151
|
Hoblos R, Kefalakes H. Immunology of hepatitis D virus infection: General concepts and present evidence. Liver Int 2023; 43 Suppl 1:47-59. [PMID: 36074070 DOI: 10.1111/liv.15424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 08/27/2022] [Accepted: 09/05/2022] [Indexed: 02/13/2023]
Abstract
Infection with the hepatitis D virus induces the most severe form of chronic viral hepatitis, affecting over 12 million people worldwide. Chronic HDV infection leads to rapid development of liver cirrhosis and hepatocellular carcinoma in ~70% of patients within 15 years of infection. Recent evidence suggests that an interplay of different components of the immune system are contributing to viral control and may even be implicated in liver disease pathogenesis. This review will describe general concepts of antiviral immune response and elicit the present evidence concerning the interplay of the hepatitis D virus with the immune system.
Collapse
Affiliation(s)
- Reem Hoblos
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Helenie Kefalakes
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
152
|
Jia D, Lu Y, Lv M, Wang F, Lu X, Zhu W, Wei J, Guo W, Liu R, Li G, Wang R, Li J, Yuan F. Targeted co-delivery of resiquimod and a SIRPα variant by liposomes to activate macrophage immune responses for tumor immunotherapy. J Control Release 2023; 360:858-871. [PMID: 37473808 DOI: 10.1016/j.jconrel.2023.07.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/25/2023] [Accepted: 07/18/2023] [Indexed: 07/22/2023]
Abstract
Tumor-associated macrophages (TAMs) are the major immune cells infiltrating the tumor microenvironment (TME) and typically exhibit an immunosuppressive M2-like phenotype, which facilitates tumor growth and promotes resistance to immunotherapy. Additionally, tumor cells tend to express high levels of CD47, a "don't eat me" signal, that obstructs macrophage phagocytosis. Consequently, re-educating TAMs in combination with CD47 blockage is promising to trigger intense macrophage immune responses against tumors. As a toll-like receptor 7/8 agonist, resiquimod (R848) possesses the capacity to re-educate TAMs from M2 type to M1 type. We found that intratumoral administration of R848 synergistically improved the antitumor immunotherapeutic effect of CV1 protein (a SIRPα variant with high antagonism to CD47). However, the poor bioavailability and potential toxicity of this combo strategy remain a challenge. Here, a TAMs-targeted liposome (named: R-LS/M/CV1) co-delivering R848 and CV1 protein was constructed via decorating mannose on the liposomal surface. R-LS/M/CV1 exhibited high abilities of targeting, re-education and pro-phagocytosis of tumor cells to M2 macrophages in vitro. Intratumoral administration of R-LS/M/CV1 remarkedly eliminated tumor burden in the MC38 tumor model via repolarization of TAMs to M1 type, pro-phagocytosis of TAMs against tumors, and recruitment of tumor-infiltrating T cells. More encouragingly, due to the double targeting to TAMs and tumor cells of mannose and CV1 protein, R-LS/M/CV1 effectively accumulated at the tumor site, thereby not only remarkedly inhibiting tumors, but also exerting no hematological and histopathological toxicity when administered systemically. Our integrated strategy based on re-educating TAMs and CD47 blockade provides a promising approach to trigger macrophage immune responses against tumors for immunotherapy.
Collapse
Affiliation(s)
- Dianlong Jia
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Yue Lu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China.
| | - Mingjia Lv
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Feifei Wang
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Xiaomeng Lu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Weifan Zhu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Jianmei Wei
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China
| | - Wen Guo
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Renmin Liu
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Guangyong Li
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Rui Wang
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China
| | - Jun Li
- Laboratory of Drug Discovery and Design, School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, Shandong 252000, PR China.
| | - Fengjiao Yuan
- Joint Laboratory for Translational Medicine Research, Liaocheng People's Hospital, Liaocheng, Shandong 252000, PR China.
| |
Collapse
|
153
|
Zhang Y, Jiang M, Du G, Zhong X, He C, Qin M, Hou Y, Liu R, Sun X. An antigen self-assembled and dendritic cell-targeted nanovaccine for enhanced immunity against cancer. Acta Pharm Sin B 2023; 13:3518-3534. [PMID: 37655327 PMCID: PMC10465870 DOI: 10.1016/j.apsb.2022.03.017] [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: 01/06/2022] [Revised: 02/22/2022] [Accepted: 02/23/2022] [Indexed: 11/30/2022] Open
Abstract
The rise of nanotechnology has opened new horizons for cancer immunotherapy. However, most nanovaccines fabricated with nanomaterials suffer from carrier-related concerns, including low drug loading capacity, unpredictable metabolism, and potential systemic toxicity, which bring obstacles for their clinical translation. Herein, we developed an antigen self-assembled nanovaccine, which was resulted from a simple acryloyl modification of the antigen to induce self-assembly. Furthermore, a dendritic cell targeting head mannose monomer and a mevalonate pathway inhibitor zoledronic acid (Zol) were integrated or absorbed onto the nanoparticles (denoted as MEAO-Z) to intensify the immune response. The synthesized nanovaccine with a diameter of around 70 nm showed successful lymph node transportation, high dendritic cell internalization, promoted costimulatory molecule expression, and preferable antigen cross-presentation. In virtue of the above superiorities, MEAO-Z induced remarkably higher titers of serum antibody, stronger cytotoxic T lymphocyte immune responses and IFN-γ secretion than free antigen and adjuvants. In vivo, MEAO-Z significantly suppressed EG7-OVA tumor growth and prolonged the survival time of tumor-bearing mice. These results indicated the translation promise of our self-assembled nanovaccine for immune potentiation and cancer immunotherapy.
Collapse
Affiliation(s)
| | | | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaofang Zhong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Chunting He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Ming Qin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yingying Hou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Rong Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
154
|
Ildefonso GV, Finley SD. A data-driven Boolean model explains memory subsets and evolution in CD8+ T cell exhaustion. NPJ Syst Biol Appl 2023; 9:36. [PMID: 37524735 PMCID: PMC10390540 DOI: 10.1038/s41540-023-00297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023] Open
Abstract
T cells play a key role in a variety of immune responses, including infection and cancer. Upon stimulation, naïve CD8+ T cells proliferate and differentiate into a variety of memory and effector cell types; however, failure to clear antigens causes prolonged stimulation of CD8+ T cells, ultimately leading to T cell exhaustion (TCE). The functional and phenotypic changes that occur during CD8+ T cell differentiation are well characterized, but the underlying gene expression state changes are not completely understood. Here, we utilize a previously published data-driven Boolean model of gene regulatory interactions shown to mediate TCE. Our network analysis and modeling reveal the final gene expression states that correspond to TCE, along with the sequence of gene expression patterns that give rise to those final states. With a model that predicts the changes in gene expression that lead to TCE, we could evaluate strategies to inhibit the exhausted state. Overall, we demonstrate that a common pathway model of CD8+ T cell gene regulatory interactions can provide insights into the transcriptional changes underlying the evolution of cell states in TCE.
Collapse
Affiliation(s)
- Geena V Ildefonso
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA
| | - Stacey D Finley
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA.
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, California, USA.
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
155
|
Li H, Xu H, Guo H, Du K, Chen D. Integrative analysis illustrates the role of PCDH7 in lung cancer development, cisplatin resistance, and immunotherapy resistance: an underlying target. Front Pharmacol 2023; 14:1217213. [PMID: 37538171 PMCID: PMC10394841 DOI: 10.3389/fphar.2023.1217213] [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: 05/05/2023] [Accepted: 06/30/2023] [Indexed: 08/05/2023] Open
Abstract
Background: Cisplatin resistance is a common clinical problem in lung cancer. However, the underlying mechanisms have not yet been fully elucidated, highlighting the importance of searching for biological targets. Methods: Bioinformatics analysis is completed through downloaded public data (GSE21656, GSE108214, and TCGA) and specific R packages. The evaluation of cell proliferation ability is completed through CCK8 assay, colony formation, and EdU assay. The evaluation of cell invasion and migration ability is completed through transwell and wound-healing assays. In addition, we evaluated cell cisplatin sensitivity by calculating IC50. Results: Here, we found that PCDH7 may be involved in cisplatin resistance in lung cancer through public database analysis (GSE21656 and GSE108214). Then, a series of in vitro experiments was performed, which verified the cancer-promoting role of PCDH7 in NSCLC. Moreover, the results of IC50 detection showed that PCDH7 might be associated with cisplatin resistance of NSCLC. Next, we investigated the single-cell pattern, biological function, and immune analysis of PCDH7. Importantly, we noticed PCDH7 may regulate epithelial-mesenchymal transition activity, and the local infiltration of CD8+ T and activated NK cells. Furthermore, we noticed that patients with high PCDH7 expression might be more sensitive to bortezomib, docetaxel, and gemcitabine, and resistant to immunotherapy. Finally, a prognosis model based on three PCDH7-derived genes (GPX8, BCAR3, and TNS4) was constructed through a machine learning algorithm, which has good prediction ability on NSCLC patients' survival. Conclusion: Our research has improved the regulatory framework for cisplatin resistance in NSCLC and can provide direction for subsequent related research, especially regarding PCDH7.
Collapse
|
156
|
Wu X, Zhang Y, Liang G, Ye H. Cuproptosis-related lncRNAs potentially predict prognosis and therapy sensitivity of breast cancer. Front Pharmacol 2023; 14:1199883. [PMID: 37529698 PMCID: PMC10390311 DOI: 10.3389/fphar.2023.1199883] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 07/03/2023] [Indexed: 08/03/2023] Open
Abstract
Background: Cuproptosis-related lncRNAs regulate the biological functions of various cancers. However, the role of cuproptosis-related lncRNAs in breast cancer remains unclear. In this study, we investigated the biological functions and clinical applications of cuproptosis-related lncRNAs in breast cancer. Methods: The Cancer Genome Atlas (TCGA) database and the GSE20685 dataset were used for screening cuproptosis-related lncRNAs. Colony formation and CCK-8 kit assays were performed for detecting the proliferative function of cuproptosis-related lncRNAs, whereas wound healing, migration, and invasion assays were performed for detecting the metastatic regulation of cuproptosis-related lncRNAs in breast cancer. Finally, a prognostic cuproptosis-related lncRNA model was constructed using LASSO Cox regression analysis for detecting survival and sensitivity to conventional treatment (endocrine therapy, chemotherapy, and radiotherapy) and novel therapy (PARP and CDK4/6 inhibitors). Results: In this study, we screened six cuproptosis-related lncRNAs associated with the survival of patients with breast cancer. Biofunctional experiments indicated that cuproptosis-related lncRNAs play essential roles in regulating the proliferation and metastasis of breast cancer cells. Finally, we applied a model of six cuproptosis-related lncRNAs to classify the patients into high- and low-risk groups. High-risk group patients exhibited worse survival rates (p < 0.001) and lower sensitivity to chemotherapy, endocrine therapy, and radiation therapy. Compared with high-risk patients, low-risk patients exhibited a lower expression of CDK4/6 inhibitor-resistant biomarkers (CCNE1, E2F1, and E2F2) and PARP inhibitor-resistant biomarkers (BRCA1/BRCA2), indicating that patients in the low-risk group were more suitable for PARP inhibitor and CDK4/6 inhibitor application. Conclusion: Cuproptosis-related lncRNAs are essential for regulating the biological functions of breast cancer, and they have the potential to predict prognosis and sensitivity of breast cancer to various therapies.
Collapse
Affiliation(s)
- Xiwen Wu
- Department of Clinical Nutrition, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying Zhang
- Staff and Faculty Clinic, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Gehao Liang
- Department of Breast Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Huizhen Ye
- Staff and Faculty Clinic, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
157
|
Wang YY, Zhen C, Hu W, Huang HH, Li YJ, Zhou MJ, Li J, Fu YL, Zhang P, Li XY, Yang T, Song JW, Fan X, Zou J, Meng SR, Qin YQ, Jiao YM, Xu R, Zhang JY, Zhou CB, Yuan JH, Huang L, Shi M, Cheng L, Wang FS, Zhang C. Elevated glutamate impedes anti-HIV-1 CD8 + T cell responses in HIV-1-infected individuals on antiretroviral therapy. Commun Biol 2023; 6:696. [PMID: 37419968 PMCID: PMC10328948 DOI: 10.1038/s42003-023-04975-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/24/2023] [Indexed: 07/09/2023] Open
Abstract
CD8 + T cells are essential for long-lasting HIV-1 control and have been harnessed to develop therapeutic and preventive approaches for people living with HIV-1 (PLWH). HIV-1 infection induces marked metabolic alterations. However, it is unclear whether these changes affect the anti-HIV function of CD8 + T cells. Here, we show that PLWH exhibit higher levels of plasma glutamate than healthy controls. In PLWH, glutamate levels positively correlate with HIV-1 reservoir and negatively correlate with the anti-HIV function of CD8 + T cells. Single-cell metabolic modeling reveals glutamate metabolism is surprisingly robust in virtual memory CD8 + T cells (TVM). We further confirmed that glutamate inhibits TVM cells function via the mTORC1 pathway in vitro. Our findings reveal an association between metabolic plasticity and CD8 + T cell-mediated HIV control, suggesting that glutamate metabolism can be exploited as a therapeutic target for the reversion of anti-HIV CD8 + T cell function in PLWH.
Collapse
Affiliation(s)
- You-Yuan Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Cheng Zhen
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Wei Hu
- Department of Emergency, Fifth Medical Center of Chinese PLA Hospital, Beijing, China
| | - Hui-Huang Huang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yan-Jun Li
- Guangxi AIDS Clinical Treatment Centre, The Fourth People's Hospital of Nanning, Nanning, China
| | - Ming-Ju Zhou
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jing Li
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Yu-Long Fu
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Peng Zhang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xiao-Yu Li
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Tao Yang
- Medical School of Chinese PLA, Beijing, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Wen Song
- Medical School of Chinese PLA, Beijing, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xing Fan
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jun Zou
- Guangxi AIDS Clinical Treatment Centre, The Fourth People's Hospital of Nanning, Nanning, China
| | - Si-Run Meng
- Guangxi AIDS Clinical Treatment Centre, The Fourth People's Hospital of Nanning, Nanning, China
| | - Ya-Qin Qin
- Guangxi AIDS Clinical Treatment Centre, The Fourth People's Hospital of Nanning, Nanning, China
| | - Yan-Mei Jiao
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruonan Xu
- Medical School of Chinese PLA, Beijing, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ji-Yuan Zhang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chun-Bao Zhou
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Hong Yuan
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Lei Huang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ming Shi
- Medical School of Chinese PLA, Beijing, China
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Liang Cheng
- Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Fu-Sheng Wang
- Medical School of Chinese PLA, Beijing, China.
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
- Guangxi AIDS Clinical Treatment Centre, The Fourth People's Hospital of Nanning, Nanning, China.
| | - Chao Zhang
- Medical School of Chinese PLA, Beijing, China.
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
- Guangxi AIDS Clinical Treatment Centre, The Fourth People's Hospital of Nanning, Nanning, China.
| |
Collapse
|
158
|
Pan Y, Yang W, Tang B, Wang X, Zhang Q, Li W, Li L. The protective and pathogenic role of Th17 cell plasticity and function in the tumor microenvironment. Front Immunol 2023; 14:1192303. [PMID: 37457739 PMCID: PMC10339829 DOI: 10.3389/fimmu.2023.1192303] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/19/2023] [Indexed: 07/18/2023] Open
Abstract
At the turn of the century, researchers discovered a unique subtype of T helper cells that secretes IL-17 and defined it as Th17. The latest study found that Th17 cells play both positive and negative definitive roles in the regulation of antitumor immune responses. Although the function of Th17 in the tumor microenvironment remains poorly understood, more and more studies have shown that this paradoxical dual role is closely related to the plasticity of Th17 cells in recent decades. Further understanding of the characteristics of Th17 cells in the tumor microenvironment could yield novel and useful therapeutic approaches to treat cancer. In this review, we further present the high plasticity of Th17 cells and the function of Th17-producing IL-17 in tumor immunity.
Collapse
|
159
|
Lista F, Peragallo MS, Biselli R, De Santis R, Mariotti S, Nisini R, D'Amelio R. Have Diagnostics, Therapies, and Vaccines Made the Difference in the Pandemic Evolution of COVID-19 in Comparison with "Spanish Flu"? Pathogens 2023; 12:868. [PMID: 37513715 PMCID: PMC10384375 DOI: 10.3390/pathogens12070868] [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: 04/26/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
In 1918 many countries, but not Spain, were fighting World War I. Spanish press could report about the diffusion and severity of a new infection without censorship for the first-time, so that this pandemic is commonly defined as "Spanish flu", even though Spain was not its place of origin. "Spanish flu" was one of the deadliest pandemics in history and has been frequently compared with the coronavirus disease (COVID)-19 pandemic. These pandemics share similarities, being both caused by highly variable and transmissible respiratory RNA viruses, and diversity, represented by diagnostics, therapies, and especially vaccines, which were made rapidly available for COVID-19, but not for "Spanish flu". Most comparison studies have been carried out in the first period of COVID-19, when these resources were either not yet available or their use had not long started. Conversely, we wanted to analyze the role that the advanced diagnostics, anti-viral agents, including monoclonal antibodies, and innovative COVID-19 vaccines, may have had in the pandemic containment. Early diagnosis, therapies, and anti-COVID-19 vaccines have markedly reduced the pandemic severity and mortality, thus preventing the collapse of the public health services. However, their influence on the reduction of infections and re-infections, thus on the transition from pandemic to endemic condition, appears to be of minor relevance. The high viral variability of influenza and coronavirus may probably be contained by the development of universal vaccines, which are not easy to be obtained. The only effective weapon still remains the disease prevention, to be achieved with the reduction of promiscuity between the animal reservoirs of these zoonotic diseases and humans.
Collapse
Affiliation(s)
- Florigio Lista
- Istituto di Scienze Biomediche della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
| | - Mario Stefano Peragallo
- Centro Studi e Ricerche di Sanità e Veterinaria, Comando Logistico dell'Esercito, 00184 Roma, Italy
| | - Roberto Biselli
- Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
| | - Riccardo De Santis
- Istituto di Scienze Biomediche della Difesa, Ispettorato Generale della Sanità Militare, Stato Maggiore della Difesa, 00184 Roma, Italy
- Dipartimento di Sanità Pubblica e Malattie Infettive, Sapienza, Università di Roma, 00161 Roma, Italy
| | - Sabrina Mariotti
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, 00161 Roma, Italy
| | - Raffaele D'Amelio
- Dipartimento di Medicina Clinica e Molecolare, Sapienza, Università di Roma, 00198 Roma, Italy
| |
Collapse
|
160
|
Guan Q, Han M, Guo Q, Yan F, Wang M, Ning Q, Xi D. Strategies to reinvigorate exhausted CD8 + T cells in tumor microenvironment. Front Immunol 2023; 14:1204363. [PMID: 37398660 PMCID: PMC10311918 DOI: 10.3389/fimmu.2023.1204363] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
CD8+ T cell exhaustion is a stable dysfunctional state driven by chronic antigen stimulation in the tumor microenvironment (TME). Differentiation of exhausted CD8+ T cells (CD8+ TEXs) is accompanied by extensive transcriptional, epigenetic and metabolic reprogramming. CD8+ TEXs are mainly characterized by impaired proliferative and cytotoxic capacity as well as the increased expression of multiple co-inhibitory receptors. Preclinical tumor studies and clinical cohorts have demonstrated that T cell exhaustion is firmly associated with poor clinical outcomes in a variety of cancers. More importantly, CD8+ TEXs are regarded as the main responder to immune checkpoint blockade (ICB). However, to date, a large number of cancer patients have failed to achieve durable responses after ICB. Therefore, improving CD8+ TEXs may be a breakthrough point to reverse the current dilemma of cancer immunotherapy and eliminate cancers. Strategies to reinvigorate CD8+ TEXs in TME mainly include ICB, transcription factor-based therapy, epigenetic therapy, metabolism-based therapy and cytokine therapy, which target on different aspects of exhaustion progression. Each of them has its advantages and application scope. In this review, we mainly focus on the major advances of current strategies to reinvigorate CD8+ TEXs in TME. We summarize their efficacy and mechanisms, identify the promising monotherapy and combined therapy and propose suggestions to enhance the treatment efficacy to significantly boost anti-tumor immunity and achieve better clinical outcomes.
Collapse
|
161
|
Ramgopal A, Braverman EL, Sun LK, Monlish D, Wittmann C, Ramsey MJ, Caitley R, Hawse W, Byersdorfer CA. AMPK Drives Both Glycolytic and Oxidative Metabolism in T Cells During Graft-versus-host Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.12.544686. [PMID: 37398326 PMCID: PMC10312647 DOI: 10.1101/2023.06.12.544686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Allogeneic T cells reprogram their metabolism during acute graft-versus-host disease (GVHD) in a process reliant on the cellular energy sensor AMP-activated protein kinase (AMPK). Deletion of AMPK in donor T cells limits GVHD but still preserves homeostatic reconstitution and graft-versus-leukemia (GVL) effects. In the current studies, murine T cells lacking AMPK decreased oxidative metabolism at early timepoints post-transplant and were also unable to mediate a compensatory increase in glycolysis following inhibition of the electron transport chain. Human T cells lacking AMPK gave similar results, with glycolytic compensation impaired both in vitro and following expansion in vivo in a modified model of GVHD. Immunoprecipitation of proteins from day 7 allogeneic T cells, using an antibody specific to phosphorylated AMPK targets, recovered lower levels of multiple glycolysis-related proteins including the glycolytic enzymes aldolase, enolase, pyruvate kinase M (PKM), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). Functionally, murine T cells lacking AMPK exhibited impaired aldolase activity following anti-CD3/CD28 stimulation and a decrease in GAPDH activity on day 7 post-transplant. Importantly, these changes in glycolysis correlated with an impaired ability of AMPK KO T cells to produce significant amounts of interferon gamma (IFNγ) upon antigenic re-stimulation. Together these data highlight a significant role for AMPK in controlling oxidative and glycolytic metabolism in both murine and human T cells during GVHD and endorse further study of AMPK inhibition as a potential target for future clinical therapies. KEY POINTS AMPK plays a key role in driving both and oxidative and glycolytic metabolism in T cells during graft-versus-host disease (GVHD)Absence of AMPK simultaneously impairs both glycolytic enzyme activity, most notably by aldolase, and interferon gamma (IFNγ) production.
Collapse
|
162
|
Hibino S, Eto S, Hangai S, Endo K, Ashitani S, Sugaya M, Osawa T, Soga T, Taniguchi T, Yanai H. Tumor cell-derived spermidine is an oncometabolite that suppresses TCR clustering for intratumoral CD8 + T cell activation. Proc Natl Acad Sci U S A 2023; 120:e2305245120. [PMID: 37276392 PMCID: PMC10268234 DOI: 10.1073/pnas.2305245120] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/03/2023] [Indexed: 06/07/2023] Open
Abstract
The activation and expansion of T cells that recognize cancer cells is an essential aspect to antitumor immunity. Tumors may escape destruction by the immune system through ectopic expression of inhibitory immune ligands typically exemplified by the PD-L1/PD-1 pathway. Here, we reveal another facet of tumor evasion from T cell surveillance. By secretome profiling of necrotic tumor cells, we identified an oncometabolite spermidine as a unique inhibitor of T cell receptor (TCR) signaling. Mechanistically, spermidine causes the downregulation of the plasma membrane cholesterol levels, resulting in the suppression of TCR clustering. Using syngeneic mouse models, we show that spermidine is abundantly detected in the tumor immune microenvironment (TIME) and that administration of the polyamine synthesis inhibitor effectively enhanced CD8+ T cell-dependent antitumor responses. Further, the combination of the polyamine synthesis inhibitor with anti-PD-1 immune checkpoint antibody resulted in a much stronger antitumor immune response. This study reveals an aspect of immunosuppressive TIME, wherein spermidine functions as a metabolic T cell checkpoint that may offer a unique approach for promoting tumor immunotherapy.
Collapse
Affiliation(s)
- Sana Hibino
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-0041, Japan
| | - Shotaro Eto
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-0041, Japan
| | - Sho Hangai
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-0041, Japan
| | - Keiko Endo
- Institute for Advanced Biosciences, Keio University, Yamagata997-0052, Japan
| | - Sanae Ashitani
- Institute for Advanced Biosciences, Keio University, Yamagata997-0052, Japan
| | - Maki Sugaya
- Division of Nutriomics and Oncology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-8904, Japan
| | - Tsuyoshi Osawa
- Division of Nutriomics and Oncology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-8904, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata997-0052, Japan
| | - Tadatsugu Taniguchi
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-0041, Japan
| | - Hideyuki Yanai
- Department of Inflammology, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo153-0041, Japan
| |
Collapse
|
163
|
Lu W, Bai Y, Zhang S, Zhao X, Jin J, Zhu X, Wang R, Wu Y, Zhang A, Zhang G, Zhuang G, Sun A. An Intracellular Epitope of ASFV CD2v Protein Elicits Humoral and Cellular Immune Responses. Animals (Basel) 2023; 13:1967. [PMID: 37370477 DOI: 10.3390/ani13121967] [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: 05/04/2023] [Revised: 06/10/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
The African swine fever virus (ASFV) causes high mortality in domestic pigs. ASFV encodes an important protein target for subunit vaccine development, CD2v, but its most effective immunological regions are not known. Herein, we generated a monoclonal antibody (mAb) named IF3 by immunizing mice against the intracellular region of the CD2v protein (CD2v-IR). 1F3 specifically recognized CD2v, which is expressed transiently in transfected Sf9 cells and also in inactivated ASFV-infected porcine alveolar macrophage (PAM) cells. The epitope recognized by 1F3 is 264EPSPREP270, which is highly conserved in ASFV genotypes. Immunization of mice with this epitope elicited an increased IgG response, including IgG1 and IgG2a subtypes, and also increased CD8+ T cells and cytokine expression. Overall, these results indicate that this epitope induces both humoral and cellular immune responses that may be used for ASFV-related subunit vaccine design and development.
Collapse
Affiliation(s)
- Wenlong Lu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yilin Bai
- Laboratory of Indigenous Cattle Germplasm Innovation, School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Shuai Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Xuyang Zhao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Jiaxin Jin
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Xiaojing Zhu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Rui Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Yanan Wu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Angke Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou 450046, China
| | - Guoqing Zhuang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| | - Aijun Sun
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
164
|
Turner JA, Fredrickson MA, D'Antonio M, Katsnelson E, MacBeth M, Van Gulick R, Chimed TS, McCarter M, D'Alessandro A, Robinson WA, Couts KL, Pelanda R, Klarquist J, Tobin RP, Torres RM. Lysophosphatidic acid modulates CD8 T cell immunosurveillance and metabolism to impair anti-tumor immunity. Nat Commun 2023; 14:3214. [PMID: 37270644 PMCID: PMC10239450 DOI: 10.1038/s41467-023-38933-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 05/19/2023] [Indexed: 06/05/2023] Open
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid which increases in concentration locally and systemically across different cancer types. Yet, the exact mechanism(s) of how LPA affects CD8 T cell immunosurveillance during tumor progression remain unknown. We show LPA receptor (LPAR) signaling by CD8 T cells promotes tolerogenic states via metabolic reprogramming and potentiating exhaustive-like differentiation to modulate anti-tumor immunity. We found LPA levels predict response to immunotherapy and Lpar5 signaling promotes cellular states associated with exhausted phenotypes on CD8 T cells. Importantly, we show that Lpar5 regulates CD8 T cell respiration, proton leak, and reactive oxygen species. Together, our findings reveal that LPA serves as a lipid-regulated immune checkpoint by modulating metabolic efficiency through LPAR5 signaling on CD8 T cells. Our study offers key insights into the mechanisms governing adaptive anti-tumor immunity and demonstrates LPA could be exploited as a T cell directed therapy to improve dysfunctional anti-tumor immunity.
Collapse
Affiliation(s)
- Jacqueline A Turner
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
- Medical Scientist Training Program, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Malia A Fredrickson
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Marc D'Antonio
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Elizabeth Katsnelson
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Morgan MacBeth
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Robert Van Gulick
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Tugs-Saikhan Chimed
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Martin McCarter
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - William A Robinson
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Kasey L Couts
- Division of Medical Oncology, Department of Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Roberta Pelanda
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Richard P Tobin
- Division of Surgical Oncology, Department of Surgery, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Raul M Torres
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
165
|
Rochussen AM, Lippert AH, Griffiths GM. Imaging the T-cell receptor: new approaches, new insights. Curr Opin Immunol 2023; 82:102309. [PMID: 37011462 DOI: 10.1016/j.coi.2023.102309] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/02/2023] [Accepted: 03/03/2023] [Indexed: 04/03/2023]
Abstract
T cells recognize pathogenic antigens via the T-cell antigen receptor (TCR). This protein complex binds to antigen fragments on the surface of antigen-presenting cells. To understand how cellular activation can ensue rapidly from molecular recognition, the localization and distribution of the TCR on the surface of the resting T cell are of particular importance. Conflicting results regarding TCR distribution have emerged from recent studies using a range of imaging techniques, including total internal reflection and single-molecule localization microscopy modalities. Here, we review the differing results and the potential biases inherent in differing imaging approaches. In addition, we review studies showing the impact of differing imaging surfaces on T-cell activation.
Collapse
|
166
|
Gao C, Ni J, Gao Y, Xie D, Yang L, Yang B, Lu X, Guo Q. Association of current hepatitis B virus infection with mortality in adults with sepsis. Epidemiol Infect 2023; 151:e94. [PMID: 37203184 PMCID: PMC10311682 DOI: 10.1017/s0950268823000729] [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: 03/19/2023] [Accepted: 05/04/2023] [Indexed: 05/20/2023] Open
Abstract
This study aimed to determine the impact of current hepatitis B virus (HBV) infection on patients hospitalised with sepsis. This was a retrospective cohort study. Patients from three medical centres in Suzhou from 10 January 2016 to 23 July 2022 participated in this study. Demographic characteristics and clinical characteristics were collected. A total of 945 adult patients with sepsis were included. The median age was 66.0 years, 68.6% were male, 13.1% presented with current HBV infection, and 34.9% of all patients died. In the multivariable-adjusted Cox model, patients with current HBV infection had significantly higher mortality than those without (hazard ratio (HR) 1.50, 95% confidence interval (CI) 1.11-2.02). A subgroup analysis showed that being infected with HBV significantly increased in-hospital mortality in patients younger than 65 years old (HR 1.74, 95% CI 1.16-2.63), whereas no significant impact was observed in patients ≥65 years. The propensity score-matched case-control analysis showed that the rate of septic shock (91.4% vs. 62.1%, P < 0.001) and in-hospital mortality (48.3% vs. 35.3%, P = 0.045) were much higher in the propensity score-matched HBV infection group compared with the control group. In conclusion, current HBV infection was associated with mortality in adults with sepsis.
Collapse
Affiliation(s)
- Chang Gao
- Department of Emergency and Critical Care Medicine, Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Suzhou, China
- Medical Center of Soochow University, Suzhou, China
- Institute of Critical Care Medicine, Soochow University, Suzhou, China
| | - Jingjing Ni
- Department of Critical Care Medicine, Zhangjiagang Hospital Affiliated to Soochow University, Suzhou, China
| | - Ye Gao
- Department of Critical Care Medicine, Taicang Hospital Affiliated to Soochow University, Suzhou, China
| | - Dan Xie
- Department of Emergency and Critical Care Medicine, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Lijuan Yang
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Bining Yang
- Department of Emergency and Critical Care Medicine, Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Suzhou, China
- Medical Center of Soochow University, Suzhou, China
- Institute of Critical Care Medicine, Soochow University, Suzhou, China
| | - Xiaoting Lu
- Department of Emergency and Critical Care Medicine, Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Suzhou, China
- Medical Center of Soochow University, Suzhou, China
- Institute of Critical Care Medicine, Soochow University, Suzhou, China
| | - Qiang Guo
- Department of Emergency and Critical Care Medicine, Suzhou Dushu Lake Hospital (Dushu Lake Hospital Affiliated to Soochow University), Suzhou, China
- Medical Center of Soochow University, Suzhou, China
- Institute of Critical Care Medicine, Soochow University, Suzhou, China
- Department of Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
167
|
Siggins RW, McTernan PM, Simon L, Souza-Smith FM, Molina PE. Mitochondrial Dysfunction: At the Nexus between Alcohol-Associated Immunometabolic Dysregulation and Tissue Injury. Int J Mol Sci 2023; 24:8650. [PMID: 37239997 PMCID: PMC10218577 DOI: 10.3390/ijms24108650] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Alcohol misuse, directly or indirectly as a result of its metabolism, negatively impacts most tissues, including four with critical roles in energy metabolism regulation: the liver, pancreas, adipose, and skeletal muscle. Mitochondria have long been studied for their biosynthetic roles, such as ATP synthesis and initiation of apoptosis. However, current research has provided evidence that mitochondria participate in myriad cellular processes, including immune activation, nutrient sensing in pancreatic β-cells, and skeletal muscle stem and progenitor cell differentiation. The literature indicates that alcohol impairs mitochondrial respiratory capacity, promoting reactive oxygen species (ROS) generation and disrupting mitochondrial dynamics, leading to dysfunctional mitochondria accumulation. As discussed in this review, mitochondrial dyshomeostasis emerges at a nexus between alcohol-disrupted cellular energy metabolism and tissue injury. Here, we highlight this link and focus on alcohol-mediated disruption of immunometabolism, which refers to two distinct, yet interrelated processes. Extrinsic immunometabolism involves processes whereby immune cells and their products influence cellular and/or tissue metabolism. Intrinsic immunometabolism describes immune cell fuel utilization and bioenergetics that affect intracellular processes. Alcohol-induced mitochondrial dysregulation negatively impacts immunometabolism in immune cells, contributing to tissue injury. This review will present the current state of literature, describing alcohol-mediated metabolic and immunometabolic dysregulation from a mitochondrial perspective.
Collapse
Affiliation(s)
- Robert W. Siggins
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Patrick M. McTernan
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Liz Simon
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Flavia M. Souza-Smith
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
| | - Patricia E. Molina
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (R.W.S.); (P.M.M.); (L.S.); (F.M.S.-S.)
- Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
168
|
Koo SY, Park EJ, Noh HJ, Jo SM, Ko BK, Shin HJ, Lee CW. Ubiquitination Links DNA Damage and Repair Signaling to Cancer Metabolism. Int J Mol Sci 2023; 24:ijms24098441. [PMID: 37176148 PMCID: PMC10179089 DOI: 10.3390/ijms24098441] [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: 04/12/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Changes in the DNA damage response (DDR) and cellular metabolism are two important factors that allow cancer cells to proliferate. DDR is a set of events in which DNA damage is recognized, DNA repair factors are recruited to the site of damage, the lesion is repaired, and cellular responses associated with the damage are processed. In cancer, DDR is commonly dysregulated, and the enzymes associated with DDR are prone to changes in ubiquitination. Additionally, cellular metabolism, especially glycolysis, is upregulated in cancer cells, and enzymes in this metabolic pathway are modulated by ubiquitination. The ubiquitin-proteasome system (UPS), particularly E3 ligases, act as a bridge between cellular metabolism and DDR since they regulate the enzymes associated with the two processes. Hence, the E3 ligases with high substrate specificity are considered potential therapeutic targets for treating cancer. A number of small molecule inhibitors designed to target different components of the UPS have been developed, and several have been tested in clinical trials for human use. In this review, we discuss the role of ubiquitination on overall cellular metabolism and DDR and confirm the link between them through the E3 ligases NEDD4, APC/CCDH1, FBXW7, and Pellino1. In addition, we present an overview of the clinically important small molecule inhibitors and implications for their practical use.
Collapse
Affiliation(s)
- Seo-Young Koo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Eun-Ji Park
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hyun-Ji Noh
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Su-Mi Jo
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Bo-Kyoung Ko
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Hyun-Jin Shin
- Team of Radiation Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul 01812, Republic of Korea
| | - Chang-Woo Lee
- Department of Molecular Cell Biology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
- SKKU Institute for Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
169
|
Cable J, Rathmell JC, Pearce EL, Ho PC, Haigis MC, Mamedov MR, Wu MJ, Kaech SM, Lynch L, Febbraio MA, Bapat SP, Hong HS, Zou W, Belkaid Y, Sullivan ZA, Keller A, Wculek SK, Green DR, Postic C, Amit I, Benitah SA, Jones RG, Reina-Campos M, Torres SV, Beyaz S, Brennan D, O'Neill LAJ, Perry RJ, Brenner D. Immunometabolism at the crossroads of obesity and cancer-a Keystone Symposia report. Ann N Y Acad Sci 2023; 1523:38-50. [PMID: 36960914 PMCID: PMC10367315 DOI: 10.1111/nyas.14976] [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] [Indexed: 03/25/2023]
Abstract
Immunometabolism considers the relationship between metabolism and immunity. Typically, researchers focus on either the metabolic pathways within immune cells that affect their function or the impact of immune cells on systemic metabolism. A more holistic approach that considers both these viewpoints is needed. On September 5-8, 2022, experts in the field of immunometabolism met for the Keystone symposium "Immunometabolism at the Crossroads of Obesity and Cancer" to present recent research across the field of immunometabolism, with the setting of obesity and cancer as an ideal example of the complex interplay between metabolism, immunity, and cancer. Speakers highlighted new insights on the metabolic links between tumor cells and immune cells, with a focus on leveraging unique metabolic vulnerabilities of different cell types in the tumor microenvironment as therapeutic targets and demonstrated the effects of diet, the microbiome, and obesity on immune system function and cancer pathogenesis and therapy. Finally, speakers presented new technologies to interrogate the immune system and uncover novel metabolic pathways important for immunity.
Collapse
Affiliation(s)
| | - Jeffrey C Rathmell
- Vanderbilt-Ingram Cancer Center; Vanderbilt Center for Immunobiology; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Erika L Pearce
- Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, Baltimore, Maryland, USA
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Ping-Chih Ho
- Department of Fundamental Oncology and Ludwig Institute for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Murad R Mamedov
- Gladstone-UCSF Institute of Genomic Immunology and Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Meng-Ju Wu
- Cancer Center, Massachusetts General Hospital; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Lydia Lynch
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Sagar P Bapat
- Diabetes Center and Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Hanna S Hong
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Weiping Zou
- Department of Surgery; Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan Rogel Cancer Center; Department of Pathology; Graduate Program in Immunology; Graduate Program in Cancer Biology, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology, and NIAID Microbiome Program National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Zuri A Sullivan
- Department of Immunobiology, Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Andrea Keller
- Department of Biological Chemistry and Pharmacology, College of Medicine; and Comprehensive Cancer Center, Wexner Medical Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Stefanie K Wculek
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Douglas R Green
- St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Catherine Postic
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Ido Amit
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Salvador Aznar Benitah
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST) and Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | | | - Santiago Valle Torres
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, USA
| | - Donal Brennan
- UCD Gynecological Oncology Group, UCD School of Medicine, Catherine McAuley Research Centre, Mater Misericordiae University Hospital, Belfield, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin, Ireland
| | - Rachel J Perry
- Department of Cellular and Molecular Physiology and Department of Internal Medicine (Endocrinology), Yale University School of Medicine, New Haven, Connecticut, USA
| | - Dirk Brenner
- Experimental and Molecular Immunology, Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
- Immunology and Genetics, Luxembourg Centre for System Biomedicine (LCSB), University of Luxembourg, Belval, Luxembourg
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
170
|
Wang Y, Jin X, Sun Y, Zhao Y, Qu Z, Wang L, Sun L. Muramyl dipeptide CD10 monoclonal antibody immunoconjugates inhibited acute leukemia in nude mice. Biosci Rep 2023; 43:BSR20222668. [PMID: 37039042 PMCID: PMC10126809 DOI: 10.1042/bsr20222668] [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: 01/13/2023] [Revised: 03/11/2023] [Accepted: 04/03/2023] [Indexed: 04/12/2023] Open
Abstract
Minimal residual disease (MRD) is one of the causes of leukemia recurrence. Previously, we developed anti-CD10 mAb conjugated to muramyl dipeptide immunoconjugate (MDP-Ab) for immune enhancement. The present study aimed to investigate anti-leukemia effect of MDP-Ab administered via different methods in leukemia ectopic graft nude mouse model. BALB/c nude mice were injected with Nalm-6 cells subcutaneously to establish leukemia xenografts in nude mice as a model. MDP-Ab or/and human lymphocytes (LYM) was injected into different sites of the nude mice. Immunohistochemistry staining of CDs in the bone marrow, liver and spleen was performed. IFN-γ was detected by ELISA. We detected the metastasis of leukemia cells to the liver, spleen and bone marrow in nude mouse leukemia model. MDP-Ab and LYM inhibited the growth of tumors, and simultaneous injection of MDP-Ab and LYM into the tumor inhibited the growth of tumors. IFN-γ levels in MDP-Ab (ca) + h-LYM (ca) group, MDP-Ab (ca) + h-LYM (ip) group, MDP-Ab (iv) + h-LYM (ip) group and PBS (ca) + h-LYM (ca) group were significantly higher than those in control group, while IFN-γ level in MDP-Ab (ca) + h-LYM (ca) group was the highest. Moreover, MDP-Ab and h-LYM promoted the expression of hCD4 and hCD8, with the highest expression in MDP-Ab (ca) + h-LYM (ca) group. In conclusion, MDP-Ab effectively promoted the production of IFN-γ, enhanced the antitumor immunity of T lymphocytes and inhibited leukemia.
Collapse
Affiliation(s)
- Yilin Wang
- Department of Pediatric Hematology, The Affiliated Hospital of Qingdao University, Shandong 266003, PR China
| | - Xiaofu Jin
- Department of Pediatric Internal Medicine, Jinhua Municipal Central Hospital Medicine Group, Jinhua, Zhejiang 321000, PR China
| | - Yan Sun
- Department of Pediatric Hematology, The Affiliated Hospital of Qingdao University, Shandong 266003, PR China
| | - Yanxia Zhao
- Department of Pediatric Hematology, The Affiliated Hospital of Qingdao University, Shandong 266003, PR China
| | - Zhenghai Qu
- Department of Pediatric, The Affiliated Hospital of Qingdao University, Shandong 266003, PR China
| | - Lingzhen Wang
- Department of Pediatric Hematology, The Affiliated Hospital of Qingdao University, Shandong 266003, PR China
| | - Lirong Sun
- Department of Pediatric Hematology, The Affiliated Hospital of Qingdao University, Shandong 266003, PR China
| |
Collapse
|
171
|
Lin K, Zhou Y, Lin Y, Feng Y, Chen Y, Cai L. Senescence-Related lncRNA Signature Predicts Prognosis, Response to Immunotherapy and Chemotherapy in Skin Cutaneous Melanoma. Biomolecules 2023; 13:biom13040661. [PMID: 37189408 DOI: 10.3390/biom13040661] [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: 11/25/2022] [Revised: 02/15/2023] [Accepted: 03/27/2023] [Indexed: 05/17/2023] Open
Abstract
Skin cutaneous melanoma (SKCM) is a highly malignant and aggressive cancer. Previous studies have shown that cellular senescence is a promising therapeutic strategy to limit melanoma cell progression. However, models to predict the prognosis of melanoma based on senescence-related lncRNAs and the efficacy of immune checkpoint therapy remain undefined. In this study, we developed a predictive signature consisting of four senescence-related lncRNAs (AC009495.2, U62317.1, AATBC, MIR205HG), and we then classified patients into high- and low-risk groups. GSEA (Gene set enrichment analysis) showed different activation of immune-related pathways in two groups. In addition, there were significant differences between the scores of tumor immune microenvironment, tumor burden mutation, immune checkpoint expression, and chemotherapeutic drug sensitivity between the two groups of patients. It provides new insights to guide more personalized treatment for patients with SKCM.
Collapse
Affiliation(s)
- Kefan Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yingtong Zhou
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yanling Lin
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuanyuan Feng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Yuting Chen
- First Clinical Medical College, Southern Medical University, Guangzhou 510515, China
| | - Longmei Cai
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
172
|
Zhao LM, Shi AD, Yang Y, Liu ZL, Hu XQ, Shu LZ, Tang YC, Zhang ZL. Advances in molecular and cell therapy for immunotherapy of cholangiocarcinoma. Front Oncol 2023; 13:1140103. [PMID: 37064120 PMCID: PMC10090456 DOI: 10.3389/fonc.2023.1140103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a highly malignant tumor of the hepatobiliary system that has failed to respond to many traditional therapies to a certain extent, including surgery, chemotherapy and radiotherapy. In recent years, the new therapeutic schemes based on immunology have fundamentally changed the systemic treatment of various malignant tumors to a certain extent. In view of the immunogenicity of CCA, during the occurrence and development of CCA, some immunosuppressive substances are released from cells and immunosuppressive microenvironment is formed to promote the escape immune response of its own cells, thus enhancing the malignancy of the tumor and reducing the sensitivity of the tumor to drugs. Some immunotherapy regimens for cholangiocarcinoma have produced good clinical effects. Immunotherapy has more precise characteristics and less adverse reactions compared with traditional treatment approaches. However, due to the unique immune characteristics of CCA, some patients with CCA may not benefit in the long term or not benefit at all after current immunotherapy. At present, the immunotherapy of CCA that have been clinically studied mainly include molecular therapy and cell therapy. In this article, we generalized and summarized the current status of immunotherapy strategies including molecular therapy and cell therapy in CCA in clinical studies, and we outlined our understanding of how to enhance the clinical application of these immunotherapy strategies.
Collapse
Affiliation(s)
- Li-ming Zhao
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - An-da Shi
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yan Yang
- Department of General Surgery, Shanxian Central Hospital, Heze, China
| | - Zeng-li Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- Department of General Surgery, Qilu Hospital (Qingdao), Shandong University, Jinan, China
| | - Xiao-Qiang Hu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Li-Zhuang Shu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yong-chang Tang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- *Correspondence: Yong-chang Tang, ; Zong-li Zhang,
| | - Zong-li Zhang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- *Correspondence: Yong-chang Tang, ; Zong-li Zhang,
| |
Collapse
|
173
|
Lebedev T, Kousar R, Patrick B, Usama M, Lee MK, Tan M, Li XG. Targeting ARID1A-Deficient Cancers: An Immune-Metabolic Perspective. Cells 2023; 12:cells12060952. [PMID: 36980292 PMCID: PMC10047504 DOI: 10.3390/cells12060952] [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] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Epigenetic remodeling and metabolic reprogramming, two well-known cancer hallmarks, are highly intertwined. In addition to their abilities to confer cancer cell growth advantage, these alterations play a critical role in dynamically shaping the tumor microenvironment and antitumor immunity. Recent studies point toward the interplay between epigenetic regulation and metabolic rewiring as a potentially targetable Achilles' heel in cancer. In this review, we explore the key metabolic mechanisms that underpin the immunomodulatory role of AT-rich interaction domain 1A (ARID1A), the most frequently mutated epigenetic regulator across human cancers. We will summarize the recent advances in targeting ARID1A-deficient cancers by harnessing immune-metabolic vulnerability elicited by ARID1A deficiency to stimulate antitumor immune response, and ultimately, to improve patient outcome.
Collapse
Affiliation(s)
- Timofey Lebedev
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Rubina Kousar
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 110122, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 110122, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 110122, Taiwan
| | - Bbumba Patrick
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 110122, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 110122, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 110122, Taiwan
| | - Muhammad Usama
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 110122, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 110122, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 110122, Taiwan
| | - Meng-Kuei Lee
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 110122, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 110122, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 110122, Taiwan
| | - Ming Tan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 110122, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 110122, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 110122, Taiwan
| | - Xing-Guo Li
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 110122, Taiwan
- Research Center for Cancer Biology, China Medical University, Taichung 110122, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, China Medical University, Taichung 110122, Taiwan
| |
Collapse
|
174
|
Shi X, Ding J, Zheng Y, Wang J, Sobhani N, Neeli P, Wang G, Zheng J, Chai D. HMGB1/GPC3 dual targeting vaccine induces dendritic cells-mediated CD8 +T cell immune response and elicits potential therapeutic effect in hepatocellular carcinoma. iScience 2023; 26:106143. [PMID: 36879804 PMCID: PMC9984564 DOI: 10.1016/j.isci.2023.106143] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/15/2022] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a fatal malignant tumor, but effective clinical interventions are limited. PLGA/PEI-mediated DNA vaccine encoding the dual targets of high-mobility group box 1 (HMGB1) or GPC3 was developed for HCC treatment. Compared with PLGA/PEI-GPC3 immunization, PLGA/PEI-HMGB1/GPC3 co-immunization significantly inhibited the subcutaneous tumor growth, while increasing the infiltration of CD8+T cells and DCs. Furthermore, the PLGA/PEI-HMGB1/GPC3 vaccine induced a strong CTL effect and promoted functional CD8+T cell proliferation. Intriguingly, the depletion assay proved that the therapeutic effect PLGA/PEI-HMGB1/GPC3 vaccine was dependent on antigen-specific CD8+T cell immune responses. In the rechallenge experiment, PLGA/PEI-HMGB1/GPC3 vaccine provided a long-lasting resistance to the growth of the contralateral tumor by inducing the memory CD8+T cell responses. Collectively, PLGA/PEI-HMGB1/GPC3 vaccine could induce a strong and long-lasting CTL effect and inhibit the tumor progression or re-attack. Therefore, the combined co-immunization of PLGA/PEI-HMGB1/GPC3 might be served as an effective anti-tumor strategy against HCC.
Collapse
Affiliation(s)
- Xiaoqing Shi
- Department of General Surgery, Lianyungang Hospital of Traditional Chinese Medicine, Lianyungang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Lianyungang, Jiangsu 222004, China
| | - Jiage Ding
- Department of Oncology, Xuzhou Central Hospital, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221009, China.,Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Yanyan Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Jiawei Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Navid Sobhani
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Praveen Neeli
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China.,Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
175
|
Li T, Chen S, Zhang Y, Zhao Q, Ma K, Jiang X, Xiang R, Zhai F, Ling G. Ensemble learning-based gene signature and risk model for predicting prognosis of triple-negative breast cancer. Funct Integr Genomics 2023; 23:81. [PMID: 36917262 DOI: 10.1007/s10142-023-01009-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/15/2023]
Abstract
Although medical science has been fully developed, due to the high heterogeneity of triple-negative breast cancer (TNBC), it is still difficult to use reasonable and precise treatment. In this study, based on local optimization-feature screening and genomics screening strategy, we screened 25 feature genes. In multiple machine learning algorithms, feature genes have excellent discriminative diagnostic performance among samples composed of multiple large datasets. After screening at the single-cell level, we identified genes expressed substantially in myeloid cells (MCGs) that have a potential association with TNBC. Based on MCGs, we distinguished two types of TNBC patients who showed considerable differences in survival status and immune-related characteristics. Immune-related gene risk scores (IRGRS) were established, and their validity was verified using validation cohorts. A total of 25 feature genes were obtained, among which CXCL9, CXCL10, CCL7, SPHK1, and TREM1 were identified as the result after single-cell level analysis and screening. According to these entries, the cohort was divided into MCA and MCB subtypes, and the two subtypes had significant differences in survival status and tumor-immune microenvironment. After Lasso-Cox screening, IDO1, GNLY, IRF1, CTLA4, and CXCR6 were selected for constructing IRGRS. There were significant differences in drug sensitivity and immunotherapy sensitivity among high-IRGRS and low-IRGRS groups. We revealed the dynamic relationship between TNBC and TIME, identified a potential biomarker called Granulysin (GNLY) related to immunity, and developed a multi-process machine learning package called "MPMLearning 1.0" in Python.
Collapse
Affiliation(s)
- Tiancheng Li
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Siqi Chen
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yuqi Zhang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Qianqian Zhao
- School of Life Sciences and Biopharmaceutical Science, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Kai Ma
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Xiwei Jiang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Rongwu Xiang
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China
- Liaoning Medical Big Data and Artificial Intelligence Engineering Technology Research Center, Shenyang, 110016, China
| | - Fei Zhai
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Guixia Ling
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
176
|
Ning L, Shishi Z, Bo W, Huiqing L. Targeting immunometabolism against acute lung injury. Clin Immunol 2023; 249:109289. [PMID: 36918041 PMCID: PMC10008193 DOI: 10.1016/j.clim.2023.109289] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023]
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening conditions triggered by multiple intra- and extra-pulmonary injury factors, characterized by complicated molecular mechanisms and high mortality. Great strides have been made in the field of immunometabolism to clarify the interplay between intracellular metabolism and immune function in the past few years. Emerging evidence unveils the crucial roles of immunometabolism in inflammatory response and ALI. During ALI, both macrophages and lymphocytes undergo robust metabolic reprogramming and discrete epigenetic changes after activated. Apart from providing ATP and biosynthetic precursors, these metabolic cellular reactions and processes in lung also regulate inflammation and immunity.In fact, metabolic reprogramming involving glucose metabolism and fatty acidoxidation (FAO) acts as a double-edged sword in inflammatory response, which not only drives inflammasome activation but also elicits anti-inflammatory response. Additionally, the features and roles of metabolic reprogramming in different immune cells are not exactly the same. Here, we outline the evidence implicating how adverse factors shape immunometabolism in differentiation types of immune cells during ALI and summarize key proteins associated with energy expenditure and metabolic reprogramming. Finally, novel therapeutic targets in metabolic intermediates and enzymes together with current challenges in immunometabolism against ALI were discussed.
Collapse
Affiliation(s)
- Li Ning
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Zou Shishi
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Wang Bo
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| | - Lin Huiqing
- Department of Thoracic Surgery, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China.
| |
Collapse
|
177
|
Guo YC, Fu ZY, Ding ZJ. Immune infiltration associated C1q acts as a novel prognostic biomarker of cutaneous melanoma. Medicine (Baltimore) 2023; 102:e33088. [PMID: 36897727 PMCID: PMC9997796 DOI: 10.1097/md.0000000000033088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 02/03/2023] [Indexed: 03/11/2023] Open
Abstract
C1q (complement C1q A chain, complement C1q B chain, and complement C1q C chain) is a recognized component of the classical complement pathway that influences the prognosis of various cancers. However, the effects of C1q on cutaneous melanoma (SKCM) outcomes and immune infiltration remain unknown. Gene expression profiling interactive analysis 2 and the human protein atlas were used to evaluate differential expression of C1q mRNA and protein. The relationship between C1q expression and clinicopathological features was also examined. The genetic alterations of C1q and their impact on survival were analyzed using the cbioportal database. The Kaplan-Meier approach was used to assess the significance of C1q in individuals with SKCM. The cluster profiler R package and the cancer single-cell state atlas database were used to investigate the function and mechanism of C1q in SKCM. The relationship between C1q and immune cell infiltration was estimated using single-sample gene set enrichment analysis. C1q expression was increased, and predicted a favorable prognosis. High C1q expression correlated with clinicopathological T stage, pathological stage, overall survival, and disease specific survival events. Moreover, C1q genetic alterations range from 2.7% to 4%, with no impact on prognosis. According to the enrichment analysis, C1q and immune-related pathways were closely connected. The link between complement C1q B chain and the functional state of inflammation was determined using the cancer single-cell state atlas database. In particular, C1q expression was significantly associated with infiltration of most immune cells and checkpoints PDCD1, CD274, and HAVCR2. The results of this study suggest that C1q is associated with prognosis and immune cell infiltration, supporting its value as a diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Yi-Cheng Guo
- Dermatology Hospital of Jiangxi Province, Nanchang, China
- Jiangxi Province Clinical Research Center for Skin Diseases, Nanchang, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Nanchang, Jiangxi, China
| | - Zhi-Yuan Fu
- Dermatology Hospital of Jiangxi Province, Nanchang, China
| | - Zhi-Jun Ding
- Jiangxi Province Clinical Research Center for Skin Diseases, Nanchang, China
| |
Collapse
|
178
|
Zhang S, Tu Q, Qian X, Wang J, Ma C, Yang L, Liu Y, Wu R, Liu M. Deficiency of Kif15 gene inhibits tumor growth due to host CD8 +T lymphocytes increase. Biochem Biophys Res Commun 2023; 655:110-117. [PMID: 36934586 DOI: 10.1016/j.bbrc.2023.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Abstract
Kif15, also name kinesin-12, is a microtubule (MT) associate protein, which functions as a regulator of MT-dependent transport or spindle organization. Previous studies reported Kif15 increases in many tumors, however the effect of host Kif15 gene lack on tumor growth is not investigated. In this study, CRISPR/Cas9 mediated Kif15 gene knockout (Kif15-/-) mice were established and HE (Hematoxylin-Eosin) assay revealed no significant differences of morphology in most adult tissues (heart, liver, lung, kidney, and brain) except a retarded development of spleen in adult Kif15-/- mice. RNA sequence analysis of adult spleen tissues of Kif15-/- and Kif15+/+ mice was performed, and the results revealed that a total of 438 mRNAs were significantly differentially expressed in Kif15 knockout spleen, showing the top biological process was immune system process. FCM (Flow Cytometry) assay showed the percentage of CD8+ T lymphocytes notably increased in spleens of 9 w and 12 w old Kif15-/- mice. The CD8+ T lymphocytes are cytotoxic effector cells fighting against tumor. We thus detected the tumor growth in Kif15-/- mice using the melanoma cells inoculated subcutaneously. The tumor size significantly reduced in Kif15-/- mice. We finally detected whether Kif15 dysfunction affects the phagocytic function of macrophages on tumor cells, and the result showed Kif15 inhibitor treated macrophages significantly promoted the phagocytosis in vitro. In summary, this study revealed that the tumor-bearing mice of Kif15 gene deficiency notably inhibited tumor growth due to innate immune activation, which was the first report of the relation of Kif15 on the immunoreactivity.
Collapse
Affiliation(s)
- Siming Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China; Cancer Research Center Nantong, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Qifeng Tu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Xiaowei Qian
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Junpei Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Chao Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Liu Yang
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China
| | - Ronghua Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China.
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, China.
| |
Collapse
|
179
|
Liu Y, Zhang M, Wang X, Yang F, Cao Z, Wang L, Liu J. Dressing Bacteria With a Hybrid Immunoactive Nanosurface to Elicit Dual Anticancer and Antiviral Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2210949. [PMID: 36564893 DOI: 10.1002/adma.202210949] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Indexed: 06/17/2023]
Abstract
Approaches capable of simultaneously treating cancer and protecting susceptible patients from lethal infections such as coronavirus disease 2019, are highly desirable but prove to be difficult. Here, dressing bacteria with a hybrid immunoactive nanosurface is reported to elicit dual anticancer and antiviral immunity. A combination of a checkpoint blocking antibody and a virus-specific antigen is covalently conjugated to polydopamine nanoparticles, which can be anchored onto bacterial surface, by a one-step in situ polymerization of dopamine under a cell-friendly condition. By virtue of the ability to colonize and penetrate deep tumor tissue, dressed bacteria enable sustained release and expanded exposure of carried immunoactivators to stimulate immune cells. In addition to a carrier role, bacteria are able to further provoke innate immunity due to the native immunogenicity of the pathogen-associated molecular patterns. Immunization with dressed bacteria promotes the maturation, and activation of antigen-presenting cells, which induces robust humoral and cellular immune responses in tumor-bearing mice. As evidenced by efficient production of viral-antigen-specific immunoglobulin G antibody in serum and significantly suppressed tumor growth in different models, dressing bacteria with a hybrid immunoactive nanosurface paves an avenue to prepare next-generation therapeutics for synergistic treatment and prevention.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mengmeng Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Xinyue Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fengmin Yang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zhenping Cao
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Jinyao Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| |
Collapse
|
180
|
Zhang J, Huang L, Ge G, Hu K. Emerging Epigenetic-Based Nanotechnology for Cancer Therapy: Modulating the Tumor Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206169. [PMID: 36599655 PMCID: PMC9982594 DOI: 10.1002/advs.202206169] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 12/05/2022] [Indexed: 06/02/2023]
Abstract
Dysregulated epigenetic modifications dynamically drive the abnormal transcription process to affect the tumor microenvironment; thus, promoting cancer progression, drug resistance, and metastasis. Nowadays, therapies targeting epigenetic dysregulation of tumor cells and immune cells in the tumor microenvironment appear to be promising adjuncts to other cancer therapies. However, the clinical results of combination therapies containing epigenetic agents are disappointing due to systemic toxicities and limited curative effects. Here, the role of epigenetic processes, including DNA methylation, post-translational modification of histones, and noncoding RNAs is discussed, followed by detailed descriptions of epigenetic regulation of the tumor microenvironment, as well as the application of epigenetic modulators in antitumor therapy, with an emphasis on the epigenetic-based advanced drug delivery system in targeting the tumor microenvironment.
Collapse
Affiliation(s)
- Jiaxin Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular PharmaceuticsEshelman School of PharmacyUniversity of North Carolina at Chapel HillChapel HillNC27599USA
| | - Guangbo Ge
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| | - Kaili Hu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghai201203China
| |
Collapse
|
181
|
Li Y, Xin W, Liu F, Li F, Yang C, Liu C, Liu J. Dysfunction of the ST7-AS1/miR-301b-3p/BTG1 ceRNA network promotes immune escape of triple-negative breast cancer. Int Immunopharmacol 2023. [DOI: 10.1016/j.intimp.2023.109805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
182
|
Han Y, Fan X, Fan L, Wu Y, Zhou Z, Wang G, Guo L, Gao W, Chen Y, Gao Q. Liujunzi decoction exerts potent antitumor activity in oesophageal squamous cell carcinoma by inhibiting miR-34a/STAT3/IL-6R feedback loop, and modifies antitumor immunity. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 111:154672. [PMID: 36701994 DOI: 10.1016/j.phymed.2023.154672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 12/30/2022] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Liujunzi decoction (LJZD), a traditional herbal formula and one of the most commonly used adjuvant medications for the treatment of oesophageal squamous cell carcinoma (ESCC), exerts good antitumor and immunomodulatory activity. However, its specific mechanism of action remains largely unclear. PURPOSE In order to examine the potential primary and adjuvant antitumor mechanisms of LJZD, both in vitro and in vivo. METHODS IL-6 and miR-34a inhibitors were used to activate the miR-34a/STAT3/IL-6R feedback loop to observe the effects of LJZD. A humanised mouse model with a functional human immune system was constructed to evaluate the antitumor efficacy of LJZD in vivo on xenograft tumours, which was compared to that of the positive control drug anti-PD-1 monoclonal antibodies (mAb). Finally, a co-culture system of peripheral blood mononuclear and tumour cells in vitro was used to analyse the cytotoxic activity of LJZD on T cells. RESULTS LJZD significantly interfered with IL-6-induced activation of the miR-34a/STAT3/IL-6R feedback loop in ESCC by restoring the expression of the tumour suppressor miR-34a, and inhibited the proliferation of EC109 oesophageal cancer cells in a dose-dependant manner. Furthermore, LJZD effectively suppressed oesophageal tumour growth in vivo and alleviated organ injury and visceral index. Furthermore, LJZD boosted antitumor immunity by increasing IFN-γ expression and CD8+tumour-infiltrating lymphocytes (TILs) infiltration in the peripheral blood and tumour tissues, respectively, which may be related to a decrease in PD-1, but not PD-L1 expression. Finally, we confirmed that LJZD strengthens the killing ability of T cells by suppressing PD-1 expression in a co-culture system in vitro. CONCLUSION LJZD exerts excellent antitumor effect by interfering with the miR-34a/STAT3/IL-6R feedback loop and augmenting antitumor immune responses. Which provides new insights into mechanisms for LJZD and sheds light on the multifaceted role of phytomedicine in cancer.
Collapse
Affiliation(s)
- Yicun Han
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Xiuqi Fan
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Liyan Fan
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Yaosong Wu
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Zhexu Zhou
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Ge Wang
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Lanwei Guo
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China
| | - Wendong Gao
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China
| | - Yulong Chen
- Henan International Joint Laboratory of TCM Syndrome and Prescription in Signaling, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan Province 450046, China.
| | - Qilong Gao
- The Tumor Hospital Affiliated to Zhengzhou University, Zhengzhou, Henan Province 450008, China.
| |
Collapse
|
183
|
Metabolic interaction: tumor-derived lactate inhibiting CD8 + T cell cytotoxicity in a novel route. Signal Transduct Target Ther 2023; 8:52. [PMID: 36737430 PMCID: PMC9898493 DOI: 10.1038/s41392-023-01320-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/12/2022] [Accepted: 12/22/2022] [Indexed: 02/05/2023] Open
|
184
|
Trefny MP, Kirchhammer N, Auf der Maur P, Natoli M, Schmid D, Germann M, Fernandez Rodriguez L, Herzig P, Lötscher J, Akrami M, Stinchcombe JC, Stanczak MA, Zingg A, Buchi M, Roux J, Marone R, Don L, Lardinois D, Wiese M, Jeker LT, Bentires-Alj M, Rossy J, Thommen DS, Griffiths GM, Läubli H, Hess C, Zippelius A. Deletion of SNX9 alleviates CD8 T cell exhaustion for effective cellular cancer immunotherapy. Nat Commun 2023; 14:86. [PMID: 36732507 PMCID: PMC9895440 DOI: 10.1038/s41467-022-35583-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 12/12/2022] [Indexed: 02/04/2023] Open
Abstract
Tumor-specific T cells are frequently exhausted by chronic antigenic stimulation. We here report on a human antigen-specific ex vivo model to explore new therapeutic options for T cell immunotherapies. T cells generated with this model resemble tumor-infiltrating exhausted T cells on a phenotypic and transcriptional level. Using a targeted pooled CRISPR-Cas9 screen and individual gene knockout validation experiments, we uncover sorting nexin-9 (SNX9) as a mediator of T cell exhaustion. Upon TCR/CD28 stimulation, deletion of SNX9 in CD8 T cells decreases PLCγ1, Ca2+, and NFATc2-mediated T cell signaling and reduces expression of NR4A1/3 and TOX. SNX9 knockout enhances memory differentiation and IFNγ secretion of adoptively transferred T cells and results in improved anti-tumor efficacy of human chimeric antigen receptor T cells in vivo. Our findings highlight that targeting SNX9 is a strategy to prevent T cell exhaustion and enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Marcel P Trefny
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.
| | - Nicole Kirchhammer
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Priska Auf der Maur
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Marina Natoli
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Dominic Schmid
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Markus Germann
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Laura Fernandez Rodriguez
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Jonas Lötscher
- Laboratory of Immunobiology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Maryam Akrami
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Jane C Stinchcombe
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Michal A Stanczak
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Andreas Zingg
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Melanie Buchi
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Julien Roux
- Bioinformatics Core Facility, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Romina Marone
- Laboratory of Molecular Immune Regulation, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.,Transplantation Immunology & Nephrology, Basel University Hospital, Basel, Switzerland
| | - Leyla Don
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Didier Lardinois
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Mark Wiese
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Lukas T Jeker
- Laboratory of Molecular Immune Regulation, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.,Transplantation Immunology & Nephrology, Basel University Hospital, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Laboratory of Tumor Heterogeneity, Metastasis and Resistance, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland
| | - Jérémie Rossy
- Biotechnology Institute Thurgau, University of Konstanz, Kreuzlingen, Switzerland
| | - Daniela S Thommen
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.,Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gillian M Griffiths
- Cambridge Institute for Medical Research, Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Heinz Läubli
- Laboratory of Cancer Immunotherapy, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.,Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Christoph Hess
- Laboratory of Immunobiology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland.,Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, CB2 0AW, UK
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital of Basel, Basel, Switzerland. .,Medical Oncology, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
185
|
Amitrano AM, Kim M. Metabolic Challenges in Anticancer CD8 T Cell Functions. Immune Netw 2023; 23:e9. [PMID: 36911801 PMCID: PMC9995993 DOI: 10.4110/in.2023.23.e9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/13/2022] [Accepted: 12/21/2022] [Indexed: 03/07/2023] Open
Abstract
Cancer immunotherapies continue to face numerous obstacles in the successful treatment of solid malignancies. While immunotherapy has emerged as an extremely effective treatment option for hematologic malignancies, it is largely ineffective against solid tumors due in part to metabolic challenges present in the tumor microenvironment (TME). Tumor-infiltrating CD8+ T cells face fierce competition with cancer cells for limited nutrients. The strong metabolic suppression in the TME often leads to impaired T-cell recruitment to the tumor site and hyporesponsive effector functions via T-cell exhaustion. Growing evidence suggests that mitochondria play a key role in CD8+ T-cell activation, migration, effector functions, and persistence in tumors. Therefore, targeting the mitochondrial metabolism of adoptively transferred T cells has the potential to greatly improve the effectiveness of cancer immunotherapies in treating solid malignancies.
Collapse
Affiliation(s)
- Andrea M. Amitrano
- Department of Pathology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
186
|
Jin K, Qian C, Lin J, Liu B. Cyclooxygenase-2-Prostaglandin E2 pathway: A key player in tumor-associated immune cells. Front Oncol 2023; 13:1099811. [PMID: 36776289 PMCID: PMC9911818 DOI: 10.3389/fonc.2023.1099811] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/12/2023] [Indexed: 01/29/2023] Open
Abstract
Cyclooxygenases-2 (COX-2) and Prostaglandin E2 (PGE2), which are important in chronic inflammatory diseases, can increase tumor incidence and promote tumor growth and metastasis. PGE2 binds to various prostaglandin E receptors to activate specific downstream signaling pathways such as PKA pathway, β-catenin pathway, NF-κB pathway and PI3K/AKT pathway, all of which play important roles in biological and pathological behavior. Nonsteroidal anti-inflammatory drugs (NSAIDs), which play as COX-2 inhibitors, and EP antagonists are important in anti-tumor immune evasion. The COX-2-PGE2 pathway promotes tumor immune evasion by regulating myeloid-derived suppressor cells, lymphocytes (CD8+ T cells, CD4+ T cells and natural killer cells), and antigen presenting cells (macrophages and dendritic cells). Based on conventional treatment, the addition of COX-2 inhibitors or EP antagonists may enhance immunotherapy response in anti-tumor immune escape. However, there are still a lot of challenges in cancer immunotherapy. In this review, we focus on how the COX-2-PGE2 pathway affects tumor-associated immune cells.
Collapse
Affiliation(s)
- Kaipeng Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Chao Qian
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jinti Lin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China,*Correspondence: Bing Liu, ; Jinti Lin,
| | - Bing Liu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China,Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China,Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China,*Correspondence: Bing Liu, ; Jinti Lin,
| |
Collapse
|
187
|
Kim J, Nguyen T, Cifello J, Ahmad R, Zhang Y, Yang Q, Lee JE, Li X, Kai Y, De S, Peng W, Ge K, Weng NP. Lysine methyltransferase Kmt2d regulates naive CD8 + T cell activation-induced survival. Front Immunol 2023; 13:1095140. [PMID: 36741385 PMCID: PMC9892454 DOI: 10.3389/fimmu.2022.1095140] [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: 11/10/2022] [Accepted: 12/30/2022] [Indexed: 01/20/2023] Open
Abstract
Lysine specific methyltransferase 2D (Kmt2d) catalyzes the mono-methylation of histone 3 lysine 4 (H3K4me1) and plays a critical role in regulatory T cell generation via modulating Foxp3 gene expression. Here we report a role of Kmt2d in naïve CD8+ T cell generation and survival. In the absence of Kmt2d, the number of CD8+ T cells, particularly naïve CD8+ T cells (CD62Lhi/CD44lo), in spleen was greatly decreased and in vitro activation-related death significantly increased from Kmt2d fl/flCD4cre+ (KO) compared to Kmt2d fl/flCD4cre- (WT) mice. Furthermore, analyses by ChIPseq, RNAseq, and scRNAseq showed reduced H3K4me1 levels in enhancers and reduced expression of apoptosis-related genes in activated naïve CD8+ T cells in the absence of Kmt2d. Finally, we confirmed the activation-induced death of antigen-specific naïve CD8+ T cells in vivo in Kmt2d KO mice upon challenge with Listeria monocytogenes infection. These findings reveal that Kmt2d regulates activation-induced naïve CD8+ T cell survival via modulating H3K4me1 levels in enhancer regions of apoptosis and immune function-related genes.
Collapse
Affiliation(s)
- Jaekwan Kim
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Thomas Nguyen
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Jeffrey Cifello
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Raheel Ahmad
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Yongqing Zhang
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Qian Yang
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Ji-Eun Lee
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Xiang Li
- Department of Physics, George Washington University, Washington DC, WA, United States
| | - Yan Kai
- Department of Physics, George Washington University, Washington DC, WA, United States
| | - Supriyo De
- Laboratory of Genetics and Genomics, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Weiqun Peng
- Department of Physics, George Washington University, Washington DC, WA, United States
| | - Kai Ge
- Laboratory of Endocrinology and Receptor Biology, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Nan-ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States,*Correspondence: Nan-ping Weng,
| |
Collapse
|
188
|
Tian Z, Khan AI, Rehman AU, Deng T, Ma C, Wang L. Virulence factors and mechanisms of paediatric pneumonia caused by Enterococcus faecalis. Gut Pathog 2023; 15:2. [PMID: 36624474 PMCID: PMC9830894 DOI: 10.1186/s13099-022-00522-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 12/09/2022] [Indexed: 01/11/2023] Open
Abstract
Paediatric pneumonia is a respiratory infection that affects infants and young children under the age of 3. This disease is the leading cause of infant and child mortality in developing countries because of the weak immune system of young children. The difficulty and length of time required to identify the pathogen and causative agent are the main reasons for this high mortality rate. In addition, the identification of certain causative agents is particularly important for the treatment of paediatric pneumonia. In this study, we explored the possible mechanisms by which pathogenic Enterococcus faecalis induced pneumonia in vivo. The potential virulence factors of bacteria isolated from the intestines of paediatric pneumonia patients were determined. Taken together, the results suggested that lysophosphatidic acid (LTA) from pathogenic E. faecalis decreases the expression of platelet-activating factor receptor (PAFR), which in turn disrupts the function of intestinal tight junctions (Occ and Ccldn1), leading to the entry of LE-LTA into the bloodstream because of the disruption of the intestinal barrier. Although LTA can enter circulation, it cannot directly infiltrate the lungs, which indicates that lung inflammation in mice is not caused by the direct entry of LE-LTA into the lungs. We further found that LTA activates immune cells, such as CD8 + T cells and type 2 innate lymphocytes, in vivo. Interleukin-6 and interleukin-17 can produce large amounts of inflammatory factors and thus promote the development of pneumonia. In conclusion, our findings demonstrate that the LTA of pathogenic E. faecalis in the intestine is a virulence factor that can cause paediatric pneumonia. This study found that intestinal bacterial virulence factors can induce immune responses in the lungs and blood. These findings could provide further insight into the mechanism of infectious diseases in the lung that are caused by bacteria in the intestine.
Collapse
Affiliation(s)
- Zhiying Tian
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Asif Iqbal Khan
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Ata Ur Rehman
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Ting Deng
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Chao Ma
- Laboratory of Biochemistry and Molecular Biology, Department of Biotechnology, College of Basic Medicine, Dalian Medical University, Dalian, China
| | - Liang Wang
- National Joint Engineering Laboratory, Regenerative Medicine Centre, Stem Cell Clinical Research Centre, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
189
|
Feng Y, Tang F, Li S, Wu D, Liu Q, Li H, Zhang X, Liu Z, Zhang L, Feng H. Mannose-modified erythrocyte membrane-encapsulated chitovanic nanoparticles as a DNA vaccine carrier against reticuloendothelial tissue hyperplasia virus. Front Immunol 2023; 13:1066268. [PMID: 36776397 PMCID: PMC9910308 DOI: 10.3389/fimmu.2022.1066268] [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: 10/10/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Introduction The erythrocyte membranes used in nanovaccines include high membrane stability, long circulation life, adaptability and extremely good bio compatibility. Nanoparticles encapsulated by erythrocyte membranes are widely used as ideal drug delivery vehicles because of their high drug loading, long circulation time, and excellent biocompatibility. The mannose modification of delivery materials can help target mannose receptors (MRs) to deliver antigens to antigen-presenting cells (APCs). Methods In this study, the antigen gene gp90 of avian reticuloendotheliosis virus (REV) was encapsulated with carboxymethyl chitosan (CS) to obtain CSgp90 nanoparticles, which were coated with mannose-modied fowl erythrocyte membranes to yield CS-gp90@M-M nanoparticles. The physicochemical characterization and immune response of the CS-gp90@M-M nanoparticles were investigated in vitro and in vivo. Results CS-gp90@M-M nanoparticles were rapidly phagocytized in vitro by macrophages to induce the production of cytokines and nitric oxide. In vivo, CS-gp90@M-M nanoparticles increased cytokine levels, the CD4+/8+ ratio, REV-specific antibodies in the peripheral blood of chicks, and the mRNA levels of immune-related genes in the spleen and bursa of immunized chicks. CS-gp90@M-M nanoparticles could be targeted to lymphoid organs to prolong the retention time of the nanoparticles at the injection site and lymphatic organs, leading to a strong, sustained immune response. Moreover, the CS-gp90@M-M nano-vaccine showed a lasting immunoprotective effect and improved the body weight of chicks after the challenge. Conclusion Overall, CS-gp90@M-M nanoparticles can be used in vaccine designs as an effective delivery carrier with immune response-enhancing effects.
Collapse
Affiliation(s)
- Yangyang Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Feng Tang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Sheng Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Daiyan Wu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Qianqian Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Hangyu Li
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Xinnan Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Ziwei Liu
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Linzi Zhang
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China
| | - Haibo Feng
- College of Animal Husbandry and Veterinary Medicine, Southwest Minzu University, Chengdu, China,Key Laboratory of Ministry of Education and Sichuan Province for Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Southwest-Minzu University, Chengdu, China,*Correspondence: Haibo Feng,
| |
Collapse
|
190
|
Han S, Georgiev P, Ringel AE, Sharpe AH, Haigis MC. Age-associated remodeling of T cell immunity and metabolism. Cell Metab 2023; 35:36-55. [PMID: 36473467 PMCID: PMC10799654 DOI: 10.1016/j.cmet.2022.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 10/14/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
Aging results in remodeling of T cell immunity and is associated with poor clinical outcomes in age-related diseases such as cancer. Among the hallmarks of aging, changes in host and cellular metabolism critically affect the development, maintenance, and function of T cells. Although metabolic perturbations impact anti-tumor T cell responses, the link between age-associated metabolic dysfunction and anti-tumor immunity remains unclear. In this review, we summarize recent advances in our understanding of aged T cell metabolism, with a focus on the bioenergetic and immunologic features of T cell subsets unique to the aging process. We also survey insights into mechanisms of metabolic T cell dysfunction in aging and discuss the impacts of aging on the efficacy of cancer immunotherapy. As the average life expectancy continues to increase, understanding the interplay between age-related metabolic reprogramming and maladaptive T cell immunity will be instrumental for the development of therapeutic strategies for older patients.
Collapse
Affiliation(s)
- SeongJun Han
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Peter Georgiev
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alison E Ringel
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arlene H Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Marcia C Haigis
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
191
|
Chang L, Zhang C, Lu J, Shen J, Hamal K, Liu D. Clinical and Pathological Features of Hydroa Vacciniforme-Like Lymphoproliferative Disorder Along with Risk Factors Indicating Poor Prognosis. Infect Drug Resist 2023; 16:1545-1559. [PMID: 36960391 PMCID: PMC10027612 DOI: 10.2147/idr.s402040] [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: 01/07/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Purpose To examine the clinical and pathological features, laboratory markers, therapeutic options and risk factors indicating poor prognosis of hydroa vacciniforme-like lymphoproliferative disorder (HVLPD). Patients and Methods Seven patients with HVLPD had their clinical and pathological data collected. Immunohistochemical staining, Epstein-Barr virus-encoded RNA (EBER) in situ hybridization experiments, T-cell receptor (TCR) gene rearrangement, RT-PCR tests and the Elisa assay were carried out. Results The main clinical manifestations were papulovesicular lesions and ulcers on the face, neck, or trunk. Five cases had systemic symptoms. Three of the deceased patients had significant facial edema, deep body necrosis, and ulceration. The pathological results demonstrated that lymphocytes infiltrated blood vessels and sweat glands in addition to the dermis and subcutaneous tissues. All patients tested positive for CD3 and EBER. Six cases tested positive for TCRβF1, but none tested positive for TCRδ. TCRγ monoclonal rearrangement, strongly positive expression of TIA-1 and a Ki67 proliferation index of 40% occurred in 3 fatal cases. When compared to the survival group, the plasma EBV DNA in the deceased group was considerably higher (P<0.05). IFN-γ and TNF-α cytokine levels in patients were higher than in the control group, particularly in the deceased group (P<0.05). The skin lesions on all patients recovered quickly underwent conservative care. Nonetheless, 3 patients passed away as the disease progressed in its latter stages. Conclusion In our cases, the main infiltrating cells were T cells and the dominant lymphocyte subclass was αβT cells. A significant increase in lgE level, plasma EBV DNA, IFN-γ, and TNF-α cytokine levels, decreased hemoglobin level, strongly positive expression of TIA-1, high Ki67 proliferation index, and positive TCR gene rearrangement are all indicators of a poor prognosis.
Collapse
Affiliation(s)
- Li Chang
- Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Nanning, People’s Republic of China
| | - Chaoyin Zhang
- Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jingjing Lu
- Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Jiahui Shen
- Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Krishna Hamal
- Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
| | - Donghua Liu
- Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, Nanning, People’s Republic of China
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Nanning, People’s Republic of China
- Correspondence: Donghua Liu, Department of Dermatology and Venereology, the First Affiliated Hospital of Guangxi Medical University, No. 6, Shuang Yong Road, Nanning, Guangxi Province, 530021, People’s Republic of China, Tel +86 771-5356752, Email
| |
Collapse
|
192
|
Gao W, Hou R, Chen Y, Wang X, Liu G, Hu W, Yao K, Hao Y. A Predictive Disease Risk Model for Ankylosing Spondylitis: Based on Integrated Bioinformatic Analysis and Identification of Potential Biomarkers Most Related to Immunity. Mediators Inflamm 2023; 2023:3220235. [PMID: 37152368 PMCID: PMC10159744 DOI: 10.1155/2023/3220235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/08/2022] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
Background The pathogenesis of ankylosing spondylitis (AS) is still not clear, and immune-related genes have not been systematically explored in AS. The purpose of this paper was to identify the potential early biomarkers most related to immunity in AS and develop a predictive disease risk model with bioinformatic methods and the Gene Expression Omnibus database (GEO) to improve diagnostic and therapeutic efficiency. Methods To identify differentially expressed genes and create a gene coexpression network between AS and healthy samples, we downloaded the AS-related datasets GSE25101 and GSE73754 from the GEO database and employed weighted gene coexpression network analysis (WGCNA). We used the GSVA, GSEABase, limma, ggpubr, and reshape2 packages to score immune data and investigated the links between immune cells and immunological functions by using single-sample gene set enrichment analysis (ssGSEA). The value of the core gene set and constructed model for early AS diagnosis was investigated by using receiver operating characteristic (ROC) curve analysis. Results Biological function and immune score analyses identified central genes related to immunity, key immune cells, key related pathways, gene modules, and the coexpression network in AS. Granulysin (GNLY), Granulysin (GZMK), CX3CR1, IL2RB, dysferlin (DYSF), and S100A12 may participate in AS development through NK cells, CD8+ T cells, Th1 cells, and other immune cells and represent potential biomarkers for the early diagnosis of AS occurrence and progression. Furthermore, the T cell coinhibitory pathway may be involved in AS pathogenesis. Conclusion The AS disease risk model constructed based on immune-related genes can guide clinical diagnosis and treatment and may help in the development of personalized immunotherapy.
Collapse
Affiliation(s)
- Wenxin Gao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Ruirui Hou
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yungang Chen
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Xiaoying Wang
- Jinan Vocational College of Nursing, Jinan, Shandong Province, China
| | - Guoyan Liu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Wanli Hu
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Kang Yao
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yanke Hao
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| |
Collapse
|
193
|
Zhao PY, Yao RQ, Zheng LY, Wu Y, Li YX, Dong N, Li JY, Du XH, Yao YM. Nuclear fragile X mental retardation-interacting protein 1-mediated ribophagy protects T lymphocytes against apoptosis in sepsis. BURNS & TRAUMA 2023; 11:tkac055. [PMID: 36873287 PMCID: PMC9976742 DOI: 10.1093/burnst/tkac055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/08/2022] [Indexed: 03/04/2023]
Abstract
Background Ribophagy is a selective autophagic process that specifically degrades dysfunctional or superfluous ribosomes to maintain cellular homeostasis. Whether ribophagy can ameliorate the immunosuppression in sepsis similar to endoplasmic reticulum autophagy (ERphagy) and mitophagy remains unclear. This study was conducted to investigate the activity and regulation of ribophagy in sepsis and to further explore the potential mechanism underlying the involvement of ribophagy in T-lymphocyte apoptosis. Methods The activity and regulation of nuclear fragile X mental retardation-interacting protein 1 (NUFIP1)-mediated ribophagy in T lymphocytes during sepsis were first investigated by western blotting, laser confocal microscopy and transmission electron microscopy. Then, we constructed lentivirally transfected cells and gene-defective mouse models to observe the impact of NUFIP1 deletion on T-lymphocyte apoptosis and finally explored the signaling pathway associated with T-cell mediated immune response following septic challenge. Results Both cecal ligation and perforation-induced sepsis and lipopolysaccharide stimulation significantly induced the occurrence of ribophagy, which peaked at 24 h. When NUFIP1 was knocked down, T-lymphocyte apoptosis was noticeably increased. Conversely, the overexpression of NUFIP1 exerted a significant protective impact on T-lymphocyte apoptosis. Consistently, the apoptosis and immunosuppression of T lymphocytes and 1-week mortality rate in NUFIP1 gene-deficient mice were significantly increased compared with those in wild-type mice. In addition, the protective effect of NUFIP1-mediated ribophagy on T lymphocytes was identified to be closely related to the endoplasmic reticulum stress apoptosis pathway, and PERK-ATF4-CHOP signaling was obviously involved in downregulating T-lymphocyte apoptosis in the setting of sepsis. Conclusions NUFIP1-mediated ribophagy can be significantly activated to alleviate T lymphocyte apoptosis through the PERK-ATF4-CHOP pathway in the context of sepsis. Thus, targeting NUFIP1-mediated ribophagy might be of importance in reversing the immunosuppression associated with septic complications.
Collapse
Affiliation(s)
- Peng-Yue Zhao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.,Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Ren-Qi Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.,Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, China
| | - Li-Yu Zheng
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Yao Wu
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Yu-Xuan Li
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.,Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Ning Dong
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Jing-Yan Li
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.,Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Xiao-Hui Du
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Yong-Ming Yao
- Translational Medicine Research Center, Medical Innovation Research Division and Fourth Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
194
|
Lu J, Liang T, Li P, Yin Q. Regulatory effects of IRF4 on immune cells in the tumor microenvironment. Front Immunol 2023; 14:1086803. [PMID: 36814912 PMCID: PMC9939821 DOI: 10.3389/fimmu.2023.1086803] [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: 11/01/2022] [Accepted: 01/18/2023] [Indexed: 02/09/2023] Open
Abstract
The tumor microenvironment (TME) is implicated in tumorigenesis, chemoresistance, immunotherapy failure and tumor recurrence. Multiple immunosuppressive cells and soluble secreted cytokines together drive and accelerate TME disorders, T cell immunodeficiency and tumor growth. Thus, it is essential to comprehensively understand the TME status, immune cells involved and key transcriptional factors, and extend this knowledge to therapies that target dysfunctional T cells in the TME. Interferon regulatory factor 4 (IRF4) is a unique IRF family member that is not regulated by interferons, instead, is mainly induced upon T-cell receptor signaling, Toll-like receptors and tumor necrosis factor receptors. IRF4 is largely restricted to immune cells and plays critical roles in the differentiation and function of effector cells and immunosuppressive cells, particularly during clonal expansion and the effector function of T cells. However, in a specific biological context, it is also involved in the transcriptional process of T cell exhaustion with its binding partners. Given the multiple effects of IRF4 on immune cells, especially T cells, manipulating IRF4 may be an important therapeutic target for reversing T cell exhaustion and TME disorders, thus promoting anti-tumor immunity. This study reviews the regulatory effects of IRF4 on various immune cells in the TME, and reveals its potential mechanisms, providing a novel direction for clinical immune intervention.
Collapse
Affiliation(s)
- Jing Lu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Taotao Liang
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Ping Li
- Department of Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Qingsong Yin
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| |
Collapse
|
195
|
Osuna-Espinoza KY, Rosas-Taraco AG. Metabolism of NK cells during viral infections. Front Immunol 2023; 14:1064101. [PMID: 36742317 PMCID: PMC9889541 DOI: 10.3389/fimmu.2023.1064101] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/04/2023] [Indexed: 01/19/2023] Open
Abstract
Cellular metabolism is essential for the correct function of immune system cells, including Natural Killer cells (NK). These cells depend on energy to carry out their effector functions, especially in the early stages of viral infection. NK cells participate in the innate immune response against viruses and tumors. Their main functions are cytotoxicity and cytokine production. Metabolic changes can impact intracellular signals, molecule production, secretion, and cell activation which is essential as the first line of immune defense. Metabolic variations in different immune cells in response to a tumor or pathogen infection have been described; however, little is known about NK cell metabolism in the context of viral infection. This review summarizes the activation-specific metabolic changes in NK cells, the immunometabolism of NK cells during early, late, and chronic antiviral responses, and the metabolic alterations in NK cells in SARS-CoV2 infection. The modulation points of these metabolic routes are also discussed to explore potential new immunotherapies against viral infections.
Collapse
Affiliation(s)
- Kenia Y Osuna-Espinoza
- Faculty of Medicine, Department of Immunology, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon, Mexico
| | - Adrián G Rosas-Taraco
- Faculty of Medicine, Department of Immunology, Universidad Autonoma de Nuevo Leon, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
196
|
Heuser C, Renner K, Kreutz M, Gattinoni L. Targeting lactate metabolism for cancer immunotherapy - a matter of precision. Semin Cancer Biol 2023; 88:32-45. [PMID: 36496155 DOI: 10.1016/j.semcancer.2022.12.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors and adoptive T cell therapies have been valuable additions to the toolbox in the fight against cancer. These treatments have profoundly increased the number of patients with a realistic perspective toward a return to a cancer-free life. Yet, in a number of patients and tumor entities, cancer immunotherapies have been ineffective so far. In solid tumors, immune exclusion and the immunosuppressive tumor microenvironment represent substantial roadblocks to successful therapeutic outcomes. A major contributing factor to the depressed anti-tumor activity of immune cells in tumors is the harsh metabolic environment. Hypoxia, nutrient competition with tumor and stromal cells, and accumulating noxious waste products, including lactic acid, pose massive constraints to anti-tumor immune cells. Numerous strategies are being developed to exploit the metabolic vulnerabilities of tumor cells in the hope that these would also alleviate metabolism-inflicted immune suppression. While promising in principle, especially in combination with immunotherapies, these strategies need to be scrutinized for their effect on tumor-fighting immune cells, which share some of their key metabolic properties with tumor cells. Here, we provide an overview of strategies that seek to tackle lactate metabolism in tumor or immune cells to unleash anti-tumor immune responses, thereby opening therapeutic options for patients whose tumors are currently not treatable.
Collapse
Affiliation(s)
- Christoph Heuser
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany.
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Department of Otorhinolaryngology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Clinical Cooperation Group Immunometabolomics, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology (CITO), University Hospital Regensburg, 93053 Regensburg, Germany
| | - Luca Gattinoni
- Division of Functional Immune Cell Modulation, Leibniz Institute for Immunotherapy (LIT), 93053 Regensburg, Germany; Center for Immunomedicine in Transplantation and Oncology (CITO), University Hospital Regensburg, 93053 Regensburg, Germany; University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
197
|
Chen C, Wang Z, Ding Y, Qin Y. Manipulating T-cell metabolism to enhance immunotherapy in solid tumor. Front Immunol 2022; 13:1090429. [PMID: 36618408 PMCID: PMC9812959 DOI: 10.3389/fimmu.2022.1090429] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
Cellular metabolism is not only essential for tumor cells to sustain their rapid growth and proliferation, but also crucial to maintain T cell fitness and robust immunity. Dysregulated metabolism has been recognized as a hallmark of cancer, which provides survival advantages for tumor cells under stress conditions. Also, emerging evidence suggests that metabolic reprogramming impacts the activation, differentiation, function, and exhaustion of T cells. Normal stimulation of resting T cells promotes the conversion of catabolic and oxidative metabolism to aerobic glycolysis in effector T cells, and subsequently back to oxidative metabolism in memory T cells. These metabolic transitions profoundly affect the trajectories of T-cell differentiation and fate. However, these metabolic events of T cells could be dysregulated by their interplays with tumor or the tumor microenvironment (TME). Importantly, metabolic competition in the tumor ecosystem is a new mechanism resulting in strong suppression of effector T cells. It is appreciated that targeting metabolic reprogramming is a promising way to disrupt the hypermetabolic state of tumor cells and enhance the capacity of immune cells to obtain nutrients. Furthermore, immunotherapies, such as immune checkpoint inhibitor (ICI), adoptive cell therapy (ACT), and oncolytic virus (OV) therapy, have significantly refashioned the clinical management of solid tumors, they are not sufficiently effective for all patients. Understanding how immunotherapy affects T cell metabolism provides a bright avenue to better modulate T cell anti-tumor response. In this review, we provide an overview of the cellular metabolism of tumor and T cells, provide evidence on their dynamic interaction, highlight how metabolic reprogramming of tumor and T cells regulate the anti-tumor responses, describe T cell metabolic patterns in the context of ICI, ACT, and OV, and propose hypothetical combination strategies to favor potent T cell functionality.
Collapse
|
198
|
Wang S, Shi Y. Exosomes Derived from Immune Cells: The New Role of Tumor Immune Microenvironment and Tumor Therapy. Int J Nanomedicine 2022; 17:6527-6550. [PMID: 36575698 PMCID: PMC9790146 DOI: 10.2147/ijn.s388604] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 12/09/2022] [Indexed: 12/29/2022] Open
Abstract
Exosomes are small vesicles secreted by living cells, with a typical lipid bilayer structure. They carry a variety of proteins, lipids, RNA and other important information, play an important role in the transmission of substances and information between cells, and gradually become a marker for early diagnosis of many diseases and an important tool in drug delivery system. Immune cells are an important part of tumor microenvironment, and they can affect tumor progression by secreting a variety of immunoreactive substances. This review focuses on the effects of various immune cell-derived exosomes on tumor cells, different immune cells and other stromal cells in tumor microenvironment. Exosomes derived from different immune cells can not only reshape a pro-inflammatory microenvironment to inhibit tumor progression, but also promote tumor progression by inhibiting the killing effect of NK cells, CD8+T cells and other cells or promoting tumor cells and immunosuppressive immune cells. In addition, we also discussed that some exosomes derived from immune cells (such as DC, M1 macrophages and neutrophils) play a tumor inhibitory role after being engineered.
Collapse
Affiliation(s)
- Shiyang Wang
- Department of Geriatric Surgery, The First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China
| | - Yue Shi
- Department of Geriatric Surgery, The First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China,Correspondence: Yue Shi, Department of Geriatric Surgery, The First Hospital of China Medical University, Shenyang, 110001, People’s Republic of China, Tel +86-13842073309, Email
| |
Collapse
|
199
|
Li H, Zhao A, Li M, Shi L, Han Q, Hou Z. Targeting T-cell metabolism to boost immune checkpoint inhibitor therapy. Front Immunol 2022; 13:1046755. [PMID: 36569893 PMCID: PMC9768337 DOI: 10.3389/fimmu.2022.1046755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have shown promising therapeutic effects in the treatment of advanced solid cancers, but their overall response rate is still very low for certain tumor subtypes, limiting their clinical scope. Moreover, the high incidence of drug resistance (including primary and acquired) and adverse effects pose significant challenges to the utilization of these therapies in the clinic. ICIs enhance T cell activation and reverse T cell exhaustion, which is a complex and multifactorial process suggesting that the regulatory mechanisms of ICI therapy are highly heterogeneous. Recently, metabolic reprogramming has emerged as a novel means of reversing T-cell exhaustion in the tumor microenvironment; there is increasing evidence that T cell metabolic disruption limits the therapeutic effect of ICIs. This review focuses on the crosstalk between T-cell metabolic reprogramming and ICI therapeutic efficacy, and summarizes recent strategies to improve drug tolerance and enhance anti-tumor effects by targeting T-cell metabolism alongside ICI therapy. The identification of potential targets for altering T-cell metabolism can significantly contribute to the development of methods to predict therapeutic responsiveness in patients receiving ICI therapy, which are currently unknown but would be of great clinical significance.
Collapse
Affiliation(s)
- Haohao Li
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Alison Zhao
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve School of Medicine, Cleveland, OH, United States
| | - Menghua Li
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Lizhi Shi
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China
| | - Qiuju Han
- Institute of Immunopharmaceutical Sciences, School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong, China,*Correspondence: Qiuju Han, ; Zhaohua Hou,
| | - Zhaohua Hou
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, United States,*Correspondence: Qiuju Han, ; Zhaohua Hou,
| |
Collapse
|
200
|
Abstract
Significance: Cancer immunotherapy has yielded striking antitumor effects in many cancers, yet the proportion of benefited patients is still limited. As key mediators of tumor suppression, CD8+ T cells are crucial for cancer immunotherapy. It has been widely appreciated that the modulation of CD8+ T cell immunity could be an effective way to further improve the therapeutic benefit of immunotherapy. Recent Advances: Emerging evidence has underlined a close link between metabolism and immune functions, providing a metabolism-immune axis that is increasingly investigated for understanding CD8+ T cell regulation. On the other hand, growing findings have reported that tumors adopt multiple approaches to induce metabolic reprogramming of CD8+ T cells, leading to compromised immunotherapy. Critical Issues: CD8+ T cell metabolism in the tumor microenvironment (TME) is often adapted to diminish antitumor immune responses and thereby evade from immune surveillance. A better understanding of metabolic regulation of CD8+ T cells in the TME is believed to hold promise for opening a new therapeutic window to further improve the benefit of immunotherapy. We herein review the mechanistic understanding of how CD8+ T cell metabolism is reprogrammed in the TME, mainly focusing on the impact of nutrient availability and bioactive molecules secreted by surrounding cells. Future Directions: Future research should pay attention to tumor heterogeneity in the metabolic microenvironment and associated immune responses. It is also important to include the trending opinion of "precision medicine" in cancer immunotherapies to tailor metabolic interventions for individual patients in combination with immunotherapy treatments. Antioxid. Redox Signal. 37, 1234-1253.
Collapse
Affiliation(s)
- Ying Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiaomin Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|