1
|
Horbatok K, Semchuk I, Horbach O, Khranovska N, Kosach V, Borysko P, Koniev S, Ulrich AS, Afonin S, Komarov IV. In vitro evaluation of the immunogenic potential of gramicidin S and its photocontrolled analogues. RSC Med Chem 2025:d5md00075k. [PMID: 40270993 PMCID: PMC12013366 DOI: 10.1039/d5md00075k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Three hallmarks of ICD (immunogenic cell death), release of adenosine triphosphate (ATP), release of high mobility group box 1 protein, and calreticulin exposure on the cell surface, were studied upon treatment of mammalian cells with small cyclic peptides, namely, the natural antibiotic gramicidin S (GS) and two photocontrolled GS analogues (LMB002 and LMB033). The analogues contained a photoisomerizable diarylethene fragment, and they exhibited different bioactivities in their "open" and "closed" photoisomeric forms. The data (obtained from cell cultures and spheroids) were collected in a concentration-dependent manner to assess cytotoxicity. Results showed that treatment with all peptides induced ICD at sub-IC50 and higher concentrations, indicating that GS and its derivatives have promising immunogenic potential. The "open" photoisomers of the photoswitchable GS analogues generated using visible light were as efficient as ICD inducers and the parent GS, while the UV-generated "closed" photoforms induced ICD only at higher concentrations. Herein, the cell specificity and time dependency of the observed effects are presented.
Collapse
Affiliation(s)
- Kateryna Horbatok
- Taras Shevchenko National University of Kyiv Volodymyrska street 60 01601 Kyiv Ukraine
- Enamine Ltd. Winston Churchill street 78 02094 Kyiv Ukraine
| | - Iryna Semchuk
- Nonprofit organization "National Cancer Institute" Yulii Zdanovskoi street 33/43 03022 Kyiv Ukraine
| | - Oleksandr Horbach
- Nonprofit organization "National Cancer Institute" Yulii Zdanovskoi street 33/43 03022 Kyiv Ukraine
| | - Natalia Khranovska
- Nonprofit organization "National Cancer Institute" Yulii Zdanovskoi street 33/43 03022 Kyiv Ukraine
| | | | - Petro Borysko
- Enamine Ltd. Winston Churchill street 78 02094 Kyiv Ukraine
- V. P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry Akademician Kukhar street 1 02094 Kyiv Ukraine
| | - Serhii Koniev
- Karlsruhe Institute of Technology POB 3640 76021 Karlsruhe Germany
| | - Anne S Ulrich
- Karlsruhe Institute of Technology POB 3640 76021 Karlsruhe Germany
| | - Sergii Afonin
- Karlsruhe Institute of Technology POB 3640 76021 Karlsruhe Germany
| | - Igor V Komarov
- Taras Shevchenko National University of Kyiv Volodymyrska street 60 01601 Kyiv Ukraine
- Enamine Ltd. Winston Churchill street 78 02094 Kyiv Ukraine
- Lumobiotics Auerstraße 2 76227 Karlsruhe Germany
| |
Collapse
|
2
|
Aubrun C, Doussineau T, Carmès L, Meyzaud A, Boux F, Dufort S, Delfour A, De Beaumont O, Mirjolet C, Le Duc G. Mechanisms of Action of AGuIX as a Pan-Cancer Nano-Radiosensitizer: A Comprehensive Review. Pharmaceuticals (Basel) 2025; 18:519. [PMID: 40283954 PMCID: PMC12030438 DOI: 10.3390/ph18040519] [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: 02/19/2025] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Objective: This review provides an overview of the current knowledge regarding the mechanisms of action of AGuIX, a clinical-stage theranostic nano-radiosensitizer composed of gadolinium. It covers the steps following the administration, from the internalization in tumor cells to the interaction with X-rays and the subsequent physical, chemical, biological, and immunological events. Results: After intravenous injection, AGuIX accumulates in tumors through the enhanced permeability and retention (EPR) effect, and its specific retention properties allow its persistence in tumors for several days. At the cellular level, the nanomedicine is internalized by endocytic processes and mainly located in the cytoplasm, especially in lysosomes. AGuIX enhances the effects of radiotherapy (RT) at several levels, starting from radiation-matter interactions to a chemical stage of reactive oxygen species (ROS) production, followed by a cascade of biological events leading to tumor cell death and immune response. Indeed, AGuIX induces a local increase in radiation dose deposition through the emission of Auger electrons, leading to a subsequent increase in ROS generation. AGuIX also impacts RT-induced biological mechanisms, including DNA damage and cell death mechanisms such as apoptosis, autophagic cell death, and ferroptosis. Last, the combination of AGuIX and RT stimulates an antitumor immune response through the induction of immunogenic cell death (ICD), the activation of dendritic and T cells, and the reprogramming of tumor-associated macrophages (TAMs) into a pro-inflammatory phenotype. Conclusions: AGuIX is a clinical-stage nanoparticle (NP) intravenously administered with pan-cancer potential due to its specific biodistribution properties and a strong ability to amplify RT-induced mechanisms.
Collapse
Affiliation(s)
- Clémentine Aubrun
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Tristan Doussineau
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Léna Carmès
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Aurélien Meyzaud
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Fabien Boux
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Sandrine Dufort
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Adeline Delfour
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Radiation Therapy Department, Centre Georges-François Leclerc, 21000 Dijon, France;
- TIReCS Team, CTM (Center for Translational and Molecular Medicine), INSERM UMR 1231, 21000 Dijon, France
| | - Olivier De Beaumont
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| | - Céline Mirjolet
- X-Rain: Research Unit in Radiotherapy Combined with Immunotherapies and Nanoparticles, IMATHERA, Radiation Therapy Department, Centre Georges-François Leclerc, 21000 Dijon, France;
- TIReCS Team, CTM (Center for Translational and Molecular Medicine), INSERM UMR 1231, 21000 Dijon, France
| | - Géraldine Le Duc
- NH TherAguix SA, 19 Chemin des Prés, 38240 Meylan, France; (T.D.); (L.C.); (A.M.); (F.B.); (S.D.); (A.D.); (O.D.B.)
| |
Collapse
|
3
|
Amiri M, Jafari S, Lavasanifar A, Molavi O, Montazersaheb S. Nano-delivery of Silibinin Potentiate the Induction of Immunogenic Cell Death (ICD) in Melanoma Cells. Curr Pharm Biotechnol 2025; 26:392-401. [PMID: 38482616 DOI: 10.2174/0113892010280336240227062954] [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: 10/25/2023] [Revised: 01/17/2024] [Accepted: 02/01/2024] [Indexed: 03/25/2025]
Abstract
BACKGROUND Induction of immunogenic cell death (ICD) in tumors can enhance antitumor immunity and modulate immunosuppression in the tumor microenvironment (TME). OBJECTIVE In the current study, we investigated the effect of silibinin, a natural compound with anticancer activity, and its polymer-based nanoformulations on the induction of apoptosis and ICD in cancer cells. METHODS Free and nanoparticulate silibinin were evaluated for their growth-inhibitory effects using an MTT assay. Annexin V/PI staining was used to analyze apoptosis. Calreticulin (CRT) expression was measured by flow cytometry. Western blotting was conducted to examine the levels of elf2α, which plays a role in the ICD pathway. The HSP90 and ATP levels were determined using specific detection kits. RESULTS Compared to the free drug, silibinin-loaded nanocarriers significantly increased the induction of apoptosis and ICD in B16F10 cells. ICD induction was characterized by significantly increased levels of ICD biomarkers, including CRT, HSP90, and ATP. We also observed an increased expression of p-elf-2α/ elf-2α in B16F10 cells treated with silibinin-loaded micelles compared to cells that received free silibinin. CONCLUSION Our findings showed that the encapsulation of silibinin in polymeric nanocarriers can potentiate the effects of this drug on the induction of apoptosis and ICD in B16F10 melanoma cells.
Collapse
Affiliation(s)
- Mina Amiri
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sevda Jafari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afsaneh Lavasanifar
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Canada
| | - Ommoleila Molavi
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soheila Montazersaheb
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
Xia L, Mei J, Huang M, Bao D, Wang Z, Chen Y. O-GlcNAcylation in ovarian tumorigenesis and its therapeutic implications. Transl Oncol 2025; 51:102220. [PMID: 39616984 DOI: 10.1016/j.tranon.2024.102220] [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: 09/13/2024] [Revised: 11/17/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024] Open
Abstract
Ovarian cancer is a prevalent malignancy among women, often associated with a poor prognosis. Post-translational modifications (PTMs), particularly O-GlcNAcylation, have been implicated in the progression of ovarian cancer. Emerging evidence indicates that dysregulation of O-GlcNAcylation contributes to the initiation and malignant progression of ovarian cancer. This review discusses the potential role of O-GlcNAcylation in ovarian tumorigenesis, with a focus on its regulation of various cellular signaling pathways, including p53, RhoA/ROCK/MLC, Ezrin/Radixin/Moesin (ERM), and β-catenin. This review also emphasizes the O-GlcNAcylation of critical proteins in ovarian cancer, such as SNAP-23, SNAP-29, E-cadherin, and calreticulin. Additionally, the potential of O-GlcNAcylation to enhance immunotherapy for ovarian cancer patients is explored. Several compounds targeting OGT and OGA in ovarian cancer are also highlighted. Targeting the dynamic and versatile nature of O-GlcNAcylation could undoubtedly contribute to more effective treatments and improved outcomes for ovarian cancer patients.
Collapse
Affiliation(s)
- Lu Xia
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Jie Mei
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Min Huang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Dandan Bao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Zhiwei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Yizhe Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
5
|
He W, Lv W, Liu L, Gong Y, Song K, Xu J, Zhao W, Li S, Min Z, Chen Q, Yin J, Chen Y, Fang H, Xin H, Fang X. Enhanced Antiglioma Effect by a Vitamin D3-Inserted Lipid Hybrid Neutrophil Membrane Biomimetic Multimodal Nanoplatform. ACS NANO 2024; 18:35559-35574. [PMID: 39696957 DOI: 10.1021/acsnano.4c13470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Glioblastoma, the most prevalent malignant brain tumor, is a lethal threat to human health, with aggressive and infiltrative growth characteristics that compromise the clinical treatment. Herein, we developed a vitamin D3-inserted lipid hybrid neutrophil membrane biomimetic multimodal nanoplatform (designated as NED@MnO2-DOX) through doxorubicin (DOX)-loaded manganese dioxide nanoparticles (MnO2) which were coated with a vitamin D3-inserted lipid hybrid neutrophil membrane. It was demonstrated that in addition to chemotherapy and chemo-dynamic therapy efficacy, NED@MnO2-DOX exhibited great power to activate and amplify immune responses related to the cGAS STING pathway, bolstering the secretion of type I interferon-β and proinflammatory cytokines, promoting the maturation of DC cells and infiltration of CD8+T cells into the glioma tissue, thereby reversing the immunosuppressive microenvironment of glioma from a "cold" tumor to a "hot" tumor. The biomimetic multimodal nanoplatform has potential as a multimodal strategy for glioma-targeted treatment, especially holding considerable promise for the development of innate immune therapy for glioma.
Collapse
Affiliation(s)
- Weichong He
- Department of Pharmacy, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou 213100, China
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Wei Lv
- Department of Pharmacy, The Jiangyin Clinical College of Xuzhou Medical University, Wuxi 214499, China
| | - Linfeng Liu
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Yue Gong
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Kefan Song
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Jiangna Xu
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Wei Zhao
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Shengnan Li
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Zhiyi Min
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Qinhua Chen
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Jiaqing Yin
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Yuqin Chen
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
| | - Hufeng Fang
- Department of Pharmacy, The Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical University, Changzhou 213100, China
| | - Hongliang Xin
- Department of Pharmaceutics, Pharmacy of School, Nanjing Medical University, Nanjing 211166, China
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi 214400, China
| | - Xiangming Fang
- Department of Radiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center of Nanjing Medical University, Wuxi 214400, China
| |
Collapse
|
6
|
Xu C. CRISPR/Cas9-mediated knockout strategies for enhancing immunotherapy in breast cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8561-8601. [PMID: 38907847 DOI: 10.1007/s00210-024-03208-2] [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: 12/28/2023] [Accepted: 05/31/2024] [Indexed: 06/24/2024]
Abstract
Breast cancer, a prevalent disease with significant mortality rates, often presents treatment challenges due to its complex genetic makeup. This review explores the potential of combining Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) gene knockout strategies with immunotherapeutic approaches to enhance breast cancer treatment. The CRISPR/Cas9 system, renowned for its precision in inducing genetic alterations, can target and eliminate specific cancer cells, thereby minimizing off-target effects. Concurrently, immunotherapy, which leverages the immune system's power to combat cancer, has shown promise in treating breast cancer. By integrating these two strategies, we can potentially augment the effectiveness of immunotherapies by knocking out genes that enable cancer cells to evade the immune system. However, safety considerations, such as off-target effects and immune responses, necessitate careful evaluation. Current research endeavors aim to optimize these strategies and ascertain the most effective methods to stimulate the immune response. This review provides novel insights into the integration of CRISPR/Cas9-mediated knockout strategies and immunotherapy, a promising avenue that could revolutionize breast cancer treatment as our understanding of the immune system's interplay with cancer deepens.
Collapse
Affiliation(s)
- Chenchen Xu
- Department of Gynecology and Obstetrics, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
7
|
Li Z, Han B, Qi M, Li Y, Duan Y, Yao Y. Modulating macrophage-mediated programmed cell removal: An attractive strategy for cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189172. [PMID: 39151808 DOI: 10.1016/j.bbcan.2024.189172] [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: 01/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Macrophage-mediated programmed cell removal (PrCR) is crucial for the identification and elimination of needless cells that maintain tissue homeostasis. The efficacy of PrCR depends on the balance between pro-phagocytic "eat me" signals and anti-phagocytic "don't eat me" signals. Recently, a growing number of studies have shown that tumourigenesis and progression are closely associated with PrCR. In the tumour microenvironment, PrCR activated by the "eat me" signal is counterbalanced by the "don't eat me" signal of CD47/SIRPα, resulting in tumour immune escape. Therefore, targeting exciting "eat me" signalling while simultaneously suppressing "don't eat me" signalling and eventually inducing macrophages to produce effective PrCR will be a very attractive antitumour strategy. Here, we comprehensively review the functions of PrCR-activating signal molecules (CRT, PS, Annexin1, SLAMF7) and PrCR-inhibiting signal molecules (CD47/SIRPα, MHC-I/LILRB1, CD24/Siglec-10, SLAMF3, SLAMF4, PD-1/PD-L1, CD31, GD2, VCAM1), the interactions between these molecules, and Warburg effect. In addition, we highlight the molecular regulatory mechanisms that affect immune system function by exciting or suppressing PrCR. Finally, we review the research advances in tumour therapy by activating PrCR and discuss the challenges and potential solutions to smooth the way for tumour treatment strategies that target PrCR.
Collapse
Affiliation(s)
- Zhenzhen Li
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bingqian Han
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Menghui Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yinchao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongtao Duan
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China.
| | - Yongfang Yao
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
8
|
Wang ZQ, Qu TR, Zhang ZS, Zeng FS, Song HJ, Zhang K, Guo P, Tong Z, Hou DY, Liu X, Wang L, Wang H, Xu W. A Transformable Specific-Responsive Peptide for One-Step Synergistic Therapy of Bladder Cancer. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310416. [PMID: 38660815 DOI: 10.1002/smll.202310416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 04/07/2024] [Indexed: 04/26/2024]
Abstract
Synergistic therapy has shown greater advantages compared with monotherapy. However, the complex multiple-administration plan and potential side effects limit its clinical application. A transformable specific-responsive peptide (TSRP) is utilized to one-step achieve synergistic therapy integrating anti-tumor, anti-angiogenesis and immune response. The TSRP is composed of: i) Recognition unit could specifically target and inhibit the biological function of FGFR-1; ii) Transformable unit could self-assembly and trigger nanofibers formation; iii) Reactive unit could specifically cleaved by MMP-2/9 in tumor micro-environment; iv) Immune unit, stimulate the release of immune cells when LTX-315 (Immune-associated oncolytic peptide) exposed. Once its binding to FGFR-1, the TSRP could cleaved by MMP-2/9 to form the nanofibers on the cell membrane, with a retention time of up to 12 h. Through suppressing the phosphorylation levels of ERK 1/2 and PI3K/AKT signaling pathways downstream of FGFR-1, the TSRP significant inhibit the growth of tumor cells and the formation of angioginesis. Furthermore, LTX-315 is exposed after TSRP cleavage, resulting in Calreticulin activation and CD8+ T cells infiltration. All above processes together contribute to the increasing survival rate of tumor-bearing mice by nearly 4-folds. This work presented a unique design for the biological application of one-step synergistic therapy of bladder cancer.
Collapse
Affiliation(s)
- Zi-Qi Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Tian-Rui Qu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, China
| | - Zhi-Shuai Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Fan-Shu Zeng
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hong-Jian Song
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Kuo Zhang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Pengyu Guo
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Zhichao Tong
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, China
| | - Da-Yong Hou
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Xiao Liu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Lu Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, the Fourth Hospital of Harbin Medical University, Harbin, 150001, China
| | - Hao Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology (NCNST), Beijing, 100190, China
| | - Wanhai Xu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Harbin Medical University, Harbin, 150001, China
- Heilongjiang Key Laboratory of Scientific Research in Urology, Harbin, 150001, China
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| |
Collapse
|
9
|
Xu S, Liu H, Li X, Zhao J, Wang J, Crans DC, Yang X. Approaches to selective and potent inhibition of glioblastoma by vanadyl complexes: Inducing mitotic catastrophe and methuosis. J Inorg Biochem 2024; 257:112610. [PMID: 38761580 DOI: 10.1016/j.jinorgbio.2024.112610] [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: 01/04/2024] [Revised: 04/08/2024] [Accepted: 05/09/2024] [Indexed: 05/20/2024]
Abstract
Drug resistance has been a major problem for cancer chemotherapy, especially for glioblastoma multiforme that is aggressive, heterogeneous and recurrent with <3% of a five-year survival and limited methods of clinical treatment. To overcome the problem, great efforts have recently been put in searching for agents inducing death of tumor cells via various non-apoptotic pathways. In the present work, we report for the first time that vanadyl complexes, i.e. bis(acetylacetonato)oxidovanadium (IV) (VO(acac)2), can cause mitotic catastrophe and methuotic death featured by catastrophic macropinocytic vacuole accumulation particularly in glioblastoma cells (GCs). Hence, VO(acac)2 strongly suppressed growth of GCs with both in vitro (IC50 = 4-6 μM) and in vivo models, and is much more potent than the current standard-of-care drug Temozolomide. The selective index is as high as ∼10 or more on GCs over normal neural cells. Importantly, GCs respond well to vanadium treatment regardless whether they are carrying IDH1 wild type gene that causes drug resistance. VO(acac)2 may induce methuosis via the Rac-Mitogen-activated protein kinase kinase 4 (MKK4)-c-Jun N-terminal kinase (JNK) signaling pathway. Furthermore, VO(acac)2-induced methuosis is not through a immunogenicity mechanism, making vanadyl complexes safe for interventional therapy. Overall, our results may encourage development of novel vanadium complexes promising for treatment of neural malignant tumor cells.
Collapse
Affiliation(s)
- Sha Xu
- State Key Laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Huixue Liu
- State Key Laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xin Li
- State Key Laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jingyan Zhao
- State Key Laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jiayu Wang
- State Key Laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Debbie C Crans
- Department of Chemistry and Cell and Molecular Biology Program, College of Natural Science, Colorado State University, Fort Collins, CO 80523-1872, USA.
| | - Xiaoda Yang
- State Key Laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China; SATCM Key Laboratory of Compound Drug Detoxification, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
10
|
Li X, Su N, Yu H, Li X, Sun SL. Hainanenin-1, an oncolytic peptide, triggers immunogenic cell death via STING activation in triple-negative breast cancer. Cell Commun Signal 2024; 22:352. [PMID: 38970078 PMCID: PMC11225514 DOI: 10.1186/s12964-024-01731-6] [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: 04/11/2024] [Accepted: 06/30/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND In triple-negative breast cancer (TNBC) therapy, insufficient tumor infiltration by lymphocytes significantly hinders the efficacy of immune checkpoint inhibitors. We have previously demonstrated that Hainanenin-1 (HN-1), a host defense peptide (HDP) identified from Hainan frog skin, induces breast cancer apoptosis and boots anti-tumor immunity via unknown mechanism. METHODS We used in vitro experiments to observe immunogenic cell death (ICD) indicators in HN-1-treated TNBC cell lines, a mouse tumor model to verify HN-1 promotion of mice anti-tumor immune response, and an in vitro drug sensitivity test of patient-derived breast cancer cells to verify the inhibitory effect of HN-1. RESULTS HN-1 induced ICD in TNBC in a process during which damage-associated molecular patterns (DAMPs) were released that could further increase the anti-tumor immune response. The secretion level of interleukin 2 (IL-2), IL-12, and interferon γ in the co-culture supernatant was increased, and dendritic cells (DCs) were activated via a co-culture with HN-1-pretreated TNBC cells. As a result, HN-1 increased the infiltration of anti-tumor immune cells (DCs and T lymphocytes) in the mouse model bearing both 4T1 and EMT6 tumors. Meanwhile, regulatory T cells and myeloid-derived suppressor cells were suppressed. In addition, HN-1 induced DNA damage, and double-strand DNA release in the cytosol was significantly enhanced, indicating that HN-1 might stimulate ICD via activation of STING pathway. The knockdown of STING inhibited HN-1-induced ICD. Of note, HN-1 exhibited inhibitory effects on patient-derived breast cancer cells under three-dimensional culture conditions. CONCLUSIONS Collectively, our study demonstrated that HN-1 could be utilized as a potential compound that might augment immunotherapy effects in patients with TNBC.
Collapse
Affiliation(s)
- Xiaoxi Li
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China
| | - Nan Su
- Central Laboratory, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China
| | - Haining Yu
- School of Bioengineering, Dalian University of Technology, Dalian, Liaoning, 116024, P. R. China.
| | - Xiaoyan Li
- Department of Pathology, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China.
| | - Shu-Lan Sun
- Central Laboratory, Cancer Hospital of Dalian University of Technology (Liaoning Cancer Hospital & Institute), Shenyang, Liaoning, 110042, P. R. China.
| |
Collapse
|
11
|
Zhou Q, Wu F, Zhang W, Guo Y, Jiang X, Yan X, Ke Y. Machine learning-based identification of a cell death-related signature associated with prognosis and immune infiltration in glioma. J Cell Mol Med 2024; 28:e18463. [PMID: 38847472 PMCID: PMC11157676 DOI: 10.1111/jcmm.18463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 04/27/2024] [Accepted: 05/17/2024] [Indexed: 06/10/2024] Open
Abstract
Accumulating evidence suggests that a wide variety of cell deaths are deeply involved in cancer immunity. However, their roles in glioma have not been explored. We employed a logistic regression model with the shrinkage regularization operator (LASSO) Cox combined with seven machine learning algorithms to analyse the patterns of cell death (including cuproptosis, ferroptosis, pyroptosis, apoptosis and necrosis) in The Cancer Genome Atlas (TCGA) cohort. The performance of the nomogram was assessed through the use of receiver operating characteristic (ROC) curves and calibration curves. Cell-type identification was estimated by using the cell-type identification by estimating relative subsets of known RNA transcripts (CIBERSORT) and single sample gene set enrichment analysis methods. Hub genes associated with the prognostic model were screened through machine learning techniques. The expression pattern and clinical significance of MYD88 were investigated via immunohistochemistry (IHC). The cell death score represents an independent prognostic factor for poor outcomes in glioma patients and has a distinctly superior accuracy to that of 10 published signatures. The nomogram performed well in predicting outcomes according to time-dependent ROC and calibration plots. In addition, a high-risk score was significantly related to high expression of immune checkpoint molecules and dense infiltration of protumor cells, these findings were associated with a cell death-based prognostic model. Upregulated MYD88 expression was associated with malignant phenotypes and undesirable prognoses according to the IHC. Furthermore, high MYD88 expression was associated with poor clinical outcomes and was positively related to CD163, PD-L1 and vimentin expression in the in-horse cohort. The cell death score provides a precise stratification and immune status for glioma. MYD88 was found to be an outstanding representative that might play an important role in glioma.
Collapse
Affiliation(s)
- Quanwei Zhou
- The National Key Clinical Specialty, Department of NeurosurgeryZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Fei Wu
- The National Key Clinical Specialty, Department of NeurosurgeryZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Wenlong Zhang
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
| | - Youwei Guo
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
| | - Xingjun Jiang
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
| | - Xuejun Yan
- NHC Key Laboratory of Birth Defect for Research and PreventionHunan Provincial Maternal and Child Health Care HospitalChangshaHunanChina
| | - Yiquan Ke
- The National Key Clinical Specialty, Department of NeurosurgeryZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
12
|
Zhang MR, Fang LL, Guo Y, Wang Q, Li YJ, Sun HF, Xie SY, Liang Y. Advancements in Stimulus-Responsive Co-Delivery Nanocarriers for Enhanced Cancer Immunotherapy. Int J Nanomedicine 2024; 19:3387-3404. [PMID: 38617801 PMCID: PMC11012697 DOI: 10.2147/ijn.s454004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/14/2024] [Indexed: 04/16/2024] Open
Abstract
Cancer immunotherapy has emerged as a novel therapeutic approach against tumors, with immune checkpoint inhibitors (ICIs) making significant clinical practice. The traditional ICIs, PD-1 and PD-L1, augment the cytotoxic function of T cells through the inhibition of tumor immune evasion pathways, ultimately leading to the initiation of an antitumor immune response. However, the clinical implementation of ICIs encounters obstacles stemming from the existence of an immunosuppressive tumor microenvironment and inadequate infiltration of CD8+T cells. Considerable attention has been directed towards advancing immunogenic cell death (ICD) as a potential solution to counteract tumor cell infiltration and the immunosuppressive tumor microenvironment. This approach holds promise in transforming "cold" tumors into "hot" tumors that exhibit responsiveness to antitumor. By combining ICD with ICIs, a synergistic immune response against tumors can be achieved. However, the combination of ICD inducers and PD-1/PD-L1 inhibitors is hindered by issues such as poor targeting and uncontrolled drug release. An advantageous solution presented by stimulus-responsive nanocarrier is integrating the physicochemical properties of ICD inducers and PD-1/PD-L1 inhibitors, facilitating precise delivery to specific tissues for optimal combination therapy. Moreover, these nanocarriers leverage the distinct features of the tumor microenvironment to accomplish controlled drug release and regulate the kinetics of drug delivery. This article aims to investigate the advancement of stimulus-responsive co-delivery nanocarriers utilizing ICD and PD-1/PD-L1 inhibitors. Special focus is dedicated to exploring the advantages and recent advancements of this system in enabling the combination of ICIs and ICD inducers. The molecular mechanisms of ICD and ICIs are concisely summarized. In conclusion, we examine the potential research prospects and challenges that could greatly enhance immunotherapeutic approaches for cancer treatment.
Collapse
Affiliation(s)
- Meng-Ru Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
- Department of Clinical Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Lin-Lin Fang
- RemeGen Co., Ltd, YanTai, ShanDong, 264000, People’s Republic of China
| | - Yang Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Qin Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - You-Jie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Hong-Fang Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Shu-Yang Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| | - Yan Liang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Binzhou Medical University, YanTai, ShanDong, 264003, People’s Republic of China
| |
Collapse
|
13
|
Li X, Tian W, Jiang Z, Song Y, Leng X, Yu J. Targeting CD24/Siglec-10 signal pathway for cancer immunotherapy: recent advances and future directions. Cancer Immunol Immunother 2024; 73:31. [PMID: 38279998 PMCID: PMC10821995 DOI: 10.1007/s00262-023-03606-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 12/07/2023] [Indexed: 01/29/2024]
Abstract
The small, heavily glycosylated protein CD24 is primarily expressed by many immune cells and is highly expressed mostly in cancer cells. As one of the most crucial biomarkers of cancers, CD24 is frequently highly expressed in solid tumors, while tumor-associated macrophages express Siglec-10 at high levels, Siglec-10 and CD24 can interact on innate immune cells to lessen inflammatory responses to a variety of disorders. Inhibiting inflammation brought on by SHP-1 and/or SHP-2 phosphatases as well as cell phagocytosis by macrophages, the binding of CD24 to Siglec-10 can prevent toll-like receptor-mediated inflammation. Targeted immunotherapy with immune checkpoint inhibitors (ICI) has lately gained popularity as one of the best ways to treat different tumors. CD24 is a prominent innate immune checkpoint that may be a useful target for cancer immunotherapy. In recent years, numerous CD24/Siglec-10-related research studies have made tremendous progress. This study discusses the characteristics and workings of CD24/Siglec-10-targeted immunotherapy and offers a summary of current advances in CD24/Siglec-10-related immunotherapy research for cancer. We then suggested potential directions for CD24-targeted immunotherapy, basing our speculation mostly on the results of recent preclinical and clinical trials.
Collapse
Affiliation(s)
- Xingchen Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130021, Jilin, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, 201203, China
| | - Zhongxing Jiang
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yongping Song
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiangyang Leng
- Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, 130021, Jilin, China.
| | - Jifeng Yu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004, Henan, China.
| |
Collapse
|
14
|
Chen S, Zhang W, Li X, Cao Z, Liu C. DNA polymerase beta connects tumorigenicity with the circadian clock in liver cancer through the epigenetic demethylation of Per1. Cell Death Dis 2024; 15:78. [PMID: 38245510 PMCID: PMC10799862 DOI: 10.1038/s41419-024-06462-7] [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: 09/12/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
The circadian-controlled DNA repair exhibits a strong diurnal rhythm. Disruption in circadian clock and DNA repair is closely linked with hepatocellular carcinoma (HCC) progression, but the mechanism remains unknown. Here, we show that polymerase beta (POLB), a critical enzyme in the DNA base excision repair pathway, is rhythmically expressed at the translational level in mouse livers. Hepatic POLB dysfunction dampens clock homeostasis, whereas retards HCC progression, by mediating the methylation of the 4th CpG island on the 5'UTR of clock gene Per1. Clinically, POLB is overexpressed in human HCC samples and positively associated with poor prognosis. Furthermore, the hepatic rhythmicity of POLB protein expression is orchestrated by Calreticulin (CALR). Our findings provide important insights into the molecular mechanism underlying the synergy between clock and food signals on the POLB-driven BER system and reveal new clock-dependent carcinogenetic effects of POLB. Therefore, chronobiological modulation of POLB may help to promote precise interventions for HCC.
Collapse
Affiliation(s)
- Siyu Chen
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Xiao Li
- Department of Pathology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhengyu Cao
- Jiangsu Provincial Key Laboratory for TCM Evaluation and Translational Development, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China.
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China.
| |
Collapse
|
15
|
Castellanos JP, Genereux JC. Calreticulin surface presentation: a signal for natural killer cells to attack. Signal Transduct Target Ther 2023; 8:289. [PMID: 37544934 PMCID: PMC10404582 DOI: 10.1038/s41392-023-01551-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/03/2023] [Accepted: 06/25/2023] [Indexed: 08/08/2023] Open
Affiliation(s)
| | - Joseph C Genereux
- Microbiology Graduate Program, University of California, Riverside, CA, 92521, USA.
- Department of Chemistry, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|