1
|
Wang R, Dong X, Zhang X, Liao J, Cui W, Li W. Exploring viral mimicry combined with epigenetics and tumor immunity: new perspectives in cancer therapy. Int J Biol Sci 2025; 21:958-973. [PMID: 39897033 PMCID: PMC11781167 DOI: 10.7150/ijbs.103877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025] Open
Abstract
Viral mimicry refers to an active antiviral response triggered by the activation of endogenous retroviruses (ERVs), usually manifested by the formation of double-stranded RNA (dsRNA) and activation of the cellular interferon response, which activates the immune system and produces anti-tumor effects. Epigenetic studies have shown that epigenetic modifications (e.g. DNA methylation, histone modifications, etc.) play a crucial role in tumorigenesis, progression, and treatment resistance. Particularly, alterations in DNA methylation may be closely associated with the suppression of ERVs expression, and treatment by demethylation may restore ERVs activity and thus strengthen the tumor immune response. Therefore, we propose that viral mimicry can induce immune responses in the tumor microenvironment by activating the expression of ERVs, and that epigenetic alterations may play a key regulatory role in this process. In this paper, we review the intersection of viral mimicry, epigenetics and tumor immunotherapy, and explore the possible interactions and synergistic effects among the three, aiming to provide a new theoretical basis and potential strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Ruirui Wang
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| | - Xin Dong
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiongjian Zhang
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| | - Jinzhuang Liao
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University. Tongzipo Road 138, Changsha, Hunan, People's Republic of China
| |
Collapse
|
2
|
Al Mamun A, Suchi SA, Aziz MA, Zaeem M, Munir F, Wu Y, Xiao J. Pyroptosis in acute pancreatitis and its therapeutic regulation. Apoptosis 2022; 27:465-481. [PMID: 35687256 DOI: 10.1007/s10495-022-01729-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2022] [Indexed: 12/20/2022]
Abstract
Pyroptosis defines a new type of GSDMs-mediated programmed cell death, distinguishes from the classical concepts of apoptosis and necrosis-mediated cell death and is prescribed by cell swelling and membrane denaturation, leading to the extensive secretion of cellular components and low-grade inflammatory response. However, NLRP3 inflammasome activation can trigger its downstream inflammatory cytokines, leading to the activation of pyroptosis-regulated cell death. Current studies reveal that activation of caspase-4/5/11-driven non-canonical inflammasome signaling pathways facilitates the pathogenesis and progression of acute pancreatitis (AP). In addition, a large number of studies have reported that NLRP3 inflammasome-dependent pyroptosis is a crucial player in driving the course of the pathogenesis of AP. Excessive uncontrolled GSDMD-mediated pyroptosis has been implicated in AP. Therefore, the pyroptosis-related molecule GSDMD may be an independent prognostic biomarker for AP. The present review paper summarizes the molecular mechanisms of pyroptotic signaling pathways and their pathophysiological impacts on the progress of AP. Moreover, we briefly present some experimental compounds targeting pyroptosis-regulated cell death for exploring novel therapeutic directions for the treatment and management of AP. Our review investigations strongly suggest that targeting pyroptosis could be an ideal therapeutic approach in AP.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Suzia Aktar Suchi
- Department of Pharmacy, College of Pharmacy, Chosun University, Gwangju, 501759, South Korea
| | - Md Abdul Aziz
- Department of Pharmacy, Faculty of Pharmacy and Health Sciences, State University of Bangladesh, Dhaka, 1205, Bangladesh.,Laboratory of Pharmacogenomics and Molecular Biology, Department of Pharmacy, Noakhali Science and Technology University, Noakhali, 3814, Bangladesh
| | - Muhammad Zaeem
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Zhejiang Province, Wenzhou, 325035, China
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China. .,Department of Hand Surgery and Peripheral Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| |
Collapse
|
3
|
Zhang Q, Bai X, Wang R, Zhao H, Wang L, Liu J, Li M, Chen Z, Wang Z, Li L, Wang D. 4‐octyl Itaconate inhibits lipopolysaccharide (LPS)‐induced osteoarthritis via activating Nrf2 signalling pathway. J Cell Mol Med 2022; 26:1515-1529. [PMID: 35068055 PMCID: PMC8899168 DOI: 10.1111/jcmm.17185] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/18/2021] [Accepted: 01/03/2022] [Indexed: 12/23/2022] Open
Abstract
Small molecule drug intervention for chondrocytes is a valuable method for the treatment of osteoarthritis (OA). The 4‐octyl itaconate (OI) is a cellular derivative of itaconate with sound cell permeability and transformation rate. We attempted to confirm the protective role of OI in chondrocytes and its regulatory mechanism. We used lipopolysaccharide (LPS) to induce chondrocyte inflammation injury. After the OI treatment, the secretion and mRNA expression of Il‐6, Il‐10, Mcp‐1 and Tnf‐α were detected by ELISA and qPCR. The protective effect of OI on articular cartilage was further verified in surgical destabilization of the medial meniscus model of OA. Cell death and apoptosis were evaluated based on CCK8, LDH, Typan blue staining, Annexin V and TUNEL analyses. The small interfering RNAs were used to knockout the Nrf2 gene of chondrocytes to verify the OI‐mediated Nrf2 signalling pathway. The results revealed that OI protects cells from LPS‐induced inflammatory injury and attenuates cell death and apoptosis induced by LPS. Similar protective effects were also observed on articular cartilage in mice. The OI activated Nrf2 signalling pathway and promoted the stable expression and translocation of Nrf2 into the nucleus. When the Nrf2 signalling pathway was blocked, the protective effect of OI was significantly counteracted in chondrocytes and a mouse arthritis model. Both itaconate and its derivative (i.e., OI) showed important medical effects in the treatment of OA.
Collapse
Affiliation(s)
- Qingchen Zhang
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Xiaohui Bai
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Rongrong Wang
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Hao Zhao
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Lili Wang
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Jingwen Liu
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Ming Li
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Zheng Chen
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Zejun Wang
- Department of Clinical Laboratory Shandong Provincial Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Lianxin Li
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Dawei Wang
- Department of Orthopaedics Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan China
- Department of Orthopaedics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| |
Collapse
|
4
|
Razeghian E, Suksatan W, Sulaiman Rahman H, Bokov DO, Abdelbasset WK, Hassanzadeh A, Marofi F, Yazdanifar M, Jarahian M. Harnessing TRAIL-Induced Apoptosis Pathway for Cancer Immunotherapy and Associated Challenges. Front Immunol 2021; 12:699746. [PMID: 34489946 PMCID: PMC8417882 DOI: 10.3389/fimmu.2021.699746] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 08/05/2021] [Indexed: 01/04/2023] Open
Abstract
The immune cytokine tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted rapidly evolving attention as a cancer treatment modality because of its competence to selectively eliminate tumor cells without instigating toxicity in vivo. TRAIL has revealed encouraging promise in preclinical reports in animal models as a cancer treatment option; however, the foremost constraint of the TRAIL therapy is the advancement of TRAIL resistance through a myriad of mechanisms in tumor cells. Investigations have documented that improvement of the expression of anti-apoptotic proteins and survival or proliferation involved signaling pathways concurrently suppressing the expression of pro-apoptotic proteins along with down-regulation of expression of TRAILR1 and TRAILR2, also known as death receptor 4 and 5 (DR4/5) are reliable for tumor cells resistance to TRAIL. Therefore, it seems that the development of a therapeutic approach for overcoming TRAIL resistance is of paramount importance. Studies currently have shown that combined treatment with anti-tumor agents, ranging from synthetic agents to natural products, and TRAIL could result in induction of apoptosis in TRAIL-resistant cells. Also, human mesenchymal stem/stromal cells (MSCs) engineered to generate and deliver TRAIL can provide both targeted and continued delivery of this apoptosis-inducing cytokine. Similarly, nanoparticle (NPs)-based TRAIL delivery offers novel platforms to defeat barricades to TRAIL therapeutic delivery. In the current review, we will focus on underlying mechanisms contributed to inducing resistance to TRAIL in tumor cells, and also discuss recent findings concerning the therapeutic efficacy of combined treatment of TRAIL with other antitumor compounds, and also TRAIL-delivery using human MSCs and NPs to overcome tumor cells resistance to TRAIL.
Collapse
Affiliation(s)
- Ehsan Razeghian
- Human Genetics Division, Medical Biotechnology Department, National Institute of Genetics Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Wanich Suksatan
- Faculty of Nursing, HRH Princess Chulabhorn College of Medical Science, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Suleimanyah, Iraq
- Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaimaniyah, Iraq
| | - Dmitry O. Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russia
- Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russia
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Ali Hassanzadeh
- Department of Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faroogh Marofi
- Immunology Research Center (IRC), Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahboubeh Yazdanifar
- Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, United States
| | - Mostafa Jarahian
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
5
|
Yang C, Sun Y, Ouyang X, Li J, Zhu Z, Yu R, Wang L, Jia L, Ding G, Wang Y, Jiang F. Pain May Promote Tumor Progression via Substance P-Dependent Modulation of Toll-like Receptor-4. PAIN MEDICINE 2021; 21:3443-3450. [PMID: 32914185 DOI: 10.1093/pm/pnaa265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND In a previous study, persistent pain was suggested to be a risk factor for tumor patients. However, the mechanism underlying this phenomenon is still unclear. Substance P (SP), a pain-related neuropeptide secreted by the neural system and the immune system, plays an important role in the induction and maintenance of persistent pain. METHODS In this study, in order to explore whether SP participates in the influence of pain on tumor progression, the serum samples of lung cancer and breast cancer patients were collected and tested. An elevated expression of SP was found in patients with pain. RESULTS Cell pharmacological experiments revealed that SP can upregulate the expression of Toll-like receptor-4 (TLR-4) in tumor cells and increase the proliferation, migration, and invasive activity of tumor cells. As high expression of TLR-4 has the ability to enhance the biological activity of tumor cells, TLR-4 is thought to be involved in SP-induced tumor proliferation, migration, and invasion. Treatment of tumor cells with Aprepitant, a specific blocker of the NK-1 receptor, could reduce the expression of TLR-4 and reduce the proliferation, invasion, and migration activities of tumor cells; further proof of the influence of SP on TLR-4 expression depends on the NK-1 receptor located in tumor cells. CONCLUSIONS Based on the results above, we proposed a possible mechanism underlying pain affecting tumor progression: The presence of pain increases the content of SP in patients' blood, and elevated SP increases the expression of tumor TLR-4 by acting on the NK-1 receptor, which ultimately affects the biological activity of the tumor.
Collapse
Affiliation(s)
- Chao Yang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Yunheng Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Ouyang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Jing Li
- Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhen Zhu
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Ruihua Yu
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Li Wang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Lin Jia
- Shanghai International Medical Center, Shanghai, China
| | - Gang Ding
- Shanghai International Medical Center, Shanghai, China
| | - Yaosheng Wang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| | - Feng Jiang
- Translational Institute for Cancer Pain, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Chongming Branch, Shanghai, China
| |
Collapse
|
6
|
Tanshinone IIA sensitizes TRAIL-induced apoptosis in glioblastoma through inducing the expression of death receptors (and suppressing STAT3 activation). Brain Res 2021; 1766:147515. [PMID: 33984327 DOI: 10.1016/j.brainres.2021.147515] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/12/2021] [Accepted: 05/06/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVE This work was designed to explore whether the combination of Tanshinone IIA (T-IIA) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has a direct anti-cancer effect in glioblastoma (GBM) and the possible mechanisms. METHODS GBM cells (U-87 and U-251 MG) were treated with T-IIA or/and TRAIL, or the expression of death receptors (DRs), DR4 and DR5, was suppressed in GBM cells. The activity of GBM cells was determined by MTT, and the apoptosis was assessed by Hoechst33342 staining and flow cytometry. The expression levels of cleaved caspase-3/8/9, phosphorylated (p)-STAT3 as well as DR4 and DR5 in GBM cells were assessed by Western blotting. A nude mouse xenograft model was constructed to evaluate the effects of T-IIA and TRAIL cotreatment on tumor growth and apoptosis in vivo. RESULTS After T-IIA treatment, GBM cells resumed the sensitivity to TRAIL-induced apoptosis dependent on inhibition of p-STAT3 and activation of DR4, DR5 and caspases. DR4 or/and DR5 knockdown significantly abated the co-effect of T-IIA and TRAIL on GBM cell apoptosis and proliferation. Furthermore, T-IIA and TRAIL cotreatment markedly inhibited the growth of transplanted tumor and activated U87 cell apoptosis in nude mice. CONCLUSION T-IIA increases TRAIL-induced apoptosis by downregulating STAT3 and upregulating DR4 and DR5, indicating T-IIA therapy as a novel treatment strategy for TRAIL-resistant GBM.
Collapse
|
7
|
Kashani B, Zandi Z, Pourbagheri-Sigaroodi A, Bashash D, Ghaffari SH. The role of toll-like receptor 4 (TLR4) in cancer progression: A possible therapeutic target? J Cell Physiol 2020; 236:4121-4137. [PMID: 33230811 DOI: 10.1002/jcp.30166] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/13/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022]
Abstract
The toll-like receptor (TLR) family consists of vital receptors responsible for pattern recognition in innate immunity, making them the core proteins involved in pathogen detection and eliciting immune responses. The most studied member of this family, TLR4, has been the center of attention regarding its contributory role in many inflammatory diseases including sepsis shock and asthma. Notably, mounting pieces of evidence have proved that this receptor is aberrantly expressed on the tumor cells and the tumor microenvironment in a wide range of cancer types and it is highly associated with the initiation of tumorigenesis as well as tumor progression and drug resistance. Cancer therapy using TLR4 inhibitors has recently drawn scientists' attention, and the promising results of such studies may pave the way for more investigation in the foreseeable future. This review will introduce the key proteins of the TLR4 pathway and how they interact with major growth factors in the tumor microenvironment. Moreover, we will discuss the many aspects of tumor progression affected by the activation of this receptor and provide an overview of the recent therapeutic approaches using various TLR4 antagonists.
Collapse
Affiliation(s)
- Bahareh Kashani
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Zandi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
8
|
Pan H, Dai H, Wang L, Lin S, Tao Y, Zheng Y, Jiang R, Fang F, Wu Y. MicroRNA-410-3p modulates chondrocyte apoptosis and inflammation by targeting high mobility group box 1 (HMGB1) in an osteoarthritis mouse model. BMC Musculoskelet Disord 2020; 21:486. [PMID: 32709223 PMCID: PMC7379779 DOI: 10.1186/s12891-020-03489-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Background Osteoarthritis (OA) is the most prevalent type of arthritis, which commonly involves inflammation in the articular cartilage in OA pathogenesis. MicroRNAs (miRNAs) play essential roles in the regulation and pathophysiology of various diseases including OA. MiR-410-3p has been demonstrated to mediate inflammatory pathways, however, the regulatory functions of miR-410-3p in OA remain largely unknown. Methods The regulations of miR-410-3p were investigated in OA. Mouse primary chondrocytes and mouse in vivo models were used. The expression levels of miR-410-3p and HMGB1 were measured by qPCR. The transcription activity of NF-κB was assessed by luciferase reporter assay. MTT assay was performed to assess cellular proliferation. Cell apoptosis was evaluated with the Fluorescein Isothiocyanate (FITC) Annexin V assay. Expression levels of proteins were determined by Western blot. Results The results demonstrated that miR-410-3p was markedly downregulated in articular cartilage tissues as well as in lipopolysaccharide (LPS)-treated chondrocytes in OA mice. In addition, upregulation of miR-410-3p markedly inhibited LPS-induced apoptosis of chondrocytes. The results also demonstrated that the high mobility group box 1 (HMGB1) was a target of miR-410-3p. LPS-induced upregulated expression of HMGB1 significantly suppressed expression of miR-410-3p. Furthermore, upregulation of miR-410-3p markedly inhibited HMGB1 expression, the nuclear factor (NF)-kB activity and pro-inflammatory cytokines production. Taken together, the results suggested that miR-410-3p targeted HMGB1 and modulated chondrocytes apoptosis and inflammation through the NF-κB signaling pathway. Conclusions These findings provide insights into the potential of miR-410-3p/ HMGB1 as therapeutic targets for OA treatment.
Collapse
Affiliation(s)
- Hong Pan
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China.
| | - Huming Dai
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Linzhi Wang
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Silong Lin
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Yuefeng Tao
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Yi Zheng
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Renyi Jiang
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Fan Fang
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| | - Yifan Wu
- Department of Orthopaedics, Affiliated Anqing Hospital of Anhui Medical University, No.352 Ren Min Road, Yingjiang District, Anqing City, 246003, Anhui Province, China
| |
Collapse
|
9
|
Cao Y, Kong S, Xin Y, Meng Y, Shang S, Qi Y. Lestaurtinib potentiates TRAIL-induced apoptosis in glioma via CHOP-dependent DR5 induction. J Cell Mol Med 2020; 24:7829-7840. [PMID: 32441887 PMCID: PMC7348155 DOI: 10.1111/jcmm.15415] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 04/19/2020] [Accepted: 05/03/2020] [Indexed: 12/22/2022] Open
Abstract
Lestaurtinib, also called CEP-701, is an inhibitor of tyrosine kinase, causes haematological remission in patients with AML possessing FLT3-ITD (FLT3 gene) internal tandem duplication and strongly inhibits tyrosine kinase FLT3. Treatment with lestaurtinib modulates various signalling pathways and leads to cell growth arrest and programmed cell death in several tumour types. However, the effect of lestaurtinib on glioma remains unclear. In this study, we examined lestaurtinib and TRAIL interactions in glioma cells and observed their synergistic activity on glioma cell apoptosis. While U87 and U251 cells showed resistance to TRAIL single treatment, they were sensitized to apoptosis induced by TRAIL in the presence of lestaurtinib because of increased death receptor 5 (DR5) levels through CHOP-dependent manner. We also demonstrated using a xenograft model of mouse that the tumour growth was absolutely suppressed because of the combined treatment compared to TRAIL or lestaurtinib treatment carried out singly. Our findings reveal a potential new strategy to improve antitumour activity induced by TRAIL in glioma cells using lestaurtinib through a mechanism dependent on CHOP.
Collapse
Affiliation(s)
- Yingxiao Cao
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Shiqi Kong
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Yuling Xin
- Department of NeurosurgeryXingtai People’s HospitalXingtaiChina
| | - Yan Meng
- Department of Operating RoomXingtai People’s HospitalXingtaiChina
| | - Shuling Shang
- Department of Operating RoomXingtai People’s HospitalXingtaiChina
| | - Yanhui Qi
- Department of Intensive Care UnitXingtai People’s HospitalXingtaiChina
| |
Collapse
|
10
|
Interactions of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) with the Immune System: Implications for Inflammation and Cancer. Cancers (Basel) 2019; 11:cancers11081161. [PMID: 31412671 PMCID: PMC6721490 DOI: 10.3390/cancers11081161] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily. TRAIL has historically been distinct from the Fas ligand and TNFα in terms of selective apoptosis induction in tumor cells and has a nearly non-existent systemic toxicity. Consequently, in the search for an ideal drug for tumor therapy, TRAIL rapidly drew interest, promising effective tumor control with minimal side effects. However, euphoria gave way to disillusionment as it turned out that carcinoma cells possess or can acquire resistance to TRAIL-induced apoptosis. Additionally, studies on models of inflammation and autoimmunity revealed that TRAIL can influence immune cells in many different ways. While TRAIL was initially found to be an important player in tumor defense by natural killer cells or cytotoxic T cells, additional effects of TRAIL on regulatory T cells and effector T cells, as well as on neutrophilic granulocytes and antigen-presenting cells, became focuses of interest. The tumor-promoting effects of these interactions become particularly important for consideration in cases where tumors are resistant to TRAIL-induced apoptosis. Consequently, murine models have shown that TRAIL can impair the tumor microenvironment toward a more immunosuppressive type, thereby promoting tumor growth. This review summarizes the current state of knowledge on TRAIL’s interactions with the immune system in the context of cancer.
Collapse
|
11
|
Zhang Q, Wang Y, Zhang M, Ying H. Green tea polyphenols attenuate LPS-induced inflammation through upregulating microRNA-9 in murine chondrogenic ATDC5 cells. J Cell Physiol 2019; 234:22604-22612. [PMID: 31102286 DOI: 10.1002/jcp.28826] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Osteoarthritis (OA), a universal chronic musculoskeletal disorder, is closely related to inflammation. More effective drugs for improving OA outcome are definitely needed. Herein, we attempted to verify the protective role of green tea polyphenols (GTP) after treatment with murine in ATDC5 cells to reveal the regulatory mechanism. METHODS ATDC5 cells were stimulated with lipopolysaccharide (LPS) to mimic an inflammatory response during OA. Cell activity, apoptosis, levels of relative proteins, and prophlogistic factors were tested via a Cell Counting Kit-8 experiment, a flow cytometry experiment, western blot, and RT-qPCR (ELISA and Western blot), separately. miR-9 level was detected by RT-qPCR and altered via miR-9 mimic and inhibitor transfection. We finally studied MAPK and NF-κB pathways in GTP-related modulations using western blot. RESULTS LPS caused inflammatory cell damage in ATDC5 cells, showing decreased cell activity, enhanced apoptosis, and increased levels of pro-inflammatory cytokines. GTP pretreatments could significantly attenuate LPS-induced alterations. In addition, LPS-induced miR-9 upregulation was further positively regulated in ATDC5 cells. The effects of GTP pretreatments in LPS-caused ATDC5 cells were enhanced via miR-9 upregulation, whereas they were reduced via miR-9 suppression. Finally, we found that GTP pretreatments could suppress the MAPK and NF-κB pathways through miR-9 regulation. CONCLUSION GTP pretreatments attenuated LPS-induced inflammatory response accompanied by the suppression of the MAPK and NF-κB pathways via positively regulating miR-9 in ATDC5 cells.
Collapse
Affiliation(s)
- Qiao Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Yongkun Wang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Mingran Zhang
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Hongliang Ying
- Department of Orthopaedics, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
12
|
Rubio-Manzanares Dorado M, Marín Gómez LM, Aparicio Sánchez D, Pereira Arenas S, Praena-Fernández JM, Borrero Martín JJ, Farfán López F, Gómez Bravo MÁ, Muntané Relat J, Padillo Ruiz J. Translational pancreatic cancer research: A comparative study on patient-derived xenograft models. World J Gastroenterol 2018; 24:794-809. [PMID: 29467550 PMCID: PMC5807938 DOI: 10.3748/wjg.v24.i7.794] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/14/2018] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the viability of orthotopic and heterotopic patient-derived pancreatic cancer xenografts implanted into nude mice. METHODS This study presents a prospective experimental analytical follow-up of the development of tumours in mice upon implantation of human pancreatic adenocarcinoma samples. Specimens were obtained surgically from patients with a pathological diagnosis of pancreatic adenocarcinoma. Tumour samples from pancreatic cancer patients were transplanted into nude mice in three different locations (intraperitoneal, subcutaneous and pancreatic). Histological analysis (haematoxylin-eosin and Masson's trichrome staining) and immunohistochemical assessment of apoptosis (TUNEL), proliferation (Ki-67), angiogenesis (CD31) and fibrogenesis (α-SMA) were performed. When a tumour xenograft reached the target size, it was re-implanted in a new nude mouse. Three sequential tumour xenograft generations were generated (F1, F2 and F3). RESULTS The overall tumour engraftment rate was 61.1%. The subcutaneous model was most effective in terms of tissue growth (69.9%), followed by intraperitoneal (57.6%) and pancreatic (55%) models. Tumour development was faster in the subcutaneous model (17.7 ± 2.6 wk) compared with the pancreatic (23.1 ± 2.3 wk) and intraperitoneal (25.0 ± 2.7 wk) models (P = 0.064). There was a progressive increase in the tumour engraftment rate over successive generations for all three models (F1 28.1% vs F2 71.4% vs F3 80.9%, P < 0.001). There were no significant differences in tumour xenograft differentiation and cell proliferation between human samples and the three experimental models among the sequential generations of tumour xenografts. However, a progressive decrease in fibrosis, fibrogenesis, tumour vascularisation and apoptosis was observed in the three experimental models compared with the human samples. All three pancreatic patient-derived xenograft models presented similar histological and immunohistochemical characteristics. CONCLUSION In our experience, the faster development and greatest number of viable xenografts could make the subcutaneous model the best option for experimentation in pancreatic cancer.
Collapse
Affiliation(s)
| | - Luis Miguel Marín Gómez
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Daniel Aparicio Sánchez
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Sheila Pereira Arenas
- Oncology Surgery, Cell Therapy, and Organ Transplantation Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, University of Seville, Seville 41013, Spain
| | - Juan Manuel Praena-Fernández
- Statistics, Methodology and Evaluation of Research Unit, Virgen del Rocio University Hospital, Seville 41013, Spain
| | | | | | - Miguel Ángel Gómez Bravo
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| | - Jordi Muntané Relat
- Oncology Surgery, Cell Therapy, and Organ Transplantation Group, Institute of Biomedicine of Seville (IBiS), Virgen del Rocio University Hospital, University of Seville, Seville 41013, Spain
| | - Javier Padillo Ruiz
- Department of Hepatobiliary and Pancreatic Surgery, Virgen del Rocio University Hospital, Seville 41013, Spain
| |
Collapse
|