1
|
Kong D, Duan J, Chen S, Wang Z, Ren J, Lu J, Chen T, Song Z, Wu D, Chang Y, Yin Z, Shen Z, Zheng H. Transplant oncology and anti-cancer immunosuppressants. Front Immunol 2025; 15:1520083. [PMID: 39840041 PMCID: PMC11747528 DOI: 10.3389/fimmu.2024.1520083] [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: 10/30/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
Organ transplantation is a life-saving intervention that enhances the quality of life for patients with end-stage organ failure. However, long-term immunosuppressive therapy is required to prevent allogeneic graft rejection, which inadvertently elevates the risk of post-transplant malignancies, especially for liver transplant recipients with a prior history of liver cancer. In response, the emerging field of transplant oncology integrates principles from oncology and immunology to improve outcomes for patients at high risk of tumor occurrence or recurrence following transplantation. Therefore, it is of substantial clinical significance to develop immunosuppressants that possess both immunosuppressive and anti-tumor properties. For instance, mTOR inhibitors demonstrate anti-tumor effects among antimetabolite immunosuppressive drugs, and recent studies indicate that capecitabine, an antimetabolite chemotherapeutic, may also exhibit immunosuppressive activity in the clinic for liver transplants suffering from hepatocellular carcinoma. This review systematically explores potential immunosuppressants with dual anti-tumor and immunosuppressive effects to support the management of transplant patients at elevated risk of tumor occurrence or recurrence.
Collapse
Affiliation(s)
- Dejun Kong
- Nankai University School of Medicine, Tianjin, China
| | - Jinliang Duan
- Nankai University School of Medicine, Tianjin, China
| | - Shaofeng Chen
- Nankai University School of Medicine, Tianjin, China
| | - Zhenglu Wang
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
| | - Jiashu Ren
- Tianjin First Central Clinical College, Tianjin, China
| | - Jianing Lu
- Tianjin First Central Clinical College, Tianjin, China
| | - Tao Chen
- Nankai University School of Medicine, Tianjin, China
| | - Zhuolun Song
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
| | - Di Wu
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
| | - Yuan Chang
- Nankai University School of Medicine, Tianjin, China
| | - Zhongqian Yin
- Tianjin First Central Clinical College, Tianjin, China
| | - Zhongyang Shen
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| | - Hong Zheng
- Tianjin Organ Transplantation Research Center, Tianjin First Central Hospital, Nankai University School of Medicine, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| |
Collapse
|
2
|
Alkharsan AMHMS, Safaralizadeh R, Khalaj-Kondori M, HosseinpourFeizi M. Examination of the effects of capecitabine treatment on the HT-29 colorectal cancer cell line and HCG 11, HCG 15, and HCG 18 lncRNAs in CRC patients before and after chemotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024:10.1007/s00210-024-03674-8. [PMID: 39702604 DOI: 10.1007/s00210-024-03674-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/24/2024] [Indexed: 12/21/2024]
Abstract
Colorectal cancer (CRC) is the third most common malignancy worldwide. Long noncoding RNAs (lncRNAs) are involved in several pathogenic pathways related to CRC. This study aimed to compare the expression profiles of HCG11, HCG15, and HCG18 genes in CRC patients before and after chemotherapy. Moreover, capecitabine's effects, which is a chemotherapeutic agent, were investigated on apoptosis, cell cycle, and the lncRNA expression in CRC using HT-29 cells. qRT-PCR was used to measure lncRNAs expression in patient and healthy tissues, and the HT-29 CRC cell line. Additionally, the diagnostic and prognostic utility of these lncRNAs were assessed using the ROC curve analysis. The MTT assay was used to evaluate the cytotoxicity of capecitabine, and by using flow cytometry, apoptosis induction and cell cycle progression were investigated. CRC patients expressed higher levels of HCG11 and HCG15 and lower levels of HCG18. Furthermore, those receiving capecitabine demonstrated a decrease in HCG11 and an increase in HCG18 expression. In the HT-29 cell line, capecitabine can also increase the expression of HCG18 and decrease the expression of HCG11 and HCG15. However, no statistically significant variations were determined in the expression of these lncRNAs in clinical variables. Additionally, the data show that HCG18 is a poor prognostic biomarker, and HCG11 and HCG18 are poor diagnostic biomarkers. Treatment with capecitabine caused an accumulation of sub-G1 cells, indicating a potent apoptotic effect on HT-29 cells. These findings confirmed capecitabine's anticancer effects and showed that it can increase HCG18 and reduce HCG11 and HCG15 expression.
Collapse
Affiliation(s)
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
3
|
Zhou Z, Zhang Y, Xia S, Chen X. Red-Light-Activatable AND-Gated Antitumor Immunosuppressant. Cells 2023; 12:2351. [PMID: 37830565 PMCID: PMC10571834 DOI: 10.3390/cells12192351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/22/2023] [Accepted: 09/23/2023] [Indexed: 10/14/2023] Open
Abstract
Immunosuppressants are emerging as promising candidates for cancer therapy with lower cytotoxicity compared to traditional chemotherapy drugs; yet, the intrinsic side effects such as immunosuppression remain a critical concern. Herein, we introduce a photoactivatable antitumor immunosuppressant called dmBODIPY-FTY720 (BF) that shows no cytotoxicity but can be temporally and locally activated by deep-red light illumination to induce tumor cell apoptosis. To further reduce potential side effects, we integrate BF with another classic photosensitizer called methylene blue (MB) that is activated under the same wavelength of deep-red light (>650 nm) and successfully establish a red-light-activatable AND Boolean logic gate through a mechanism that we found to be synergetic apoptotic induction. At further decreased dosages, deep-red light illumination does not induce cell death in the presence of either BF or MB, but significant cancer cell death is triggered in the presence of both drugs. Therefore, the dosage of BF is further reduced, which will be highly beneficial to minimize any potential side effects of BF. This AND-gated strategy has been successfully applied in vivo for effective suppression of hepatocarcinoma tumors in living mice.
Collapse
Affiliation(s)
- Ziqi Zhou
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
- School of Life Science and Technology, Harbin Institute of Technology (HIT), Harbin 150001, China
| | - Yan Zhang
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
- School of Life Science and Technology, Harbin Institute of Technology (HIT), Harbin 150001, China
| | - Simin Xia
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
| | - Xi Chen
- Laboratory of Chemical Biology and Frontier Biotechnologies, The HIT Center for Life Sciences (HCLS), Harbin Institute of Technology (HIT), Harbin 150001, China; (Z.Z.); (Y.Z.)
- School of Life Science and Technology, Harbin Institute of Technology (HIT), Harbin 150001, China
| |
Collapse
|
4
|
Abe H, Kamimura K, Okuda S, Watanabe Y, Inoue J, Aoyagi Y, Wakai T, Kominami R, Terai S. BCL11B expression in hepatocellular carcinoma relates to chemosensitivity and clinical prognosis. Cancer Med 2023; 12:15650-15663. [PMID: 37293953 PMCID: PMC10417273 DOI: 10.1002/cam4.6167] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/14/2023] [Accepted: 05/17/2023] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION B-cell lymphoma/leukemia 11B (BCL11B) is a subunit of SWI/SNF chromatin remodeling complexes and functions in cell cycle regulation and apoptosis upon DNA replication stress and damages via transcription. Many malignancies were reported to exhibit changes in BCL11B gene expression; however, no study has focused on the relationship between BCL11B and hepatocellular carcinoma, which potentially exhibits DNA replication stress and damages upon its oncogenesis. Thus, in this study, we examined the molecular characterization of BCL11B expression in hepatocellular carcinoma. METHODS AND RESULTS The cumulative progression-free survival and overall survival were significantly longer in the clinical cases of BCL11B-negative hepatocellular carcinoma than BCL11B-positve cases. Microarray and real-time PCR analyses in hepatocellular carcinoma cell lines indicated a correlation between BCL11B and GATA6, a gene reported to be correlated with oncogenic activities and resistance to anthracycline, which is often used for hepatocellular carcinoma chemotherapy. Consequently, BCL11B-overexpressing cell lines exhibited resistance to anthracycline in cell growth assays and the resistance has been evidenced by the increased expression of BCL-xL in cell lines. The results were supported by the analyses of human HCC samples showing the correlation between BCL11B and GATA6 expressions. DISCUSSIONS AND CONCLUSION Our results indicated that overexpression of BCL11B amplifies GATA6 expression in hepatocellular carcinoma in vitro and in vivo that leads to anti-apoptotic signal activation, and induces resistance to chemotherapy, which influenced the postoperative prognosis.
Collapse
Affiliation(s)
- Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
- Department of General MedicineNiigata University School of MedicineNiigataNiigataJapan
| | - Shujiro Okuda
- Division of Bioinformatics, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Yu Watanabe
- Division of Bioinformatics, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Jun Inoue
- Department of Agricultural Chemistry, Faculty of Applied BiosciencesTokyo University of AgricultureTokyoJapan
| | - Yutaka Aoyagi
- Department of Gastroenterology and HepatologyNiigata Medical CenterNiigataNiigataJapan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Ryo Kominami
- Department of Molecular Genetics, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental SciencesNiigata UniversityNiigataNiigataJapan
| |
Collapse
|
5
|
Wang H, Wang ZL, Zhang S, Kong DJ, Yang RN, Cao L, Wang JX, Yoshida S, Song ZL, Liu T, Fan SL, Ren JS, Li JH, Shen ZY, Zheng H. Metronomic capecitabine inhibits liver transplant rejection in rats by triggering recipients' T cell ferroptosis. World J Gastroenterol 2023; 29:3084-3102. [PMID: 37346150 PMCID: PMC10280797 DOI: 10.3748/wjg.v29.i20.3084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/19/2023] [Accepted: 04/28/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Capecitabine (CAP) is a classic antimetabolic drug and has shown potential antirejection effects after liver transplantation (LT) in clinical studies. Our previous study showed that metronomic CAP can cause the programmed death of T cells by inducing oxidative stress in healthy mice. Ferroptosis, a newly defined non-apoptotic cell death that occurs in response to iron overload and lethal levels of lipid peroxidation, is an important mechanism by which CAP induces cell death. Therefore, ferroptosis may also play an important role in CAP-induced T cell death and play an immunosuppressive role in acute rejection after trans-plantation. AIM To investigate the functions and underlying mechanisms of antirejection effects of metronomic CAP. METHODS A rat LT model of acute rejection was established, and the effect of metronomic CAP on splenic hematopoietic function and acute graft rejection was evaluated 7 d after LT. In vitro, primary CD3+ T cells were sorted from rat spleens and human peripheral blood, and co-cultured with or without 5-fluorouracil (5-FU) (active agent of CAP). The levels of ferroptosis-related proteins, ferrous ion concentration, and oxidative stress-related indicators were observed. The changes in mito-chondrial structure were observed using electron microscopy. RESULTS With no significant myelotoxicity, metronomic CAP alleviated graft injury (Banff score 9 vs 7.333, P < 0.001), prolonged the survival time of the recipient rats (11.5 d vs 16 d, P < 0.01), and reduced the infiltration rate of CD3+ T cells in peripheral blood (6.859 vs 3.735, P < 0.001), liver graft (7.459 vs 3.432, P < 0.001), and spleen (26.92 vs 12.9, P < 0.001), thereby inhibiting acute rejection after LT. In vitro, 5-FU, an end product of CAP metabolism, induced the degradation of the ferritin heavy chain by upregulating nuclear receptor coactivator 4, which caused the accumulation of ferrous ions. It also inhibited nuclear erythroid 2 p45-related factor 2, heme oxygenase-1, and glutathione peroxidase 4, eventually leading to oxidative damage and ferroptosis of T cells. CONCLUSION Metronomic CAP can suppress acute allograft rejection in rats by triggering CD3+ T cell ferroptosis, which makes it an effective immunosuppressive agent after LT.
Collapse
Affiliation(s)
- Hao Wang
- The First Central Clinical School, Tianjin Medical University, Tianjin 300190, China
| | - Zheng-Lu Wang
- Department of Organ Transplant, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300190, China
| | - Sai Zhang
- School of Medicine, Nankai University, Tianjin 300190, China
| | - De-Jun Kong
- School of Medicine, Nankai University, Tianjin 300190, China
| | - Rui-Ning Yang
- The First Central Clinical School, Tianjin Medical University, Tianjin 300190, China
| | - Lei Cao
- Research Institute of Transplant Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300071, China
| | - Jian-Xi Wang
- Research Institute of Transplant Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300071, China
| | - Sei Yoshida
- Research Institute of Transplant Medicine, Nankai University, Tianjin 300071, China
| | - Zhuo-Lun Song
- Department of Organ Transplant, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China
| | - Tao Liu
- National Health Commission’s Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300071, China
| | - Shun-Li Fan
- Department of Organ Transplant, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China
| | - Jia-Shu Ren
- The First Central Clinical School, Tianjin Medical University, Tianjin 300190, China
| | - Jiang-Hong Li
- The First Central Clinical School, Tianjin Medical University, Tianjin 300190, China
| | - Zhong-Yang Shen
- Department of Organ Transplant, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300190, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300071, China
- National Health Commission’s Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300071, China
| | - Hong Zheng
- Department of Organ Transplant, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300190, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin 300190, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin 300071, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300071, China
- National Health Commission’s Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Tianjin 300071, China
| |
Collapse
|
6
|
Donnenberg VS, Luketich JD, Sultan I, Lister J, Bartlett DL, Ghosh S, Donnenberg AD. A maladaptive pleural environment suppresses preexisting anti-tumor activity of pleural infiltrating T cells. Front Immunol 2023; 14:1157697. [PMID: 37063842 PMCID: PMC10097923 DOI: 10.3389/fimmu.2023.1157697] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/03/2023] [Indexed: 04/18/2023] Open
Abstract
INTRODUCTION Treatment options for patients with malignant pleural effusions (MPE) are limited due, at least in part, to the unique environment of the pleural space, which drives an aggressive tumor state and governs the behavior of infiltrating immune cells. Modulation of the pleural environment may be a necessary step toward the development of effective treatments. We examine immune checkpoint molecule (ICM) expression on pleural T cells, the secretomes of pleural fluid, pleural infiltrating T cells (PIT), and ability to activate PIT ex vivo. METHODS ICM expression was determined on freshly drained and in vitro activated PIT from breast, lung and renal cell cancer. Secretomics (63 analytes) of activated PIT, primary tumor cultures and MPE fluid was determined using Luminex technology. Complementary digital spatial proteomic profiling (42 analytes) of CD45+ MPE cells was done using the Nanostring GeoMx platform. Cytolytic activity was measured against autologous tumor targets. RESULTS ICM expression was low on freshy isolated PIT; regulatory T cells (T-reg) were not detectable by GeoMx. In vitro activated PIT coexpressed PD-1, LAG-3 and TIGIT but were highly cytotoxic against autologous tumor and uniquely secreted cytokines and chemokines in the > 100 pM range. These included CCL4, CCL3, granzyme B, IL-13, TNFα, IL-2 IFNγ, GM-CSF, and perforin. Activated PIT also secreted high levels of IL-6, IL-8 and sIL-6Rα, which contribute to polarization of the pleural environment toward wound healing and the epithelial to mesenchymal transition. Addition of IL-6Rα antagonist to cultures reversed tumor EMT but did not alter PIT activation, cytokine secretion or cytotoxicity. DISCUSSION Despite the negative environment, immune effector cells are plentiful, persist in MPE in a quiescent state, and are easily activated and expanded in culture. Low expression of ICM on native PIT may explain reported lack of responsiveness to immune checkpoint blockade. The potent cytotoxic activity of activated PIT and a proof-of-concept clinical scale GMP-expansion experiment support their promise as a cellular therapeutic. We expect that a successful approach will require combining cellular therapy with pleural conditioning using immune checkpoint blockers together with inhibitors of upstream master cytokines such as the IL-6/IL-6R axis.
Collapse
Affiliation(s)
- Vera S. Donnenberg
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Centers, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| | - James D. Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Centers, Pittsburgh, PA, United States
| | - Ibrahim Sultan
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
| | - John Lister
- Department of Medicine, Division of Hematology and Cellular Therapy, Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
- Drexel University College of Medicine, Philadelphia, PA, United States
| | - David L. Bartlett
- Drexel University College of Medicine, Philadelphia, PA, United States
- Department of Surgery, Division of Surgical Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA, United States
| | - Sohini Ghosh
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Allegheny Health Network, Pittsburgh, PA, United States
| | - Albert D. Donnenberg
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Centers, Pittsburgh, PA, United States
- McGowan Institute for Regenerative Medicine, Pittsburgh, PA, United States
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
7
|
Sun H, Yang Z, Teng Z, Zhang Y, Han Z, Xu C, Wang Z, Wang H, Wen H, Chen X, Qu C, Wang Y. DDX58 expression promotes inflammation and growth arrest in Sertoli cells by stabilizing p65 mRNA in patients with Sertoli cell-only syndrome. Front Immunol 2023; 14:1135753. [PMID: 37033952 PMCID: PMC10073560 DOI: 10.3389/fimmu.2023.1135753] [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: 01/01/2023] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Sertoli cell -only syndrome (SCOS) is a type of testicular pathological failure that causes male infertility and no effective treatment strategy, is available for this condition. Moreover, the molecular mechanism underlying its development remains unknown. We identified DExD/H-Box helicase 58 (DDX58) as a key gene in SCOS based on four datasets of testicular tissue samples obtained from the Gene Expression Synthesis database. DDX58 was significantly upregulated in SCOS testicular Sertoli cells. Moreover, high expression of DDX58 was positively correlated with the expression of several testicular inflammatory factors, such as IL -1β, IL-18, and IL-6. Interestingly, DDX58 could be induced in the D-galactose (D-gal)-stimulated TM4 cell injury model. Whereas silencing of DDX58 inhibited D-gal -mediated p65 expression, inflammatory cytokine release, and growth arrest. Mechanistically, we found that DDX58 acts as an RNA-binding protein, which enhances p65 expression by promoting mRNA stability. Furthermore, p65 gene silencing decreased the expression of inflammatory cytokines and inhibition of cell growth in D-gal-induced cells. In conclusion, our findings demonstrate that DDX58 promotes inflammatory responses and growth arrest in SCOS Sertoli cells by stabilizing p65 mRNA. Accordingly, the DDX58/p65 regulatory axis might be a therapeutic target for SCOS.
Collapse
Affiliation(s)
- Hao Sun
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhan Yang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Molecular Biology Laboratory, Talent and Academic Exchange Center, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhihai Teng
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yanping Zhang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhenwei Han
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chao Xu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhu Wang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hu Wang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Hongzhuang Wen
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaodong Chen
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Changbao Qu
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Changbao Qu, ; Yaxuan Wang,
| | - Yaxuan Wang
- Department of Urology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- *Correspondence: Changbao Qu, ; Yaxuan Wang,
| |
Collapse
|
8
|
Yang H, Xu L, Guan S, Hao X, Ge Z, Tong F, Cao Y, Liu P, Zhou B, Cheng L, Liu M, Liu H, Xie F, Wang S, Peng Y, Wang C, Wang S. Neoadjuvant docetaxel and capecitabine (TX) versus docetaxel and epirubicin (TE) for locally advanced or early her2-negative breast cancer: an open-label, randomized, multi-center, phase II Trial. BMC Cancer 2022; 22:1357. [PMID: 36577958 PMCID: PMC9795638 DOI: 10.1186/s12885-022-10439-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/12/2022] [Indexed: 12/29/2022] Open
Abstract
PURPOSE The combination of taxanes and anthracyclines is still the mainstay of chemotherapy for early breast cancer. Capecitabine is an active drug with a favorable toxicity profile, showing strong anti-tumor activity against metastatic breast cancer. This trial assessed the efficacy and safety of the TX regimen (docetaxel and capecitabine) and compared it with the TE (docetaxel and epirubicin) regimen in locally advanced or high risk early HER2-negative breast cancer. PATIENTS AND METHODS This randomized clinical trial was conducted at five academic centers in China. Eligible female patients were randomly assigned (1:1) to the TX (docetaxel 75 mg/m2 d1 plus capecitabine 1000 mg/m2 twice d1-14, q3w) or TE (docetaxel 75 mg/m2 d1 plus epirubicin 75 mg/m2 d1, q3w) groups for four cycles. The primary endpoint was a pathological complete response in the breast (pCR). Secondary endpoints included pCR in the breast and axilla, invasive disease-free survival (iDFS), overall survival (OS), and safety. RESULTS Between September 1, 2012, and December 31, 2018, 113 HER2-negative patients were randomly assigned to the study groups (TX: n = 54; TE: n = 59). In the primary endpoint analysis, 14 patients in the TX group achieved a pCR, and nine patients in the TE group achieved a pCR (25.9% vs. 15.3%), with a not significant difference of 10.6% (95% CI -6.0-27.3%; P = 0.241). In a subgroup with high Ki-67 score, TX increased the pCR rate by 24.2% (95% CI 2.2-46.1%; P = 0.029). At the end of the 69-month median follow-up period, both groups had equivalent iDFS and OS rates. TX was associated with a higher incidence of hand-foot syndrome and less alopecia, with a manageable toxicity profile. CONCLUSION The anthracycline-free TX regimen yielded comparable pCR and long-term survival rates to the TE regimen. Thus, this anthracycline-free regimen could be considered in selected patients. TRIAL REGISTRATION ACTRN12613000206729 on 21/02/2013, retrospectively registered.
Collapse
Affiliation(s)
- Houpu Yang
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Ling Xu
- grid.411472.50000 0004 1764 1621Breast Disease Center, Peking University First Hospital, Beijing, China
| | - Shan Guan
- grid.414373.60000 0004 1758 1243Department of Breast Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xiaopeng Hao
- grid.414252.40000 0004 1761 8894Department of General Surgery, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhicheng Ge
- grid.411610.30000 0004 1764 2878Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Fuzhong Tong
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Yingming Cao
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Peng Liu
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Bo Zhou
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Lin Cheng
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Miao Liu
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Hongjun Liu
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Fei Xie
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Siyuan Wang
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Yuan Peng
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Chaobin Wang
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| | - Shu Wang
- grid.411634.50000 0004 0632 4559Peking University People’s Hospital Breast Center, Beijing, China
| |
Collapse
|
9
|
Bai Y, Wu H, Zhang J, Zhang S, Zhang Z, Wang H, Zhang Y, Shen Z. IL-22 Protects against Biliary Ischemia-Reperfusion Injury after Liver Transplantation via Activating STAT3 and Reducing Apoptosis and Oxidative Stress Levels In Vitro and In Vivo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9635075. [PMID: 35592531 PMCID: PMC9113870 DOI: 10.1155/2022/9635075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022]
Abstract
Biliary complications are currently one of the leading causes of liver failure and patient death after liver transplantation and need to be solved urgently. Biliary ischemia-reperfusion injury (IRI) is one of the important causes of biliary complications. IL-22 has a protective effect on liver injury and hepatitis diseases, and its safety and efficacy in the treatment of hepatitis have also been proved in human clinical experiments. Furthermore, multiple studies have confirmed that IL-22 promotes the proliferation and repair of epithelial cells in various organs. Still, its function in the bile duct after transplantation has not been explored. This study was aimed at investigating the effects of IL-22 on cholangiocyte IRI in vitro and in vivo and exploring its underlying mechanisms. We simulated the hypoxia process of bile duct epithelial cells through in vitro experiments to investigate the protective function and molecular mechanism of IL-22 on bile duct epithelial cells. Subsequently, the function and mechanism of IL-22 in the biliary IRI model of autologous orthotopic liver transplantation in rats were assessed. This study confirmed that IL-22 could promote cholangiocyte proliferation, decrease the apoptosis rate of cholangiocytes and tissues, decrease MDA levels, and increase SOD levels by activating STAT3. In addition, IL-22 can also reduce the level of mitochondrial membrane depolarization, protect mitochondria, reduce ROS production, and play a role in protecting bile ducts. These findings provide evidence for IL-22 as a novel and effective treatment for biliary IRI after liver transplantation.
Collapse
Affiliation(s)
- Yi Bai
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Hao Wu
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Jinrui Zhang
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Sai Zhang
- School of Medicine, Nankai University, Tianjin, China
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
| | - Zhixin Zhang
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Hao Wang
- The First Central Clinical School, Tianjin Medical University, Tianjin, China
| | - Yamin Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Zhongyang Shen
- Department of Organ Transplantation, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin, China
| |
Collapse
|
10
|
Zhang S, Fan S, Wang Z, Hou W, Liu T, Yoshida S, Yang S, Zheng H, Shen Z. Capecitabine Regulates HSP90AB1 Expression and Induces Apoptosis via Akt/SMARCC1/AP-1/ROS Axis in T Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1012509. [PMID: 35368874 PMCID: PMC8970866 DOI: 10.1155/2022/1012509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/10/2022] [Indexed: 11/17/2022]
Abstract
Transplant oncology is a newly emerging discipline integrating oncology, transplant medicine, and surgery and has brought malignancy treatment into a new era via transplantation. In this context, obtaining a drug with both immunosuppressive and antitumor effects can take into account the dual needs of preventing both transplant rejection and tumor recurrence in liver transplantation patients with malignancies. Capecitabine (CAP), a classic antitumor drug, has been shown to induce reactive oxygen species (ROS) production and apoptosis in tumor cells. Meanwhile, we have demonstrated that CAP can induce ROS production and apoptosis in T cells to exert immunosuppressive effects, but its underlying molecular mechanism is still unclear. In this study, metronomic doses of CAP were administered to normal mice by gavage, and the spleen was selected for quantitative proteomic and phosphoproteomic analysis. The results showed that CAP significantly reduced the expression of HSP90AB1 and SMARCC1 in the spleen. It was subsequently confirmed that CAP also significantly reduced the expression of HSP90AB1 and SMARCC1 and increased ROS and apoptosis levels in T cells. The results of in vitro experiments showed that HSP90AB1 knockdown resulted in a significant decrease in p-Akt, SMARCC1, p-c-Fos, and p-c-Jun expression levels and a significant increase in ROS and apoptosis levels. HSP90AB1 overexpression significantly inhibited CAP-induced T cell apoptosis by increasing the p-Akt, SMARCC1, p-c-Fos, and p-c-Jun expression levels and reducing the ROS level. In conclusion, HSP90AB1 is a key target of CAP-induced T cell apoptosis via Akt/SMARCC1/AP-1/ROS axis, which provides a novel understanding of CAP-induced T cell apoptosis and lays the experimental foundation for further exploring CAP as an immunosuppressant with antitumor effects to optimize the medication regimen for transplantation patients.
Collapse
Affiliation(s)
- Sai Zhang
- School of Medicine, Nankai University, Tianjin, China
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
| | - Shunli Fan
- First Central Clinical Institute, Tianjin Medical University, Tianjin, China
| | - Zhenglu Wang
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin First Central Hospital, Tianjin, China
| | - Wen Hou
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Tao Liu
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin, China
| | - Sei Yoshida
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
| | - Shuang Yang
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
| | - Hong Zheng
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin First Central Hospital, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| | - Zhongyang Shen
- Organ Transplant Department, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin, China
- Key Laboratory of Transplant Medicine, Chinese Academy of Medical Sciences, Tianjin First Central Hospital, Tianjin, China
- National Health Commission's Key Laboratory for Critical Care Medicine, Tianjin, China
- Research Institute of Transplant Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory for Organ Transplantation, Tianjin, China
| |
Collapse
|