood brain barrier (BBB). Most Importantly, IGU-PLGA-NPs significantly depleted growth of U251 Temozolomide-resistant (U251TMZ-R) cells.
Collapse
|
33
|
Nandakumar DN, Ramaswamy P, Prasad C, Srinivas D, Goswami K. Glioblastoma invasion and NMDA receptors: A novel prospect. Physiol Int 2019; 106:250-260. [PMID: 31564120 DOI: 10.1556/2060.106.2019.22] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE Glioblastoma cells create glutamate-rich tumor microenvironment, which initiates activation of ion channels and modulates downstream intracellular signaling. N-methyl-D-aspartate receptors (NMDARs; a type of glutamate receptors) have a high affinity for glutamate. The role of NMDAR activation on invasion of glioblastoma cells and the crosstalk with α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs) is yet to be explored. MAIN METHODS LN18, U251MG, and patient-derived glioblastoma cells were stimulated with NMDA to activate NMDAR glutamate receptors. The role of NMDAR activation on invasion and migration and its crosstalk with AMPAR were evaluated. Invasion and migration of glioblastoma cells were investigated by in vitro trans-well Matrigel invasion and trans-well migration assays, respectively. Expression of NMDARs and AMPARs at transcript level was evaluated by quantitative real-time polymerase chain reaction. RESULTS We determined that NMDA stimulation leads to enhanced invasion in LN18, U251MG, and patient-derived glioblastoma cells, whereas inhibition of NMDAR using MK-801, a non-competitive antagonist of the NMDAR, significantly decreased the invasive capacity. Concordant with these findings, migration was significantly augmented by NMDAR in both cell lines. Furthermore, NMDA stimulation upregulated the expression of GluN2 and GluA1 subunits at the transcript level. CONCLUSIONS This study demonstrated the previously unexplored role of NMDAR in invasion of glioblastoma cells. Furthermore, the expression of the GluN2 subunit of NMDAR and the differential overexpression of the GluA1 subunit of AMPAR in both cell lines provide a plausible rationale of crosstalk between these calcium-permeable subunits in the glutamate-rich microenvironment of glioblastoma.
Collapse
Affiliation(s)
- D N Nandakumar
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - P Ramaswamy
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - C Prasad
- Department of Neuroimaging and Intervention Radiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - D Srinivas
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, India
| | - K Goswami
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raipur, India
| |
Collapse
|
34
|
Mitchell D, Chintala S, Fetcko K, Henriquez M, Tewari BN, Ahmed A, Bentley RT, Dey M. Common Molecular Alterations in Canine Oligodendroglioma and Human Malignant Gliomas and Potential Novel Therapeutic Targets. Front Oncol 2019; 9:780. [PMID: 31475119 PMCID: PMC6702544 DOI: 10.3389/fonc.2019.00780] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/31/2019] [Indexed: 01/05/2023] Open
Abstract
Spontaneous canine (Canis lupus) oligodendroglioma (ODG) holds tremendous potential as an immunocompetent large animal model of human malignant gliomas (MG). However, the feasibility of utilizing this model in pre-clinical studies depends on a thorough understanding of the similarities and differences of the molecular pathways associated with gliomas between the two species. We have previously shown that canine ODG has an immune landscape and expression pattern of commonly described oncogenes similar to that of human MG. In the current study, we performed a comprehensive analysis of canine ODG RNAseq data from 4 dogs with ODG and 2 normal controls to identify highly dysregulated genes in canine tumors. We then evaluated the expression of these genes in human MG using Xena Browser, a publicly available database. STRING-database inquiry was used in order to determine the suggested protein associations of these differentially expressed genes as well as the dysregulated pathways commonly enriched by the protein products of these genes in both canine ODG and human MG. Our results revealed that 3,712 (23%) of the 15,895 differentially expressed genes demonstrated significant up- or downregulation (log2-fold change > 2.0). Of the 3,712 altered genes, ~50% were upregulated (n = 1858) and ~50% were downregulated (n = 1854). Most of these genes were also found to have altered expression in human MG. Protein association and pathway analysis revealed common pathways enriched by members of the up- and downregulated gene categories in both species. In summary, we demonstrate that a similar pattern of gene dysregulation characterizes both human MG and canine ODG and provide additional support for the use of the canine model in order to therapeutically target these common genes. The results of such therapeutic targeting in the canine model can serve to more accurately predict the efficacy of anti-glioma therapies in human patients.
Collapse
Affiliation(s)
- Dana Mitchell
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Sreenivasulu Chintala
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Kaleigh Fetcko
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Mario Henriquez
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Brij N Tewari
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Atique Ahmed
- Department of Neurological Surgery, Northwestern University, Chicago, IL, United States
| | - R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN, United States
| | - Mahua Dey
- Department of Neurosurgery, Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
35
|
Ramaswamy P, Goswami K, Dalavaikodihalli Nanjaiah N, Srinivas D, Prasad C. TNF-α mediated MEK-ERK signaling in invasion with putative network involving NF-κB and STAT-6: a new perspective in glioma. Cell Biol Int 2019; 43:1257-1266. [PMID: 30839135 DOI: 10.1002/cbin.11125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 03/02/2019] [Indexed: 12/22/2022]
Abstract
Glioblastoma is the most common malignant primary brain tumor with poor prognosis. Invasion involves pro-inflammatory cytokines and major signaling hubs. Tumor necrosis factor-α (TNF-α) acts as a master switch in establishing an intricate link between inflammation and cancer. The present study attempted to explore the possible implication of MAPK extracellular signaling-regulated kinase kinase (MEK)-extracellular signaling-regulated kinase (ERK) signaling pathway and expression of nuclear factor-κB (NF-κB), signal transducers and activators of transcription-6 (STAT-6), ERK, and phosphorylated-ERK (p-ERK) signaling proteins in TNF-α microenvironment. U0126 and PD98059 were used to inhibit the MEK-ERK1/2 pathway. TNF-α stimulation enhanced invasion in U87MG, U251MG and patient-derived primary glioma cells, whereas cell viability was not altered. Matrix metalloproteinase-2 (MMP-2) activity was increased only in U251MG glioma cells. These data suggest that TNF-α microenvironment plays an important role in the invasion of U251MG, U87MG, and patient-derived primary glioma cells, without any cytotoxic effect. The MMP-2 activity is differentially regulated by TNF-α stimulation in these cells. TNF-α stimulation upregulated the protein expression of ERK-1, ERK-2 and also increased the level of p-ERK1/2. TNF-α stimulation further upregulated the expression of NF-κB1, STAT-6 in tandem with Ras-MEK signaling system in U87MG cells, which emphasized the possible involvement of these signaling hubs in the glioma microenvironment. MEK-ERK inhibitors significantly attenuated the invasion of U87MG cells mediated by the TNF-α stimulation, probably through their inhibitory impact on p-ERK1/2 and ERK-2. This study provides the possible rationale of invasion by glioma cells in a TNF-α-induced pro-inflammatory milieu, which involves direct role of MEK-ERK signaling, with possible implication of NF-κB and STAT-6.
Collapse
Affiliation(s)
- Palaniswamy Ramaswamy
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, 560029, India
| | - Kalyan Goswami
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raipur, 492099, India
| | | | - Dwarakanath Srinivas
- Department of Neurosurgery, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, 560029, India
| | - Chandrajit Prasad
- Department of Neuroimaging and Intervention Radiology, National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru, 560029, India
| |
Collapse
|
36
|
Gao Y, Zhang E, Liu B, Zhou K, He S, Feng L, Wu G, Cao M, Wu H, Cui Y, Zhang X, Liu X, Wang Y, Gao Y, Bian X. Integrated analysis identified core signal pathways and hypoxic characteristics of human glioblastoma. J Cell Mol Med 2019; 23:6228-6237. [PMID: 31282108 PMCID: PMC6714287 DOI: 10.1111/jcmm.14507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 04/30/2019] [Accepted: 05/30/2019] [Indexed: 12/26/2022] Open
Abstract
As a hallmark for glioblastoma (GBM), high heterogeneity causes a variety of phenotypes and therapeutic responses among GBM patients, and it contributes to treatment failure. Moreover, hypoxia is a predominant feature of GBM and contributes greatly to its phenotype. To analyse the landscape of gene expression and hypoxic characteristics of GBM cells and their clinical significance in GBM patients, we performed transcriptome analysis of the GBM cell line U87‐MG and the normal glial cell line HEB under normoxia and hypoxia conditions, with the results of which were analysed using established gene ontology databases as well as The Cancer Genome Atlas and the Cancer Cell Line Encyclopedia. We revealed core signal pathways, including inflammation, angiogenesis and migration, and for the first time mapped the components of the toll‐like receptor 6 pathway in GBM cells. Moreover, by investigating the signal pathways involved in homoeostasis, proliferation and adenosine triphosphate metabolism, the critical response of GBM to hypoxia was clarified. Experiments with cell lines, patient serum and tissue identified IL1B, CSF3 and TIMP1 as potential plasma markers and VIM, STC1, TGFB1 and HMOX1 as potential biopsy markers for GBM. In conclusion, our study provided a comprehensive understanding for signal pathways and hypoxic characteristics of GBM and identified new biomarkers for GBM patients.
Collapse
Affiliation(s)
- Yixing Gao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Erlong Zhang
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, and Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Bao Liu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, and Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Kai Zhou
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Shu He
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, and Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Lan Feng
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, and Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Gang Wu
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, and Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Mianfu Cao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Haibo Wu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Youhong Cui
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Xindong Liu
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yan Wang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China
| | - Yuqi Gao
- Institute of Medicine and Equipment for High Altitude Region, College of High Altitude Military Medicine, Army Medical University, and Key Laboratory of Extreme Environmental Medicine, Ministry of Education of China, Chongqing, China
| | - Xiuwu Bian
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), and Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Chongqing, China.,Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
37
|
Gastrodin Inhibits Inflammasome Through the STAT3 Signal Pathways in TNA2 Astrocytes and Reactive Astrocytes in Experimentally Induced Cerebral Ischemia in Rats. Neuromolecular Med 2019; 21:275-286. [PMID: 31218587 DOI: 10.1007/s12017-019-08544-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/11/2019] [Indexed: 12/28/2022]
Abstract
This study was aimed to determine Gastrodin (GAS) and its underlying signaling pathway involved in suppression of inflammasome specifically in reactive astrocytes that are featured prominently in different neurological conditions or diseases including cerebral ischemia. For this purpose, TNA2 astrocytes in cultures were exposed to oxygen-glucose-deprivation (OGD) mimicking hypoxic cerebral ischemia. Separately, TNA2 cells were pretreated with GAS prior to OGD exposure. Additionally, Stattic, an inhibitor of STAT3 signaling pathway, was used to ascertain its involvement in regulating inflammasome in astrocytes exposed to OGD. In parallel to the above, adult rats subjected to middle cerebral artery occlusion (MCAO) with or without GAS pretreatment were sacrificed at different time points to determine the effects of GAS on astrocyte inflammasome. TNA2 astrocytes in different treatments as well as reactive astrocytes in MCAO were processed for immunofluorescence labeling and Western blot analysis for various protein markers. In the latter, protein expression levels of p-STAT3, NLRP3, and NLRC4 were markedly increased in TNA2 astrocytes exposed to OGD. Remarkably, the expression levels of these biomarkers were significantly suppressed by GAS. Of note, GAS especially at dose 20 μM inhibited NLRP3 and NLRC4 expression levels most substantially. Moreover, GAS inhibited the downstream proteins caspase-1 and IL-18. Concomitantly, GAS significantly suppressed the expression of STAT3 and NF-κB signaling pathway. It is noteworthy that Stattic at dose 100 μM inhibited STAT3 pathway and NF-κB activation in TNA2 astrocytes, an effect that was shared by GAS. In MCAO, GAS was found to effectively attenuate p-STAT3 immunofluorescence intensity in reactive astrocytes. Arising from the above, it is concluded that GAS is anti-inflammatory as it effectively suppresses inflammasome in OGD-stimulated astrocytes as well as in reactive astrocytes in MCAO via STAT3 and NF-κB signaling expression coupled with decreased expression of caspase-1 and IL-18.
Collapse
|
38
|
Tong L, Xie C, Wei Y, Qu Y, Liang H, Zhang Y, Xu T, Qian X, Qiu H, Deng H. Antitumor Effects of Berberine on Gliomas via Inactivation of Caspase-1-Mediated IL-1β and IL-18 Release. Front Oncol 2019; 9:364. [PMID: 31139563 PMCID: PMC6527738 DOI: 10.3389/fonc.2019.00364] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/18/2019] [Indexed: 01/03/2023] Open
Abstract
Gliomas arise in the glial cells of the brain or spine and are the most prevalent and devastating type of brain tumors. Studies of tumor immunology have established the importance of the tumor micro-environment as a driver of oncogenesis. Inflammatory mediators such as IL-1β and IL-18 released by monocytes regulate transcriptional networks that are required for malignant cell growth. Berberine is a natural botanical alkaloid that is widely found in the Berberis species. Although it has been widely used as an anti-diarrheal treatment in North America for several decades, our study is the first to investigate berberine as an anti-tumor agent in glioma cells. In this study, we demonstrate that berberine significantly inhibits inflammatory cytokine Caspase-1 activation via ERK1/2 signaling and subsequent production of IL-1β and IL-18 by glioma cells. Moreover, we found that berberine treatment led to decreased motility and subsequently cell death in U251 and U87 cells. In addition, our study is the first to indicate that berberine can reverse the process of epithelial-mesenchymal transition, a marker of tumor invasion. Taken together, our work supports berberine as a putative anti-tumor agent targeting glioma cells.
Collapse
Affiliation(s)
- Lei Tong
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Chuncheng Xie
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yafen Wei
- Department of Neurology, The Provincal Hospital of Heilongjiang Province, Harbin, China
| | - Yunyue Qu
- Department of Microbiology, Harbin Medical University, Harbin, China
| | - Hongsheng Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yiwei Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tianye Xu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Qian
- Department of Vascular Surgery, RenJi Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Huijia Qiu
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haoyu Deng
- Department of Vascular Surgery, RenJi Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
39
|
Lyu X, Song AL, Bai YL, Xu XD, He DQ, Zhang YC. Inhibitory effects of petasin on human colon carcinoma cells mediated by inactivation of Akt/mTOR pathway. Chin Med J (Engl) 2019; 132:1071-1078. [PMID: 30896562 PMCID: PMC6595872 DOI: 10.1097/cm9.0000000000000199] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Colorectal cancer is the third most common cancer worldwide and still lack of effective therapy so far. Petasin, a natural product found in plants of the genus Petasites, has been reported to possess anticancer activity. The present study aimed to investigate the anticolon cancer activity of petasin both in vitro and in vivo. The molecular mechanism of petasin was also further explored. METHODS Caco-2, LoVo, SW-620, and HT-29 cell lines were used to detect the inhibitory effect of petasin on colon cancer proliferation. Cell viability was determined using the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) assay. Cell apoptosis was analyzed by flow cytometry. Hoechst 33258 staining was used to visualize morphological changes. Cell migration was assessed using a wound-healing migration assay, and cell invasion was investigated using Transwell chambers. Western blotting assays were employed to evaluate the expression levels of proteins in the protein kinase B/mammalian target of rapamycin (Akt/mTOR) signaling pathway. Finally, in vivo activity of petasin was evaluated using the SW-620 subcutaneous tumor model established in Balb/c nude mice. Twelve rats were randomly divided into control group and 10 mg/kg petasin group. The tumor volume was calculated every 7 days for 28 days. Terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay was performed to assess the apoptotic effect of petasin. Differences between two groups were assessed by analysis of independent-sample t tests. RESULTS Petasin significantly inhibited the proliferation of human colon carcinoma cell lines, induced apoptosis, and suppressed migration and invasion in SW-620 cells. Western blotting results showed that petasin decreased the phosphorylation of Akt (1.01 ± 0.16 vs. 0.74 ± 0.06, P = 0.042), mTOR (0.71 ± 0.12 vs. 0.32 ± 0.11, P = 0.013), and P70S6K (1.23 ± 0.21 vs. 0.85 ± 0.14, P = 0.008), elevated the expression of caspase-3 (0.41 ± 0.09 vs. 0.74 ± 0.12, P = 0.018) and caspase-9 (1.10 ± 0.27 vs. 1.98 ± 0.22, P = 0.009), decreased the Bcl-2 protein (2.75 ± 0.47 vs. 1.51 ± 0.36, P = 0.008), downregulated the expression of matrix metalloproteinase (MMP)-3 (1.51 ± 0.31 vs. 0.82 ± 0.11, P = 0.021) and MMP-9 (1.56 ± 0.32 vs. 0.94 ± 0.15, P = 0.039) in SW-620 cell. In vivo, 10 mg/kg petasin inhibited tumor growth in Balb/c nude mice (924.18 ± 101.23 vs. 577.67 ± 75.12 mm at day 28, P = 0.001) and induced apoptosis (3.6 ± 0.7% vs. 36.0 ± 4.9%, P = 0.001) in tumor tissues. CONCLUSIONS Petasin inhibits the proliferation of colon cancer SW-620 cells via inactivating the Akt/mTOR pathway. Our findings suggest petasin as a potential candidate for colon cancer therapy.
Collapse
Affiliation(s)
- Xi Lyu
- The 5th Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Ai-Lin Song
- The 5th Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Yin-Liang Bai
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Xiao-Dong Xu
- The 2nd Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Dong-Qiang He
- The 5th Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - You-Cheng Zhang
- The 2nd Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| |
Collapse
|
40
|
Abstract
Inflammasomes are molecular platforms that assemble upon sensing various intracellular stimuli. Inflammasome assembly leads to activation of caspase 1, thereby promoting the secretion of bioactive interleukin-1β (IL-1β) and IL-18 and inducing an inflammatory cell death called pyroptosis. Effectors of the inflammasome efficiently drive an immune response, primarily providing protection against microbial infections and mediating control over sterile insults. However, aberrant inflammasome signalling is associated with pathogenesis of inflammatory and metabolic diseases, neurodegeneration and malignancies. Chronic inflammation perpetuated by inflammasome activation plays a central role in all stages of tumorigenesis, including immunosuppression, proliferation, angiogenesis and metastasis. Conversely, inflammasome signalling also contributes to tumour suppression by maintaining intestinal barrier integrity, which portrays the diverse roles of inflammasomes in tumorigenesis. Studies have underscored the importance of environmental factors, such as diet and gut microbiota, in inflammasome signalling, which in turn influences tumorigenesis. In this Review, we deliver an overview of the interplay between inflammasomes and tumorigenesis and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rajendra Karki
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | | |
Collapse
|
41
|
Role of MEK-ERK signaling mediated adhesion of glioma cells to extra-cellular matrix: Possible implication on migration and proliferation. Ann Neurosci 2019; 26:52-56. [PMID: 31975773 PMCID: PMC6894623 DOI: 10.5214/ans.0972.7531.260203] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/27/2019] [Accepted: 04/03/2019] [Indexed: 01/06/2023] Open
Abstract
Background Glioblastoma represents the most common primary brain tumor with a worst
prognosis despite developments in neurosurgery and chemoradiotherapy.
Detachment of the cells from the primary tumor tissue is a prerequisite for
their dispersion and spreading. Initial and incessant dispersal of tumor
cells from the primary tumor tissue renders GBM refractory to comprehensive
surgical removal and increases the chance of recurrence and poorer
prognosis. Purposes The current study was designed to investigate the effect of inhibition of
MEK-ERK1/2 signaling by PD98059 and U0126 on the growth and migration of
glioma cells as well as their adhesion to extracellular matrix. Methods MEK-ERK1/2 signaling in U87-MG cells was inhibited by PD98059 and U0126.
Migration, proliferation and adhesion were analyzed by scratch-wound assay,
MTT assay, cell adhesion assay respectively. Results PD98059 and U0126 significantly not only reduced the proliferation of glioma
cells and attenuated their migration but also increased their adhesion to
gelatin of extracellular matrix. Conclusion This study provides the evidence that inhibition of MEK-ERK1/2 signaling
enhances the adhesion of glioma cells to gelatin/collagen component of ECM,
and decreases the proliferation and migration of the glioma cells. We
propose the possible rationale of association between ERK signaling and
cell-cell adhesion molecules in glioma microenvironment which regulates the
glioma initiation, growth and progression.
Collapse
|
42
|
Wang B, Zhao CH, Sun G, Zhang ZW, Qian BM, Zhu YF, Cai MY, Pandey S, Zhao D, Wang YW, Qiu W, Shi L. IL-17 induces the proliferation and migration of glioma cells through the activation of PI3K/Akt1/NF-κB-p65. Cancer Lett 2019; 447:93-104. [PMID: 30660646 DOI: 10.1016/j.canlet.2019.01.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/08/2018] [Accepted: 01/07/2019] [Indexed: 01/08/2023]
Abstract
Interleukin 17 (IL-17), as a pro-inflammatory cytokine, is up-regulated in the sera and tumor tissues of glioma patients; however the effects of IL-17 on glioma proliferation and migration remain unclear. In this study, the roles of IL-17 in the proliferation and migration of glioma cells and their potential mechanisms were determined. The results showed that IL-17 could not only enhance the proliferation and migration of cultured glioma cells (in vitro), but also promote the tumor formation of glioma cells in BALB/c nude mice (in vivo). Mechanical exploration revealed that IL-17 stimulation could increase the phosphorylation levels of Akt1 and NF-κB-p65 in glioma cells, and knockdown or inhibition of PI3K, Akt1 and NF-κB-p65 could also reduce the IL-17-induced proliferation and migration of the glioma cells. Moreover, PI3K/Akt1 was the upstream regulator of NF-κB-p65 activation in IL-17-incubated glioma cells. Furthermore, the inhibition of PI3K, Akt1 and NF-κB-p65 markedly suppressed the tumor formation of glioma cells induced by IL-17. Together, these data indicate that IL-17 can promote the proliferation and migration of glioma cells via PI3K/Akt1/NF-κB-p65 activation, and these findings might provide a new insight into glioma pathogenesis.
Collapse
Affiliation(s)
- Bin Wang
- Department of Neurosurgery, The First People's Hospital of Kunshan affiliated with Jiangsu University, Suzhou, Jiangsu, 215300, PR China
| | - Chen-Hui Zhao
- Department of Oncology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, PR China
| | - Guan Sun
- Department of Neurosurgery, The Fourth Affiliated Hospital of Nantong University, Yancheng City No.1 people's hospital, Yancheng, Jiangsu, 224000, PR China
| | - Zhi-Wei Zhang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Bao-Mei Qian
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Yu-Feng Zhu
- Clinical Medical Science of the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Meng-Yuan Cai
- Clinical Medical Science of the First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Samjhana Pandey
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Dan Zhao
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Ying-Wei Wang
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China
| | - Wen Qiu
- Department of Immunology, Nanjing Medical University, Nanjing, Jiangsu, 211166, PR China.
| | - Lei Shi
- Department of Neurosurgery, The First People's Hospital of Kunshan affiliated with Jiangsu University, Suzhou, Jiangsu, 215300, PR China.
| |
Collapse
|
43
|
Litmanovich A, Khazim K, Cohen I. The Role of Interleukin-1 in the Pathogenesis of Cancer and its Potential as a Therapeutic Target in Clinical Practice. Oncol Ther 2018; 6:109-127. [PMID: 32700032 PMCID: PMC7359982 DOI: 10.1007/s40487-018-0089-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Indexed: 02/07/2023] Open
Abstract
Interleukin-1 (IL-1) has long been known to be a key mediator of immunity and inflammation. Its dysregulation has been implicated in recent years in tumorigenesis and tumor progression, and its upregulation is thought to be associated with many tumors. Overexpression of the IL-1 agonists IL-1α and IL-1β has been shown to promote tumor invasiveness and metastasis by inducing the expression of angiogenic genes and growth factors. IL-1 blockers such as anakinra and canakinumab are already approved and widely used for the treatment of some autoimmune and autoinflammatory diseases and are currently being tested in preclinical and human clinical trials for cancer therapy. In this paper we review the most recent discoveries regarding the association between IL-1 dysregulation and cancer and present the novel IL-1 blockers currently being tested in cancer therapy and their corresponding clinical trials.
Collapse
Affiliation(s)
- Adi Litmanovich
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
| | - Khaled Khazim
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- Research Institute, Galilee Medical Center, Nahariya, Israel
- Department of Nephrology and Hypertension, Galilee Medical Center, Nahariya, Israel
| | - Idan Cohen
- The Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel.
- Research Institute, Galilee Medical Center, Nahariya, Israel.
| |
Collapse
|
44
|
Shang S, Wang L, Zhang Y, Lu H, Lu X. The Beta-Hydroxybutyrate Suppresses the Migration of Glioma Cells by Inhibition of NLRP3 Inflammasome. Cell Mol Neurobiol 2018; 38:1479-1489. [PMID: 30218403 PMCID: PMC11469911 DOI: 10.1007/s10571-018-0617-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 08/30/2018] [Indexed: 12/19/2022]
Abstract
Activation of inflammasome leads to the formation of an inflammatory microenvironment which plays an important role in the process of cancer development. Beta-hydroxybutyrate (BHB) is a ketone body that has recently been reported to exert anti-inflammatory effects via inhibition of NOD-like receptor pyrin domain-containing 3 (NLRP3) inflammasome. Here, we investigated the potential influence of BHB on the in vitro migration of C6 glioma cells and the activation of NLRP3 inflammasome. Our results indicated that administration of BHB suppressed C6 cells migration and NLRP3 inflammasome activation, reducing the levels of activated cysteinyl aspartate-specific proteinase 1 (caspase-1) and mature Interleukin 1β (IL-1β). Fully activation of NLRP3 inflammasome was induced by lipopolysaccharide (LPS) prime plus adenosine triphosphate (ATP) stimulation in C6 cells, which promoted in vitro migration of C6 cell. BHB also counteracted the LPS/ATP-promoted cell migration by suppressing the activation of caspase-1 and the maturation of IL-1β. The enhancement of phospho-signal transducer and activator of transcription 3 (p-STAT3), degradation of nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) as well as the overexpression of fibroblast growth factor 2 (FGF2) resulting from LPS/ATP treatment, and subsequent IL-1β maturation could also be compensated by BHB. Our results suggested that BHB inhibits the activation of NLRP3 inflammasome in C6 glioma cells and consequently suppressed the C6 cell migration. These findings also implicated that by inhibiting NLRP3 inflammasome, BHB reduced the inflammatory microenvironment which provided ancillary therapeutic benefits for the intervention of glioma.
Collapse
Affiliation(s)
- Sen Shang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, People's Republic of China
| | - Leilei Wang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, People's Republic of China
| | - Yali Zhang
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, People's Republic of China
| | - Haixia Lu
- Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Institute of Neurobiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Centre, Xi'an, 710061, Shaanxi, People's Republic of China.
| | - Xiaoyun Lu
- Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, People's Republic of China.
| |
Collapse
|
45
|
Ma M, Pei Y, Wang X, Feng J, Zhang Y, Gao MQ. LncRNA XIST mediates bovine mammary epithelial cell inflammatory response via NF-κB/NLRP3 inflammasome pathway. Cell Prolif 2018; 52:e12525. [PMID: 30362186 PMCID: PMC6430464 DOI: 10.1111/cpr.12525] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 07/16/2018] [Accepted: 08/10/2018] [Indexed: 12/26/2022] Open
Abstract
Objectives The correlations between long non‐coding RNAs (lncRNAs) and diverse mammal diseases have been clarified by many researches, but the cognition about bovine mastitis‐related lncRNAs remains limited. This study aimed to investigate the potential role of lncRNA X‐inactive specific transcript (XIST) in the inflammatory response of bovine mammary epithelial cells. Materials and methods Two inflammatory bovine mammary alveolar cell‐T (MAC‐T) models were established by infecting the cells with Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus). The expressions of pro‐inflammatory cytokines were measured, and the proliferation, viability and apoptosis of the inflammatory cells were evaluated after XIST was knocked down by an siRNA. The relationship among XIST, NF‐κB pathway and NOD‐like receptor protein 3 (NLRP3) inflammasome was investigated using an inhibitor of NF‐κB signal pathway. Results The expression of XIST was abnormally increased in bovine mastitic tissues and inflammatory MAC‐T cells. Silencing of XIST significantly increased the expression of E. coli or S. aureus‐induced pro‐inflammatory cytokines. Additionally, knockdown of XIST could inhibit cell proliferation, suppress cell viability and promote cell apoptosis under inflammatory conditions. Furthermore, XIST inhibited E. coli or S. aureus‐induced NF‐κB phosphorylation and the production of NLRP3 inflammasome. Conclusions The expression of XIST was promoted by activated NF‐κB pathway and, in turn, XIST generated a negative feedback loop to regulate NF‐κB/NLRP3 inflammasome pathway for mediating the process of inflammation.
Collapse
Affiliation(s)
- Mengru Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yifei Pei
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xixi Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jiaxin Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Ming-Qing Gao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| |
Collapse
|
46
|
Yin XF, Zhang Q, Chen ZY, Wang HF, Li X, Wang HX, Li HX, Kang CM, Chu S, Li KF, Li Y, Qiu YR. NLRP3 in human glioma is correlated with increased WHO grade, and regulates cellular proliferation, apoptosis and metastasis via epithelial-mesenchymal transition and the PTEN/AKT signaling pathway. Int J Oncol 2018; 53:973-986. [PMID: 30015880 PMCID: PMC6065456 DOI: 10.3892/ijo.2018.4480] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 06/08/2018] [Indexed: 02/07/2023] Open
Abstract
Glioma is the most prevalent and fatal primary tumor of the central nervous system in adults, while the development of effective therapeutic strategies in clinical practice remain a challenge. Nucleotide-binding domain leucine-rich family pyrin-containing 3 (NLRP3) has been reported to be associated with tumorigenesis and progression; however, its expression and function in human glioma remain unclear. The present study was designed to explore the biological role and potential mechanism of NLRP3 in human glioma. The results demonstrated that overexpression of NLRP3, apoptosis-associated speck-like protein containing a caspase-recruitment domain (ASC), caspase-1 and interleukin (IL)-1β protein in human glioma tissues were significantly correlated with higher World Health Organization grades. The in vitro biological experiments demonstrated that NLRP3 downregulation significantly inhibited the proliferation, migration and invasion, and promoted the apoptosis of SHG44 and A172 glioma cell lines. Furthermore, western blot assays revealed that the downregulation of NLRP3 significantly reduced the expression of ASC, caspase-1 and IL-1β protein. Furthermore, NLRP3 knockdown caused the inhibition of epithelial-mesenchymal transition (EMT), and inhibited the phosphorylation of AKT serine/threonine kinase (AKT) and phosphorylation of phosphatase and tensin homolog (PTEN). Consistently, the upregulation of NLRP3 significantly increased the expression of ASC, caspase-1, IL-1β and phosphorylated-PTEN, promoted proliferation, migration, invasion and EMT, inhibited apoptosis, and activated the AKT signaling pathway. The data of the present study indicate that NLRP3 affects human glioma progression and metastasis through multiple pathways, including EMT and PTEN/AKT signaling pathway regulation, enhanced inflammasome activation, and undefined inflammasome-independent mechanisms. Understanding the biological effects of NLRP3 in human glioma and the underlying mechanisms may offer novel insights for the development of glioma clinical therapeutic strategies.
Collapse
Affiliation(s)
- Xiao-Feng Yin
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qiong Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Zhuo-Yu Chen
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hai-Fang Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xin Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hong-Xia Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Hai-Xia Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Chun-Min Kang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shuai Chu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Kai-Fei Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yao Li
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yu-Rong Qiu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
47
|
Xue L, Lu B, Gao B, Shi Y, Xu J, Yang R, Xu B, Ding P. NLRP3 Promotes Glioma Cell Proliferation and Invasion via the Interleukin-1β/NF-κB p65 Signals. Oncol Res 2018; 27:557-564. [PMID: 29769161 PMCID: PMC7848456 DOI: 10.3727/096504018x15264647024196] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Because of the characteristics of high invasiveness, relapse, and poor prognosis, the management of malignant gliomas has always been a great challenge. Nod-like receptor (NLR) family pyrin domain containing 3 (NLRP3) is a crucial component of the NLRP3 inflammasome, a multiprotein complex that can trigger caspase 1/interleukin-1 (IL-1)-mediated inflammatory response once activated and participates in the pathogeny of diverse inflammatory diseases as well as cancers. We examined the function of NLRP3 in the development of glioma. Glioma cells were treated with NLRP3 interference or overexpression vectors, recombinant IL-1β, IL-1β antibody, and NF-κB inhibitor. Cell proliferation and invasion were assessed by CCK-8 and Transwell assays. Gene expression was detected by PCR, Western blot, and ELISA. NLRP3 and NF-κB p65 increased and were positively correlated in glioma tissues. NLRP3 knockdown suppressed glioma cell growth and invasion with the decrease of IL-1β and NF-κB p65. Conversely, forced expression of NLRP3 promoted cell growth. NLRP3 silencing suppressed ectogenous IL-1β-elevated cell proliferation and invasion, whereas IL-1β elimination impaired the proproliferation effect of NLRP3 hyperexpression. Furthermore, NF-κB blockage abrogated IL-1β and NLRP3 hyperexpression increased cell growth and invasion. NLRP3 promoted the growth and invasion of gliomas via the IL-1β/NF-κB p65 signals.
Collapse
Affiliation(s)
- Liping Xue
- Department of Ophthalmology, Yunnan No. 2 Provincial People's Hospital, Kunming, Yunnan, P.R. China
| | - Bin Lu
- Department of Neurosurgery, Huzhou Central Hospital, Huzhou, Zhejiang, P.R. China
| | - Bibo Gao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Yangyang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Jingqi Xu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Rui Yang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Bo Xu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| | - Peng Ding
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, P.R. China
| |
Collapse
|
48
|
Shiraki Y, Mii S, Enomoto A, Momota H, Han YP, Kato T, Ushida K, Kato A, Asai N, Murakumo Y, Aoki K, Suzuki H, Ohka F, Wakabayashi T, Todo T, Ogawa S, Natsume A, Takahashi M. Significance of perivascular tumour cells defined by CD109 expression in progression of glioma. J Pathol 2017; 243:468-480. [DOI: 10.1002/path.4981] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/29/2017] [Accepted: 09/05/2017] [Indexed: 12/11/2022]
Affiliation(s)
- Yukihiro Shiraki
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Shinji Mii
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Atsushi Enomoto
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Hiroyuki Momota
- Division of Innovative Cancer Therapy, The Institute of Medical Science; The University of Tokyo; Tokyo Japan
| | - Yi-Peng Han
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Takuya Kato
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Kaori Ushida
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Akira Kato
- Department of Neurosurgery; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Naoya Asai
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Yoshiki Murakumo
- Department of Pathology; Kitasato University School of Medicine; Sagamihara Japan
| | - Kosuke Aoki
- Department of Neurosurgery; Nagoya University Graduate School of Medicine; Nagoya Japan
- Department of Pathology and Tumor Biology, Graduate School of Medicine; Kyoto University; Kyoto Japan
| | - Hiromichi Suzuki
- Department of Neurosurgery; Nagoya University Graduate School of Medicine; Nagoya Japan
- Department of Pathology and Tumor Biology, Graduate School of Medicine; Kyoto University; Kyoto Japan
| | - Fumiharu Ohka
- Department of Neurosurgery; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Toshihiko Wakabayashi
- Department of Neurosurgery; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, The Institute of Medical Science; The University of Tokyo; Tokyo Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine; Kyoto University; Kyoto Japan
| | - Atsushi Natsume
- Department of Neurosurgery; Nagoya University Graduate School of Medicine; Nagoya Japan
| | - Masahide Takahashi
- Department of Pathology; Nagoya University Graduate School of Medicine; Nagoya Japan
| |
Collapse
|
49
|
Sangpairoj K, Vivithanaporn P, Apisawetakan S, Chongthammakun S, Sobhon P, Chaithirayanon K. RUNX1 Regulates Migration, Invasion, and Angiogenesis via p38 MAPK Pathway in Human Glioblastoma. Cell Mol Neurobiol 2017; 37:1243-1255. [PMID: 28012022 PMCID: PMC11482080 DOI: 10.1007/s10571-016-0456-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022]
Abstract
Runt-related transcription factor 1 (RUNX1) is essential for the establishment of fetal and adult hematopoiesis and neuronal development. Aberrant expression of RUNX1 led to proliferation and metastasis of several cancers. The aim of the present study was to investigate the role of RUNX1 in migration, invasion, and angiogenesis of human glioblastoma using IL-1β-treated U-87 MG human glioblastoma cells as a model. IL-1β at 10 ng/ml stimulated translocation of RUNX1 into the nucleus with increased expressions of RUNX1, MMP-1, MMP-2, MMP-9, MMP-19, and VEGFA in U-87 MG cells. In addition, silencing of RUNX1 gene significantly suppressed U-87 MG cell migration and invasion abilities. Moreover, knockdown of RUNX1 mRNA in U-87 MG cells reduced the tube formation of human umbilical vein endothelial cells. Further investigation revealed that IL-1β-induced RUNX1 expression might be mediated via the p38 mitogen-activated protein kinase (MAPK) signaling molecule for the expression of these invasion- and angiogenic-related molecules. Together with an inhibitor of p38 MAPK (SB203580) could decrease RUNX1 mRNA expression. Thus, RUNX1 may be one of the putative molecular targeted therapies against glioma metastasis and angiogenesis through the activation of p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Kant Sangpairoj
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Pornpun Vivithanaporn
- Department of Pharmacology, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Somjai Apisawetakan
- Department of Anatomy, Faculty of Medicine, Srinakharinwirot University, Wattana, Bangkok, 10110, Thailand
| | - Sukumal Chongthammakun
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
| | - Prasert Sobhon
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand
- Faculty of Allied Health Sciences, Burapha University, Mueang District, Chonburi, 20131, Thailand
| | - Kulathida Chaithirayanon
- Department of Anatomy, Faculty of Science, Mahidol University, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
50
|
Identification of IGF-1-enhanced cytokine expressions targeted by miR-181d in glioblastomas via an integrative miRNA/mRNA regulatory network analysis. Sci Rep 2017; 7:732. [PMID: 28389653 PMCID: PMC5429683 DOI: 10.1038/s41598-017-00826-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/14/2017] [Indexed: 02/03/2023] Open
Abstract
The insulin-like growth factor (IGF)-1 signaling is relevant in regulating cell growth and cytokine secretions by glioblastomas. MicroRNAs determine the cell fate in glioblastomas. However, relationships between IGF-1 signaling and miRNAs in glioblastoma pathogenesis are still unclear. Our aim was to validate the IGF-1-mediated mRNA/miRNA regulatory network in glioblastomas. Using in silico analyses of mRNA array and RNA sequencing data from The Cancer Genome Atlas (TCGA), we identified 32 core enrichment genes that were highly associated with IGF-1-promoted cytokine-cytokine receptor interactions. To investigate the IGF-1-downregulated miRNA signature, microarray-based approaches with IGF-1-treated U87-MG cells and array data in TCGA were used. Four miRNAs, including microRNA (miR)-9-5p, miR-9-3p, miR-181d, and miR-130b, exhibited an inverse correlation with IGF-1 levels. The miR-181d, that targeted the most IGF-1-related cytokine genes, was significantly reduced in IGF-1-treated glioma cells. Statistical models incorporating both high-IGF-1 and low-miR-181d statuses better predicted poor patient survival, and can be used as an independent prognostic factor in glioblastomas. The C-C chemokine receptor type 1 (CCR1) and interleukin (IL)-1b demonstrated inverse correlations with miR-181d levels and associations with patient survival. miR-181d significantly attenuated IGF-1-upregulated CCR1 and IL-1b gene expressions. These findings demonstrate a distinct role for IGF-1 signaling in glioma progression via miR-181d/cytokine networks.
Collapse
|