1
|
Kananivand M, Nouri F, Yousefi MH, Pajouhi A, Ghorbani H, Afkhami H, Razavi ZS. Mesenchymal stem cells and their exosomes: a novel approach to skin regeneration via signaling pathways activation. J Mol Histol 2025; 56:132. [PMID: 40208456 DOI: 10.1007/s10735-025-10394-7] [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: 12/01/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025]
Abstract
Accelerating wound healing is a crucial objective in surgical and regenerative medicine. The wound healing process involves three key stages: inflammation, cell proliferation, and tissue repair. Mesenchymal stem cells (MSCs) have demonstrated significant therapeutic potential in promoting tissue regeneration, particularly by enhancing epidermal cell migration and proliferation. However, the precise molecular mechanisms underlying MSC-mediated wound healing remain unclear. This review highlights the pivotal role of MSCs and their exosomes in wound repair, with a specific focus on critical signaling pathways, including PI3K/Akt, WNT/β-catenin, Notch, and MAPK. These pathways regulate essential cellular processes such as proliferation, differentiation, and angiogenesis. Moreover, in vitro and in vivo studies reveal that MSCs accelerate wound closure, enhance collagen deposition, and modulate immune responses, contributing to improved tissue regeneration. Understanding these mechanisms provides valuable insights into MSC-based therapeutic strategies for enhancing wound healing.
Collapse
Affiliation(s)
- Maryam Kananivand
- Medical Department, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Nouri
- Department of Biology, School of Basic Sciences, Science and Research Branch, Islamic Azad University (SRBIAU), Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
| | - Ali Pajouhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Hakimeah Ghorbani
- Department of Sciences, Faculty of Biological Sciences, Tabriz University of Sciences, Tabriz, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran.
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran.
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran.
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Luo AL, Zheng WY, Zhang Q, Yuan Y, Li MQ, Du K, Gao AR, Pei LJ, Xie J, Chen WH, Zhang L, Guo XZ, Yang XR, Zeng C, Yang GH, Deng M. COPS5 Triggers Ferroptosis Defense by Stabilizing MK2 in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416360. [PMID: 40198582 DOI: 10.1002/advs.202416360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/12/2025] [Indexed: 04/10/2025]
Abstract
Sorafenib, which is proven to serve as a potent ferroptosis inducer, is used as a first-line treatment for patients with advanced hepatocellular carcinoma (HCC), but it has limited clinical benefits, mainly due to drug resistance. Herein, using genome-wide CRISPR/Cas9 knockout screening and multiple functional studies, this work identifies COP9 signalosome subunit 5 (COPS5) as a driver of sorafenib resistance and a suppressor of ferroptosis in HCC. Consistently, the amplification and overexpression of COPS5 are frequently observed in clinical HCC samples, which are associated with poor patient prognosis and might predict patient response to sorafenib therapy. Mechanistically, COPS5 stabilized mitogen-activated protein kinase 2 (MK2) through deubiquitination and, in turn, induced the activation of heat shock protein beta-1 (HSPB1), a ferroptosis repressor, thereby protecting HCC cells from ferroptosis and consequently leading to sorafenib resistance and tumor progression, while its own expression could be induced by sorafenib treatment via activating transcription factor 4 (ATF4)-activated transcription. Furthermore, pharmacological inhibition of COPS5/MK2 synergize with sorafenib to induce ferroptosis and suppress HCC progression. This data reveals the crucial role of COPS5 in triggering ferroptosis defense and sorafenib resistance through the activation of the MK2-HSPB1 axis in HCC and highlights the potential of targeting COPS5/MK2 combined with sorafenib as a promising strategy for treating HCC.
Collapse
Affiliation(s)
- Ai-Ling Luo
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, 510623, China
| | - Wen-Ying Zheng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Qiong Zhang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Yan Yuan
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Mei-Qi Li
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Kai Du
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - An-Ran Gao
- Department of Laboratory Medicine, Shunde Hospital, Guangzhou University of Chinese Medicine, Foshan, 528300, China
| | - Li-Jun Pei
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Jie Xie
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Wen-Hao Chen
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Long Zhang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiu-Zhu Guo
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiao-Ran Yang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Chao Zeng
- Department of Pathology, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518033, China
| | - Guo-Hua Yang
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| | - Min Deng
- Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, 510095, China
| |
Collapse
|
3
|
Howe CS, Chulkina M, Syrcle R, McAninch C, McAninch S, Pinchuk IV, Beswick EJ. MK2 Inhibition in CD4+ T Cells Protects Against IFNγ and IL-17A, Chronic Inflammation, and Fibrosis in Inflammatory Bowel Disease Models. Inflamm Bowel Dis 2025:izaf026. [PMID: 39937137 DOI: 10.1093/ibd/izaf026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND CD4+ T cells contribute to chronic inflammation and fibrosis in inflammatory bowel disease (IBD), but the cellular mechanisms remain elusive. We have found that the mitogen-activated protein kinase 2 (MK2) pathway plays a major role in inflammation and overall pathology in IBD. Thus, here, we examined the role of MK2 in regulating CD4+ T cell responses in IBD models. METHODS Interleukin-10 (IL-10) knockout (KO) mice treated with MK2 inhibitors (MK2i) and CD4-specific MK2 knockdown mice treated with chronic dextran sodium sulfate (DSS) treatments were used to examine inflammation and fibrosis by multiplex array, gene expression, flow cytometry, and histology. Human tissues were treated with MK2i to examine Th1 and Th17 markers. RESULTS IL-10 KO mice treated with MK2i therapeutically showed significantly reduced interferon gamma (IFNγ) and interleukin-17A (IL-17A) and a significantly reduced number of IFNγ+ and IL-17A+ producing CD4+ T cells by flow cytometry. To investigate the direct role of MK2 in CD4+ T cells during IBD, we utilized CD4-specific MK2 knockdown mice in chronic DSS colitis. A decrease in colonic inflammation, IFNγ and IL-17, pro-fibrotic genes, and extracellular matrix deposition was observed in mice with MK2 knockdown in CD4+ T cells compared to control mice. Additionally, IL-17A and IFNγ directly regulated the expression of fibrosis genes in colon tissues. CONCLUSIONS The MK2 pathway regulates inflammatory CD4+ T cells and fibrosis in IBD models and is a potential therapeutic target.
Collapse
Affiliation(s)
- Cody S Howe
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Marina Chulkina
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Ryan Syrcle
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| | - Christina McAninch
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Steven McAninch
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Irina V Pinchuk
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Ellen J Beswick
- Department of Internal Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
4
|
Liu H, Yuan Y, Johnson-Stephenson TK, Jing C, Zhang M, Huang J, Zen K, Li L, Zhu D. Signal regulatory protein α dynamically mediates macrophage polarization facilitated alleviation of ischemic diseases. Cell Biosci 2024; 14:150. [PMID: 39707436 DOI: 10.1186/s13578-024-01325-2] [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: 05/26/2024] [Accepted: 11/14/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND macrophage-targeting therapy of ischemic disease has made progress in clinic trial. However, the role and underlying mechanism of pro-inflammatory or anti-inflammatory polarized macrophages in modulating ischemic diseases remain incompletely understood. RESULTS here we examine the effect of pro-inflammatory (LPS) and anti-inflammatory (IL-4) macrophage on ischemic diseases in a mouse ischemic hindlimb and heart model, and identify that signal regulatory protein α (Sirpα) modulates macrophage polarization induced angiogenesis via promoting phagocytosis or activating HIF1α nucleus relocation in macrophages, respectively. More importantly, the therapeutic effect of polarized macrophages is controlled by Sirpα in a time-dependent manner. Downregulation of macrophage Sirpα at the early-stage or upregulation of macrophage Sirpα at the late-stage of ischemic disease enhances the therapeutic effect. In contrast, increasing Sirpα at the early-stage or decreasing it at the late-stage leads to failure of inducing ischemic disease resilience. Mechanistically, we find that signal transducer and activator of transcription 3 and 6 (Stat3 and Stat6) mediate downregulation (pro-inflammatory polarization) or upregulation (anti-inflammatory polarization) of Sirpα, respectively. CONCLUSION Our results reveal that dynamic regulation of macrophage by Sirpα plays a critical role in alleviating ischemic diseases.
Collapse
Affiliation(s)
- Haiyi Liu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yonghui Yuan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | | | - Chenyang Jing
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Mingchao Zhang
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Jun Huang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ke Zen
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, Nanjing University School of Life Sciences, Nanjing, Jiangsu, China.
| | - Limin Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Dihan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Rauth S, Malafa M, Ponnusamy MP, Batra SK. Emerging Trends in Gastrointestinal Cancer Targeted Therapies: Harnessing Tumor Microenvironment, Immune Factors, and Metabolomics Insights. Gastroenterology 2024; 167:867-884. [PMID: 38759843 PMCID: PMC11793124 DOI: 10.1053/j.gastro.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024]
Abstract
Gastrointestinal (GI) cancers are the leading cause of new cancer cases and cancer-related deaths worldwide. The treatment strategies for patients with GI tumors have focused on oncogenic molecular profiles associated with tumor cells. Recent evidence has demonstrated that the tumor cell functions are modulated by its microenvironment, compromising fibroblasts, extracellular matrices, microbiome, immune cells, and the enteric nervous system. Along with the tumor microenvironment components, alterations in key metabolic pathways have emerged as a hallmark of tumor cells. From these perspectives, this review will highlight the functions of different cellular components of the GI tumor microenvironment and their implications for treatment. Furthermore, we discuss the major metabolic reprogramming in GI tumor cells and how understanding metabolic rewiring could lead to new therapeutic strategies. Finally, we briefly summarize the targeted agents currently being studied in GI cancers. Understanding the complex interplay between tumor cell-intrinsic and -extrinsic factors during tumor progression is critical for developing new therapeutic strategies.
Collapse
Affiliation(s)
- Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska
| | - Mokenge Malafa
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center at Omaha, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center at Omaha, Omaha, Nebraska.
| |
Collapse
|
6
|
Bao X, Sun M, Meng L, Zhang H, Yi X, Zhang P. Applications of pyroptosis activators in tumor immunotherapy. Mater Today Bio 2024; 28:101191. [PMID: 39221221 PMCID: PMC11363858 DOI: 10.1016/j.mtbio.2024.101191] [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: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Contemporary progress in tumor immunotherapy has solidified its role as an effective approach in combating cancer. Nonetheless, the prevalent "immune cold" state within the tumor microenvironment poses a substantial barrier to its efficacy. Addressing this, pyroptosis-a gasdermin-mediated programmed cell death characterized by its inflammatory profile-emerges as a crucial mechanism. It catalyzes the release of vast quantities of pro-inflammatory cytokines and immunogens, potentially transforming immunosuppressive "cold" tumors into reactive "hot" ones. Herein, we will initially present an overview of pyroptosis as a distinct form of cell death, along with its molecular mechanisms. Subsequently, we will focus on introducing how pyroptosis activators are utilized in the field of tumor immunotherapy. Insights gained from applications of pyroptosis activators in tumor immunotherapy could lead to the development of safe and efficient pyroptosis activators, significantly enriching the arsenal for tumor immunotherapy.
Collapse
Affiliation(s)
- Xin Bao
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Mengmeng Sun
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Lingfei Meng
- Department of Nephrology, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Hong Zhang
- Department of Thyroid, The Second Hospital of Jilin University, Changchun, 130061, PR China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Jilin Biomedical Polymers Engineering Laboratory, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| |
Collapse
|
7
|
Wang D, Sun D, Wang X, Peng X, Ji Y, Tang L, He Q, Chen D, Yang Y, Zhou X, Xiong B, Ai J. Remodeling tumor-associated macrophage for anti-cancer effects by rational design of irreversible inhibition of mitogen-activated protein kinase-activated protein kinase 2. MedComm (Beijing) 2024; 5:e634. [PMID: 38988492 PMCID: PMC11233931 DOI: 10.1002/mco2.634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 07/12/2024] Open
Abstract
Mitogen-activated protein kinase-activated protein kinase 2 (MK2) emerges as a pivotal target in developing anti-cancer therapies. The limitations of ATP-competitive inhibitors, due to insufficient potency and selectivity, underscore the urgent need for a covalent irreversible MK2 inhibitor. Our initial analyses of The Cancer Genome Atlas database revealed MK2's overexpression across various cancer types, especially those characterized by inflammation, linking it to poor prognosis and highlighting its significance. Investigating MK2's kinase domain led to the identification of a unique cysteine residue, enabling the creation of targeted covalent inhibitors. Compound 11 was developed, demonstrating robust MK2 inhibition (IC50 = 2.3 nM) and high selectivity. It binds irreversibly to MK2, achieving prolonged signal suppression and reducing pathological inflammatory cytokines in macrophages. Furthermore, compound 11 or MK2 knockdown can inhibit the tumor-promoting macrophage M2 phenotype in vitro and in vivo. In macrophage-rich tumor model, compound 11 notably slowed growth in a dose-dependent manner. These findings support MK2 as a promising anticancer target, especially relevant in cancers fueled by inflammation or dominated by macrophages, and provide compound 11 serving as an invaluable chemical tool for exploring MK2's functions.
Collapse
Affiliation(s)
- Danyi Wang
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Deqiao Sun
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Xiaoyan Wang
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Xia Peng
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Yinchun Ji
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Lu Tang
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
- State Key Laboratory of Chemical BiologyShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
| | - Qichang He
- State Key Laboratory of Chemical BiologyShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
| | - Danqi Chen
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
- State Key Laboratory of Chemical BiologyShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
| | - Ye Yang
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Xuan Zhou
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
| | - Bing Xiong
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
- State Key Laboratory of Chemical BiologyShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
| | - Jing Ai
- State Key Laboratory of Drug ResearchShanghai Institute of Materia MedicaChinese Academy of SciencesShanghaiP. R. China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijingP. R. China
- Shandong Laboratory of Yantai Drug DiscoveryBohai Rim Advanced Research Institute for Drug DiscoveryYantaiP. R. China
| |
Collapse
|
8
|
Chen H, Zhang JH, Hao Q, Wu XL, Guo JX, Huang CX, Zhang J, Xing GS, An ZL, Ling Y, Zhao JG, Bao YN. Analysis of tumor microenvironment alterations in partially responsive rectal cancer patients treated with neoadjuvant chemoradiotherapy. Int J Colorectal Dis 2024; 39:99. [PMID: 38926205 PMCID: PMC11208236 DOI: 10.1007/s00384-024-04672-1] [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] [Accepted: 06/15/2024] [Indexed: 06/28/2024]
Abstract
PURPOSE Achieving a pathologic complete response (pCR) after neoadjuvant chemoradiotherapy (NCRT) remains a challenge for most patients with rectal cancer. Exploring the potential of combining NCRT with immunotherapy or targeted therapy for those achieving a partial response (PR) offers a promising avenue to enhance treatment efficacy. This study investigated the impact of NCRT on the tumor microenvironment in locally advanced rectal cancer (LARC) patients who exhibited a PR. METHODS This was a retrospective, observational study. Five patients demonstrating a PR after neoadjuvant treatment for LARC were enrolled in the study. Biopsy samples before treatment and resected specimens after treatment were stained with a panel of 26 antibodies targeting various immune and tumor-related markers, each labeled with distinct metal tags. The labeled samples were then analyzed using the Hyperion imaging system. RESULTS Heterogeneity within the tumor microenvironment was observed both before and after NCRT. Notably, tumor-associated macrophages, CD4 + T cells, CD8 + T cells, CD56 + natural killer cells, tumor-associated neutrophils, cytokeratin, and E-cadherin exhibited slight increase in abundance within the tumor microenvironment following treatment (change ratios = 0.78, 0.2, 0.27, 0.32, 0.17, 0.46, 0.32, respectively). Conversely, the number of CD14 + monocytes, CD19 + B cells, CD45 + CD4 + T cells, collagen I, α-smooth muscle actin, vimentin, and β-catenin proteins displayed significant decreases post-treatment (change ratios = 1.73, 1.92, 1.52, 1.25, 1.52, 1.12, 2.66, respectively). Meanwhile, Foxp3 + regulatory cells demonstrated no significant change (change ratio = 0.001). CONCLUSIONS NCRT has diverse effects on various components of the tumor microenvironment in LARC patients who achieve a PR after treatment. Leveraging combination therapies may optimize treatment outcomes in this patient population.
Collapse
Affiliation(s)
- Hong Chen
- Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Ji-Hong Zhang
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Qin Hao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Xin-Lin Wu
- Department of Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Jia-Xing Guo
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Cong-Xiu Huang
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Jun Zhang
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Guo-Sheng Xing
- Department of Gastrointestinal Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Zhi-Lin An
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Yu Ling
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Jian-Guo Zhao
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China
| | - Ying-Na Bao
- Department of Radiotherapy, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010050, China.
| |
Collapse
|
9
|
Jacenik D. Tumor microenvironment and immune response: A gateway to novel therapies in gastrointestinal cancers. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167203. [PMID: 38688415 DOI: 10.1016/j.bbadis.2024.167203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/02/2024] [Accepted: 04/22/2024] [Indexed: 05/02/2024]
Affiliation(s)
- Damian Jacenik
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Cytobiochemistry, Lodz, Poland.
| |
Collapse
|
10
|
Li S, Sheng J, Zhang D, Qin H. Targeting tumor-associated macrophages to reverse antitumor drug resistance. Aging (Albany NY) 2024; 16:10165-10196. [PMID: 38787372 PMCID: PMC11210230 DOI: 10.18632/aging.205858] [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: 11/29/2023] [Accepted: 04/22/2024] [Indexed: 05/25/2024]
Abstract
Currently, antitumor drugs show limited clinical outcomes, mainly due to adaptive resistance. Clinical evidence has highlighted the importance of the tumor microenvironment (TME) and tumor-associated macrophages (TAMs) in tumor response to conventional antitumor drugs. Preclinical studies show that TAMs following antitumor agent can be reprogrammed to an immunosuppressive phenotype and proangiogenic activities through different mechanisms, mediating drug resistance and poor prognosis. Potential extrinsic inhibitors targeting TAMs repolarize to an M1-like phenotype or downregulate proangiogenic function, enhancing therapeutic efficacy of anti-tumor therapy. Moreover, pharmacological modulation of macrophages that restore the immune stimulatory characteristics is useful to reshaping the tumor microenvironment, thus further limiting tumor growth. This review aims to introduce macrophage response in tumor therapy and provide a potential therapeutic combination strategy of TAM-targeting immunomodulation with conventional antitumor drugs.
Collapse
Affiliation(s)
- Sheng Li
- The Second Hospital of Jilin University, Changchun, China
| | - Jiyao Sheng
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Dan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Second Hospital of Jilin University, Changchun, China
| | - Hanjiao Qin
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Zhou H, Yu CY, Wei H. Liposome-based nanomedicine for immune checkpoint blocking therapy and combinatory cancer therapy. Int J Pharm 2024; 652:123818. [PMID: 38253269 DOI: 10.1016/j.ijpharm.2024.123818] [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: 09/21/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
The discovery of immune checkpoint (IC) has led to a wave of leap forward in cancer immunotherapy that represents probably the most promising strategy for cancer therapy. However, the clinical use of immune checkpoint block (ICB) therapy is limited by response rates and side effects. A strategy that addresses the limitations of ICB therapies through combination therapies, using nanocarriers as mediators, has been mentioned in numerous research papers. Liposomes have been probably one of the most extensively used nanocarriers for clinical applications, with broad drug delivery and high safety. A timely review on this hot subject of research, i.e., the application of liposomes for ICB, is thus highly desirable for both fundamental and clinical translatable studies, but remains, to our knowledge, unexplored so far. For this purpose, this review is composed to address the dilemma of ICB therapy and the reasons for this dilemma. We later describe how other cancer treatments have broken this dilemma. Finally, we focus on the role of liposomes in various combinatory cancer therapy. This review is believed to serve as a guidance for the rational design and development of liposome for immunotherapy with enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Haoyuan Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| |
Collapse
|
12
|
Sun J, Wu S, Zhao W, Xue S, Zhang L, Ren J. MAPK-activated protein kinase 2 is associated with poor prognosis of glioma patients and immune inhibition in glioma. Front Oncol 2024; 14:1307992. [PMID: 38322416 PMCID: PMC10844562 DOI: 10.3389/fonc.2024.1307992] [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: 10/05/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024] Open
Abstract
Introduction An effective therapeutic method to noticeably improve the prognosis of glioma patients has not been developed thus far. MAPK-activated protein kinase 2 (MAPKAPK2) is a serine/threonine kinase, which is involved in tumorigenesis, tumor growth, metastasis, and the inflammatory process. The clinical significance and molecular function of MAPKAPK2 in glioma remain unclear. Methods MAPKAPK2 expression in human glioma tissues was detected by immunohistochemistry and analyzed from the transcriptome sequencing data in TCGA and CGGA. Prognostic nomogram was constructed to predict the survival risk of individual patients. GO and KEGG enrichment analyses were performed to analyze the function and pathways MAPKAPK2 involved. Single-cell RNA sequencing data was used to analyze the cell types in which MAPKAPK2 was enriched. Flow cytometry was used for cell cycle and apoptosis detection. The ability of cell proliferation and migration was analyzed by CCK8 and cell migration assay, respectively. Correlation analyses were performed to analyze the relationship of MAPKAPK2 with immune infiltration, immune regulators, chemokine, and chemokine receptors. Results MAPKAPK2 was not only aberrantly upregulated in glioma tissues but also correlated with poor clinical characteristics. Moreover, MAPKAPK2 was prevalent in isocitrate dehydrogenase (IDH) wild-type and 1p/19q non-codeletion glioma cohorts and predicted poor prognosis of glioma patients. MAPKAPK2 may be involved in cell proliferation, cell migration, DNA damage repair, and immune regulation in glioma. MAPKAPK2 was enriched in microglia/macrophages and malignant tumor cells. Further investigation into cellular function revealed that inhibiting MAPKAPK2 suppressed the proliferation and migration of glioblastoma multiforme (GBM) cells in vitro. The inhibition of MAPKAPK2 significantly induced the G1 cell cycle arrest and cell apoptosis of GBM cells. Consistent with the enriched function of MAPKAPK2 in immune regulation, MAPKAPK2 was correlated with immune cell infiltration in glioma tissues. Mechanistically, a series of immune regulators, immunomodulatory chemokine, and chemokine receptors were positively correlated with MAPKAPK2 expression. Discussion Our findings provide evidence of the clinical relevance of MAPKAPK2 in prognosis evaluation of glioma patients and highlight the underlying significance of MAPKAPK2 in glioma therapy.
Collapse
Affiliation(s)
- Jinmin Sun
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sicheng Wu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wenyu Zhao
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Laboratory of Clinical and Experimental Pathology, Department of Pathology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Senrui Xue
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lei Zhang
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jing Ren
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
13
|
Klapp V, Álvarez-Abril B, Leuzzi G, Kroemer G, Ciccia A, Galluzzi L. The DNA Damage Response and Inflammation in Cancer. Cancer Discov 2023; 13:1521-1545. [PMID: 37026695 DOI: 10.1158/2159-8290.cd-22-1220] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/27/2023] [Accepted: 02/23/2023] [Indexed: 04/08/2023]
Abstract
Genomic stability in normal cells is crucial to avoid oncogenesis. Accordingly, multiple components of the DNA damage response (DDR) operate as bona fide tumor suppressor proteins by preserving genomic stability, eliciting the demise of cells with unrepairable DNA lesions, and engaging cell-extrinsic oncosuppression via immunosurveillance. That said, DDR sig-naling can also favor tumor progression and resistance to therapy. Indeed, DDR signaling in cancer cells has been consistently linked to the inhibition of tumor-targeting immune responses. Here, we discuss the complex interactions between the DDR and inflammation in the context of oncogenesis, tumor progression, and response to therapy. SIGNIFICANCE Accumulating preclinical and clinical evidence indicates that DDR is intimately connected to the emission of immunomodulatory signals by normal and malignant cells, as part of a cell-extrinsic program to preserve organismal homeostasis. DDR-driven inflammation, however, can have diametrically opposed effects on tumor-targeting immunity. Understanding the links between the DDR and inflammation in normal and malignant cells may unlock novel immunotherapeutic paradigms to treat cancer.
Collapse
Affiliation(s)
- Vanessa Klapp
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Tumor Stroma Interactions, Department of Cancer Research, Luxembourg Institute of Health, Luxembourg, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Beatriz Álvarez-Abril
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Department of Hematology and Oncology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Giuseppe Leuzzi
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Alberto Ciccia
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York
- Herbert Irving Comprehensive Cancer Center, New York, New York
- Institute for Cancer Genetics, Columbia University Irving Medical Center, New York, New York
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, New York
- Sandra and Edward Meyer Cancer Center, New York, New York
- Caryl and Israel Englander Institute for Precision Medicine, New York, New York
| |
Collapse
|
14
|
Jang H, Ojha U, Jeong JH, Park KG, Lee SY, Lee YM. Myriocin suppresses tumor growth by modulating macrophage polarization and function through the PI3K/Akt/mTOR pathway. Arch Pharm Res 2023; 46:629-645. [PMID: 37468765 DOI: 10.1007/s12272-023-01454-1] [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: 02/21/2023] [Accepted: 07/02/2023] [Indexed: 07/21/2023]
Abstract
Macrophages within the tumor microenvironment (TME), referred to as tumor-associated macrophages (TAMs), are involved in various aspects of tumor progression including initiation, angiogenesis, metastasis, immunosuppression, and resistance to therapy. Myriocin, a natural compound isolated from Mycelia sterilia, is an immunosuppressant that inhibits tumor growth and angiogenesis. However, the mechanisms underlying the effects of myriocin on TAMs and TAM-mediated tumor growth have not yet been elucidated. In this study, we determined the effects of myriocin on TAMs and the underlying mechanism in vitro and in vivo. Myriocin significantly suppressed monocyte-macrophage differentiation and M2 polarization of macrophages but not M1 polarization. In addition, myriocin inhibited the expression of anti-inflammatory cytokines and secretion of proangiogenic factors, such as vascular endothelial growth factor, in M2 macrophages as well as M2-induced endothelial cell permeability. Myriocin also inhibited the PI3K/Akt/mTOR signaling pathway in M2 macrophages. Myriocin reduced the population of M2-like TAMs within the tumor tissue of a mouse allograft tumor model but not that of M1-like TAMs. Moreover, combined treatment with myriocin and cisplatin synergistically suppressed tumor growth and enhanced survival rate in mice by reducing the population of M2-like TAMs. Overall, these results suggest that myriocin inhibits tumor growth by remodeling the TME through suppression of differentiation and polarization of M2-like TAMs via the PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Hyeonha Jang
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Uttam Ojha
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ji-Hak Jeong
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Keun-Gyu Park
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - Shin Yup Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, 41944, Republic of Korea
| | - You Mie Lee
- Vessel-Organ Interaction Research Center (VOICE, MRC), College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea.
- College of Pharmacy, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
15
|
Jacenik D, Lebish EJ, Beswick EJ. MK2 drives progression of pancreas and colon cancers by suppressing CD8 + T cell cytotoxic function and is a potential immunotherapy target. Front Immunol 2023; 14:1212100. [PMID: 37415974 PMCID: PMC10321668 DOI: 10.3389/fimmu.2023.1212100] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/01/2023] [Indexed: 07/08/2023] Open
Abstract
Background Immune cell composition is a critical and dynamic component of the tumor microenvironment, which has an impact on immunosuppression and progression of cancer. T cells, especially CD8+ T cells, are one of the major immune cell types responsible for tumor cell killing employing receptor-ligand mediated apoptosis and/or releasing lytic granules among others. Accumulating evidence highlighted that adoptive transfer of activated and/or modified immune cells can enhance anti-tumorigenic immune responses and serve as promising therapy approach for patients with cancers. The mitogen-activated protein kinase-activated protein kinase 2 (MK2) is a serine/threonine protein kinase, which controls production and secretion of numerous pro-inflammatory cytokines and chemokines involved in tumorigenesis. However, limited efforts have been made to learn how MK2 may affects CD8+ T cell action and function in the tumor microenvironment especially in gastrointestinal cancers. Methods To explore the therapeutic potential of MK2 in the immune response mediated by CD8+ T cells, RAG1 knockout mice with PK5L1940 and BRAF cells-derived allograft tumors were treated with WT or MK2 knockout CD8+ T cells. The phenotype of CD8+ T cells with MK2 depletion were evaluated in vitro. Immunofluorescence staining, real-time PCR and multiplex analysis were utilized to estimate the expression of apoptotic and lytic factors. Results Here, we show that CD8+ T cells with MK2 depletion prevent gastrointestinal cancer growth, which is accompanied by enhanced expression and secretion of factors related to apoptosis. Moreover, using in vitro and in vivo approaches, we found that depletion of MK2 lead to hyperactivation of CD8+ T cells and enhanced anti-tumor immunity. Conclusion Overall, we documented that MK2 drives the progression of gastrointestinal cancers and prevents immune response generated by CD8+ T cells suggesting potential implications of MK2 in the immunotherapy of gastrointestinal cancers.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Eric J. Lebish
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
| | - Ellen J. Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, UT, United States
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
16
|
Zhao X, Fu L, Zou H, He Y, Pan Y, Ye L, Huang Y, Fan W, Zhang J, Ma Y, Chen J, Zhu M, Zhang C, Cai Y, Mou X. Optogenetic engineered umbilical cord MSC-derived exosomes for remodeling of the immune microenvironment in diabetic wounds and the promotion of tissue repair. J Nanobiotechnology 2023; 21:176. [PMID: 37269014 DOI: 10.1186/s12951-023-01886-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/06/2023] [Indexed: 06/04/2023] Open
Abstract
BACKGROUND Angiogenesis and tissue repair in chronic non-healing diabetic wounds remain critical clinical problems. Engineered MSC-derived exosomes have significant potential for the promotion of wound healing. Here, we discuss the effects and mechanisms of eNOS-rich umbilical cord MSC exosomes (UCMSC-exo/eNOS) modified by genetic engineering and optogenetic techniques on diabetic chronic wound repair. METHODS Umbilical cord mesenchymal stem cells were engineered to express two recombinant proteins. Large amounts of eNOS were loaded into UCMSC-exo using the EXPLOR system under blue light irradiation. The effects of UCMSC-exo/eNOS on the biological functions of fibroblasts and vascular endothelial cells in vitro were evaluated. Full-thickness skin wounds were constructed on the backs of diabetic mice to assess the role of UCMSC-exo/eNOS in vascular neogenesis and the immune microenvironment, and to explore the related molecular mechanisms. RESULTS eNOS was substantially enriched in UCMSCs-exo by endogenous cellular activities under blue light irradiation. UCMSC-exo/eNOS significantly improved the biological functions of cells after high-glucose treatment and reduced the expression of inflammatory factors and apoptosis induced by oxidative stress. In vivo, UCMSC-exo/eNOS significantly improved the rate of wound closure and enhanced vascular neogenesis and matrix remodeling in diabetic mice. UCMSC-exo/eNOS also improved the inflammatory profile at the wound site and modulated the associated immune microenvironment, thus significantly promoting tissue repair. CONCLUSION This study provides a novel therapeutic strategy based on engineered stem cell-derived exosomes for the promotion of angiogenesis and tissue repair in chronic diabetic wounds.
Collapse
Affiliation(s)
- Xin Zhao
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
- College of Pharmacy, Hangzhou Medical College, Hangzhou, 310059, China
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, China
| | - Luoqin Fu
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Hai Zou
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yichen He
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yi Pan
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Luyi Ye
- College of Pharmacy, Hangzhou Medical College, Hangzhou, 310059, China
| | - Yilin Huang
- College of Pharmacy, Hangzhou Medical College, Hangzhou, 310059, China
| | - Weijiao Fan
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Jungang Zhang
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Yingyu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China
| | - Jinyang Chen
- Zhejiang Healthfuture Biomedicine Co., Ltd., Hangzhou, 310052, China
| | - Mingang Zhu
- Department of Dermatology, the First People's Hospital of Jiashan, Jiaxing, 314100, Zhejiang, China
| | - Chengwu Zhang
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, China.
| | - Yu Cai
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
- College of Pharmacy, Hangzhou Medical College, Hangzhou, 310059, China.
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, China.
| | - Xiaozhou Mou
- General Surgery, Cancer Center, Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
- College of Pharmacy, Hangzhou Medical College, Hangzhou, 310059, China.
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, China.
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou, 310014, China.
| |
Collapse
|
17
|
Faget DV, Luo X, Inkman MJ, Ren Q, Su X, Ding K, Waters MR, Raut GK, Pandey G, Dodhiawala PB, Ramalho-Oliveira R, Ye J, Cole T, Murali B, Zheleznyak A, Shokeen M, Weiss KR, Monahan JB, DeSelm CJ, Lee AV, Oesterreich S, Weilbaecher KN, Zhang J, DeNardo DG, Stewart SA. p38MAPKα Stromal Reprogramming Sensitizes Metastatic Breast Cancer to Immunotherapy. Cancer Discov 2023; 13:1454-1477. [PMID: 36883955 PMCID: PMC10238649 DOI: 10.1158/2159-8290.cd-22-0907] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 01/05/2023] [Accepted: 02/14/2023] [Indexed: 03/09/2023]
Abstract
Metastatic breast cancer is an intractable disease that responds poorly to immunotherapy. We show that p38MAPKα inhibition (p38i) limits tumor growth by reprogramming the metastatic tumor microenvironment in a CD4+ T cell-, IFNγ-, and macrophage-dependent manner. To identify targets that further increased p38i efficacy, we utilized a stromal labeling approach and single-cell RNA sequencing. Thus, we combined p38i and an OX40 agonist that synergistically reduced metastatic growth and increased overall survival. Intriguingly, patients with a p38i metastatic stromal signature had better overall survival that was further improved by the presence of an increased mutational load, leading us to ask if our approach would be effective in antigenic breast cancer. The combination of p38i, anti-OX40, and cytotoxic T-cell engagement cured mice of metastatic disease and produced long-term immunologic memory. Our findings demonstrate that a detailed understanding of the stromal compartment can be used to design effective antimetastatic therapies. SIGNIFICANCE Immunotherapy is rarely effective in breast cancer. We dissected the metastatic tumor stroma, which revealed a novel therapeutic approach that targets the stromal p38MAPK pathway and creates an opportunity to unleash an immunologic response. Our work underscores the importance of understanding the tumor stromal compartment in therapeutic design. This article is highlighted in the In This Issue feature, p. 1275.
Collapse
Affiliation(s)
- Douglas V. Faget
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Xianmin Luo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Matthew J. Inkman
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Qihao Ren
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Xinming Su
- Department of Medicine, Washington University School of Medicine, St Louis, MO
| | - Kai Ding
- Womens Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Magee-Womens Research Institute, Pittsburgh, PA
- Integrative Systems Biology Graduate Program, University of Pittsburgh, Pittsburgh, PA
| | - Michael R. Waters
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Ganesh Kumar Raut
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Gaurav Pandey
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Paarth B. Dodhiawala
- Department of Medicine, Washington University School of Medicine, St Louis, MO
- Medical Scientist Training Program, University of Minnesota Medical School, Minneapolis, MN
- ICCE Institute, Washington University School of Medicine, St Louis, MO
| | - Renata Ramalho-Oliveira
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Jiayu Ye
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Thomas Cole
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Bhavna Murali
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
| | - Alexander Zheleznyak
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Monica Shokeen
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
- Department of Biomedical Engineering, Washington University School of Medicine, St Louis, MO
| | - Kurt R. Weiss
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA
| | | | - Carl J. DeSelm
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Adrian V. Lee
- Womens Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Magee-Womens Research Institute, Pittsburgh, PA
- Department of Pharmacology and Chemical Biology & Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA
| | - Steffi Oesterreich
- Womens Cancer Research Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Magee-Womens Research Institute, Pittsburgh, PA
- Department of Pharmacology and Chemical Biology & Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA
| | - Katherine N. Weilbaecher
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
- Department of Medicine, Washington University School of Medicine, St Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Jin Zhang
- Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
- Institute for Informatics (I), Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - David G. DeNardo
- Department of Medicine, Washington University School of Medicine, St Louis, MO
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO
- ICCE Institute, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| | - Sheila A. Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, MO
- Department of Medicine, Washington University School of Medicine, St Louis, MO
- ICCE Institute, Washington University School of Medicine, St Louis, MO
- Siteman Cancer Center, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
18
|
Wu Z, Fang ZX, Hou YY, Wu BX, Deng Y, Wu HT, Liu J. Exosomes in metastasis of colorectal cancers: Friends or foes? World J Gastrointest Oncol 2023; 15:731-756. [PMID: 37275444 PMCID: PMC10237026 DOI: 10.4251/wjgo.v15.i5.731] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/07/2023] [Accepted: 04/04/2023] [Indexed: 05/12/2023] Open
Abstract
Colorectal cancer (CRC), the third most common type of cancer worldwide, threaten human health and quality of life. With multidisciplinary, including surgery, chemotherapy and/or radiotherapy, patients with an early diagnosis of CRC can have a good prognosis. However, metastasis in CRC patients is the main risk factor causing cancer-related death. To elucidate the underlying molecular mechanisms of CRC metastasis is the difficult and research focus on the investigation of the CRC mechanism. On the other hand, the tumor microenvironment (TME) has been confirmed as having an essential role in the tumorigenesis and metastasis of malignancies, including CRCs. Among the different factors in the TME, exosomes as extracellular vesicles, function as bridges in the communication between cancer cells and different components of the TME to promote the progression and metastasis of CRC. MicroRNAs packaged in exosomes can be derived from different sources and transported into the TME to perform oncogenic or tumor-suppressor roles accordingly. This article focuses on CRC exosomes and illustrates their role in regulating the metastasis of CRC, especially through the packaging of miRNAs, to evoke exosomes as novel biomarkers for their impact on the metastasis of CRC progression.
Collapse
Affiliation(s)
- Zheng Wu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Ze-Xuan Fang
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yan-Yu Hou
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Bing-Xuan Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Yu Deng
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Hua-Tao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| | - Jing Liu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, Guangdong Province, China
| |
Collapse
|
19
|
Jin Y, Fu L. Engineer a double team of short-lived and glucose-sensing bacteria for cancer eradication. Cell Rep Med 2023:101043. [PMID: 37192627 DOI: 10.1016/j.xcrm.2023.101043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 02/13/2023] [Accepted: 04/19/2023] [Indexed: 05/18/2023]
Abstract
Rationally designed and engineered bacteria represent an emerging unique approach for cancer treatment. Here, we engineer a short-lived bacterium, mp105, that is effective against diverse cancer types and safe for intravenous administration. We reveal that mp105 combats cancer by direct oncolysis, depletion of tumor-associated macrophages, and elicitation of CD4+ T cell immunity. We further engineer a glucose-sensing bacterium named m6001 that selectively colonizes solid tumors. When intratumorally injected, m6001 clears tumors more efficiently than mp105 due to its post-delivery replication in tumors and potent oncolytic capacity. Finally, we combine intravenous injection of mp105 and intratumoral injection of m6001, forming a double team against cancer. The double team enhances cancer therapy compared with single treatment for subjects carrying both intratumorally injectable and uninjectable tumors. The two anticancer bacteria and their combination are applicable to different scenarios, turning bacterial therapy for cancer into a feasible solution.
Collapse
Affiliation(s)
- Ye Jin
- New Portal Limited, 130-132 Des Voeux Road Central, Hong Kong.
| | - Li Fu
- Guangdong Province Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| |
Collapse
|
20
|
Li L, Tian Y. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023; 161:114504. [PMID: 37002579 DOI: 10.1016/j.biopha.2023.114504] [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: 12/07/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Macrophages are potent immune effector cells in innate immunity and exert dual-effects in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) make up a significant portion of TME immune cells. Similar to M1/M2 macrophages, TAMs are also highly plastic, and their functions are regulated by cytokines, chemokines and other factors in the TME. The metabolic changes in TAMs are significantly associated with polarization towards a protumour or antitumour phenotype. The metabolites generated via TAM metabolic reprogramming in turn promote tumor progression and immune tolerance. In this review, we explore the metabolic reprogramming of TAMs in terms of energy, amino acid and fatty acid metabolism and the potential roles of these changes in immune suppression.
Collapse
Affiliation(s)
- Lunxu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
21
|
Shen Y, Chen JX, Li M, Xiang Z, Wu J, Wang YJ. Role of tumor-associated macrophages in common digestive system malignant tumors. World J Gastrointest Oncol 2023; 15:596-616. [PMID: 37123058 PMCID: PMC10134211 DOI: 10.4251/wjgo.v15.i4.596] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 02/12/2023] [Accepted: 03/30/2023] [Indexed: 04/12/2023] Open
Abstract
Many digestive system malignant tumors are characterized by high incidence and mortality rate. Increasing evidence has revealed that the tumor microenvironment (TME) is involved in cancer initiation and tumor progression. Tumor-associated macrophages (TAMs) are a predominant constituent of the TME, and participate in the regulation of various biological behaviors and influence the prognosis of digestive system cancer. TAMs can be mainly classified into the antitumor M1 phenotype and protumor M2 phenotype. The latter especially are crucial drivers of tumor invasion, growth, angiogenesis, metastasis, immunosuppression, and resistance to therapy. TAMs are of importance in the occurrence, development, diagnosis, prognosis, and treatment of common digestive system malignant tumors. In this review, we summarize the role of TAMs in common digestive system malignant tumors, including esophageal, gastric, colorectal, pancreatic and liver cancers. How TAMs promote the development of tumors, and how they act as potential therapeutic targets and their clinical applications are also described.
Collapse
Affiliation(s)
- Yue Shen
- Department of Dermatology, Suzhou Municipal Hospital, Suzhou 215008, Jiangsu Province, China
| | - Jia-Xi Chen
- School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Ming Li
- Department of Pathology, Suzhou Municipal Hospital, Suzhou 215008, Jiangsu Province, China
| | - Ze Xiang
- School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang Province, China
| | - Jian Wu
- Department of Clinical Laboratory, Suzhou Municipal Hospital, Suzhou 215008, Jiangsu Province, China
| | - Yi-Jin Wang
- School of Medicine, Southern University of Science and Technology, Shenzhen 518055, Guangdong Province, China
| |
Collapse
|
22
|
Zhou Y, Oki R, Tanaka A, Song L, Takashima A, Hamada N, Yokoyama S, Yano S, Sakurai H. Cellular stress induces non-canonical activation of the receptor tyrosine kinase EphA2 through the p38-MK2-RSK signaling pathway. J Biol Chem 2023; 299:104699. [PMID: 37059179 DOI: 10.1016/j.jbc.2023.104699] [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: 09/28/2022] [Revised: 04/01/2023] [Accepted: 04/03/2023] [Indexed: 04/16/2023] Open
Abstract
The receptor tyrosine kinase EphA2 is overexpressed in malignant tumors. We previously reported that non-canonical EphA2 phosphorylation at Ser-897 was catalyzed by p90 ribosomal S6 kinase (RSK) via the MEK-ERK pathway in ligand- and tyrosine kinase-independent manners. Non-canonical EphA2 activation plays a key role in tumor progression; however, its activation mechanism remains unclear. In the present study, we focused on cellular stress signaling as a novel inducer of non-canonical EphA2 activation. p38, instead of ERK in the case of epidermal growth factor signaling, activated RSK-EphA2 under cellular stress conditions, including anisomycin, cisplatin and high osmotic stress. Notably, p38 activated the RSK-EphA2 axis via downstream MAPK-activated protein kinase 2 (MK2). Furthermore, MK2 directly phosphorylated both RSK1 Ser-380 and RSK2 Ser-386, critical residues for the activation of their N-terminal kinases, which is consistent with the result showing that the C-terminal kinase domain of RSK1 was dispensable for MK2-mediated EphA2 phosphorylation. Moreover, the p38-MK2-RSK-EphA2 axis promoted glioblastoma cell migration induced by temozolomide, a chemotherapeutic agent for the treatment of glioblastoma patients. Collectively, the present results reveal a novel molecular mechanism for non-canonical EphA2 activation under stress conditions in the tumor microenvironment.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Ryota Oki
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Akihiro Tanaka
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Leixin Song
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Atsushi Takashima
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Naru Hamada
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Satoru Yokoyama
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan
| | - Seiji Yano
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Takara-Machi, Kanazawa, Ishikawa 920-0934, Japan
| | - Hiroaki Sakurai
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama; Toyama 930-0194, Japan.
| |
Collapse
|
23
|
Michaelides I, Künzel J, Ettl T, Beckhove P, Bohr C, Brochhausen C, Mamilos A. Adenoid cystic carcinoma of the salivary glands: a pilot study of potential therapeutic targets and characterization of the immunological tumor environment and angiogenesis. Eur Arch Otorhinolaryngol 2023; 280:2937-2944. [PMID: 36856809 PMCID: PMC10175421 DOI: 10.1007/s00405-023-07884-3] [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: 07/31/2022] [Accepted: 02/14/2023] [Indexed: 03/02/2023]
Abstract
BACKGROUND Adenoid cystic carcinoma (ACC) is a rare type of cancer commonly occurring in salivary glands. It is characterized by slow but infiltrative growth, nerve infiltration and overall poor prognosis, with late recurrence and distant metastasis. The treatment of ACC is still limited to surgery and/or (adjuvant) radiotherapy. Till now no promising systemic therapy option exists. However, various studies deliver promising results after treatment with anti-angiogenetic agents, such as anti-EGFR-antibody Cetuximab or Tyrosinkinase inhibitor Lenvatinib. METHODS By using of immunohistological methods we analyzed and compared the macrophage and lymphocyte populations, vascularization, and PD-L1-status in 12 ACC of the salivary glands. RESULTS All cases showed a significant elevation of macrophages with M2 polarization and a higher vascularization in ACC compared to normal salivary gland tissue. The CD4/CD8 quotient was heterogenous. ACC does not show relevant PD-L1 expression. CONCLUSIONS The predominant M2 polarization of macrophages in ACC could be responsible for elevated vascularization, as already been proved in other cancer types, that M2 macrophages promote angiogenesis.
Collapse
Affiliation(s)
- Ioannis Michaelides
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany.
| | - Julian Künzel
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Tobias Ettl
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Philipp Beckhove
- Division of Interventional Immunology, RCI Regensburg Center for Interventional Immunology, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Christopher Bohr
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | | | - Andreas Mamilos
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
24
|
Deng S, Zhu Q, Chen H, Xiao T, Zhu Y, Gao J, Li Q, Gao Y. Screening of prognosis-related Immune cells and prognostic predictors in Colorectal Cancer Patients. BMC Cancer 2023; 23:195. [PMID: 36859111 PMCID: PMC9976376 DOI: 10.1186/s12885-023-10667-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/21/2023] [Indexed: 03/03/2023] Open
Abstract
OBJECTIVE To accurately screen potential immune cells that can predict the survival of colorectal cancer (CRC) patients and identify related prognostic predictors. METHODS The sample data of CRC patients were downloaded from the GEO database as a training set to establish a prognosis-scoring model and screen prognosis-related immune cells. The sample data of CRC patients from the TCGA database were used as the validation set. Simultaneously, cancer tissue samples from 116 patients with CRC diagnosed pathologically in Shanghai Dongfang Hospital were collected to analyze the relationship of prognosis-related immune cells with patients' survival, and clinical and pathological parameters, and to screen prognostic predictors. RESULTS Prognosis-related immune cells screened from GEO and TCGA databases mainly included Follicular Helper T cells (Tfh), Monocytes and M2 Macrophages. In the training set, the 2,000- and 4,000-day survival rates were 48.3% and 10.7% in the low-risk group (N = 234), and 42.1% and 7.5% in the high-risk group (N = 214), respectively. In the validation set, the 2,000- and 4,000-day survival rates were 34.8% and 8.6% in the low-risk group (N = 187), and 28.9% and 6.1% in the high-risk group (N = 246), respectively. The prognosis of patients in the high-risk group was worse than that in the low-risk group (P < 0.05). Furthermore, the screened primary prognostic predictors were CD163 and CD4 + CXCR5. CD163 protein expression was distributed in Monocytes and M2 Macrophages. The 1,000- and 2,000-day survival rates were 56.1% and 7.0% in the CD163 low-expression group, and 40.7% and 1.7% in the high-expression group (N = 214), respectively, showing a worse prognosis in the high-expression group than that in the low-expression group. Meanwhile, the immune marker CD4 + CXCR5 could identify Tfh. The 1,000- and 2,000-day survival rates were 63.9% and 5.6% in the CD4 + CXCR5 high-expression group, and 33.3% and 2.8% in the low-expression group (N = 214), respectively, with a better prognosis in the high-expression group than that in the low-expression group. CONCLUSION Prognostic-related immune cells of CRC mainly include Tfh cells, Monocytes and M2 Macrophages. Monocytes and M2 Macrophages correlate negatively, while Tfh cells correlate positively with the prognosis of CRC patients. Immune markers CD163 and CD4 + CXCR5 can be considered as the prognostic predictors of CRC with clinical value of the application.
Collapse
Affiliation(s)
- Shuangshuang Deng
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qiping Zhu
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Hongyan Chen
- Department of Neurology, Luodian Hospital, Baoshan District, Shanghai, 201908, China
| | - Tianyu Xiao
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yinshen Zhu
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jinli Gao
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Qing Li
- Department of Pathology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
25
|
Wang F, Niu Y, Chen K, Yuan X, Qin Y, Zheng F, Cui Z, Lu W, Wu Y, Xia D. Extracellular Vesicle-Packaged circATP2B4 Mediates M2 Macrophage Polarization via miR-532-3p/SREBF1 Axis to Promote Epithelial Ovarian Cancer Metastasis. Cancer Immunol Res 2023; 11:199-216. [PMID: 36512324 PMCID: PMC9896028 DOI: 10.1158/2326-6066.cir-22-0410] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/07/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022]
Abstract
Ovarian cancer is one of the most common gynecologic malignancies with a highly immunosuppressive tumor microenvironment (TME) and poor prognosis. Circular RNA (circRNA) is a type of noncoding RNA with high stability, which has been shown to play an important role in biological processes and TME reprogramming in a variety of tumors. The biological function of a novel circRNA, circATP2B4, in epithelial ovarian cancer (EOC) was detected and evaluated. Transmission electron microscopy, differential ultracentrifugation and qRT-PCR were used to verify the existence of extracellular vesicles (EV)-packaged circATP2B4. Macrophage uptake of circATP2B4 was determined by EVs tracing. Dual luciferase reporter, FISH, Western blotting, and flow cytometry assays were used to investigate the interactions between circATP2B4 and miR-532-3p as well as sterol regulatory element-binding factor 1 (SREBF1) expression in macrophages. CircATP2B4 was upregulated in EOC tissues and positively correlated with ovarian cancer progression. Functionally, circATP2B4 promoted carcinogenic progression and metastasis of EOC both in vitro and in vivo. Mechanistically, EV-packaged circATP2B4 in EOC could be transmitted to infiltrated macrophages and acted as competing endogenous RNA of miR-532-3p to relieve the repressive effect of miR-532-3p on its target SREBF1. Furthermore, circATP2B4 induced macrophage M2 polarization by regulating the miR-532-3p/SREBF1/PI3Kα/AKT axis, thereby leading to immunosuppression and ovarian cancer metastasis. Collectively, these data indicate that circATP2B4-containing EVs generated by EOC cells promoted M2 macrophages polarization and malignant behaviors of EOC cells. Thus, targeting EVs-packaged circATP2B4 may provide a potential diagnosis and treatment strategy for ovarian cancer.
Collapse
Affiliation(s)
- Fang Wang
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuequn Niu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kelie Chen
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyu Yuan
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuheng Qin
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Zheng
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhenyan Cui
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weiguo Lu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yihua Wu
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Sciences, Hangzhou, China
| | - Dajing Xia
- Department of Toxicology of School of Public Health and Department of Gynecologic Oncology of Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Cancer Center, Zhejiang University, Hangzhou, China
| |
Collapse
|
26
|
Wu W, Liu S, Tian L, Li C, Jiang Y, Wang J, Lv Y, Guo J, Xing D, Zhai Y, Sun H, Li Y, Zhang L, He X, Luo K, Zhan H, Zhao Z. Identification of microtubule-associated biomarkers in diffuse large B-cell lymphoma and prognosis prediction. Front Genet 2023; 13:1092678. [PMID: 36761693 PMCID: PMC9902697 DOI: 10.3389/fgene.2022.1092678] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/28/2022] [Indexed: 01/25/2023] Open
Abstract
Background: Diffuse large B-cell lymphoma (DLBCL) is a genetically heterogeneous disease with a complicated prognosis. Even though various prognostic evaluations have been applied currently, they usually only use the clinical factors that overlook the molecular underlying DLBCL progression. Therefore, more accurate prognostic assessment needs further exploration. In the present study, we constructed a novel prognostic model based on microtubule associated genes (MAGs). Methods: A total of 33 normal controls and 1360 DLBCL samples containing gene-expression from the Gene Expression Omnibus (GEO) database were included. Subsequently, the univariate Cox, the least absolute shrinkage and selection operator (LASSO), and multivariate Cox regression analysis were used to select the best prognosis related genes into the MAGs model. To validate the model, Kaplan-Meier curve, and nomogram were analyzed. Results: A risk score model based on fourteen candidate MAGs (CCDC78, CD300LG, CTAG2, DYNLL2, MAPKAPK2, MREG, NME8, PGK2, RALBP1, SIGLEC1, SLC1A1, SLC39A12, TMEM63A, and WRAP73) was established. The K-M curve presented that the high-risk patients had a significantly inferior overall survival (OS) time compared to low-risk patients in training and validation datasets. Furthermore, knocking-out TMEM63A, a key gene belonging to the MAGs model, inhibited cell proliferation noticeably. Conclusion: The novel MAGs prognostic model has a well predictive capability, which may as a supplement for the current assessments. Furthermore, candidate TMEM63A gene has therapeutic target potentially in DLBCL.
Collapse
Affiliation(s)
- Wenqi Wu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Su Liu
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Linyan Tian
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Cheng Li
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yanan Jiang
- Department of Medical Oncology, Tianjin First Central Hospital, School of Medicine. Nankai University, Tianjin, China
| | - Jinhuan Wang
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yangyang Lv
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Jing Guo
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Donghui Xing
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yixin Zhai
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Huimeng Sun
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Yuhang Li
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Luying Zhang
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Jinan, China
| | - Xiang He
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Kaiping Luo
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Hongjie Zhan
- Department of Gastroenterology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Jinan, China,*Correspondence: Hongjie Zhan, ; Zhigang Zhao,
| | - Zhigang Zhao
- Department of Hematology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China,Department of Medical Oncology, Tianjin First Central Hospital, School of Medicine. Nankai University, Tianjin, China,*Correspondence: Hongjie Zhan, ; Zhigang Zhao,
| |
Collapse
|
27
|
Jacenik D, Lebish EJ, Beswick EJ. MK2 Promotes the Development and Progression of Pancreatic Neuroendocrine Tumors Mediated by Macrophages and Metabolomic Factors. Int J Mol Sci 2022; 23:13561. [PMID: 36362348 PMCID: PMC9658113 DOI: 10.3390/ijms232113561] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 09/24/2023] Open
Abstract
Cases of pancreatic neuroendocrine tumors (PNETs) are growing in number, and new treatment options are needed in order to improve patient outcomes. The mitogen-activated protein kinase-activated protein kinase 2 (MK2) is a crucial regulator of cytokine/chemokine production. The significance of MK2 expression and signaling pathway mediated by MK2 in PNETs has not been investigated. To characterize the impact of MK2 on PNET growth, we used the RipTag2 transgenic murine model of PNETs, and we developed a primary PNET cell line for both in vitro and in vivo studies. In the transgenic murine model of PNETs, we found that MK2 inhibition improves survival of mice and prevents PNET progression. MK2 blockade abolished cytokine/chemokine production, which was related to macrophage function. A role for MK2 in the regulation of metabolic factor secretion in PNETs was identified, making this the first study to identify a potential role for the MK2 pathway in regulation of tumor metabolism. Moreover, using an in vitro approach and allograft model of PNETs, we were able to show that macrophages with MK2 depletion exhibit increased cytotoxicity against PNET cells and substantially decreased production of pro-inflammatory cytokines and chemokines, as well as metabolic factors. Taken together, our work identifies MK2 as a potent driver of immune response and metabolic effectors in PNETs, suggesting it is a potential therapeutic target for patients with PNETs.
Collapse
Affiliation(s)
- Damian Jacenik
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Eric J. Lebish
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| | - Ellen J. Beswick
- Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
| |
Collapse
|
28
|
Dogan NO, Ceylan H, Suadiye E, Sheehan D, Aydin A, Yasa IC, Wild AM, Richter G, Sitti M. Remotely Guided Immunobots Engaged in Anti-Tumorigenic Phenotypes for Targeted Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2204016. [PMID: 36202751 DOI: 10.1002/smll.202204016] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/08/2022] [Indexed: 06/16/2023]
Abstract
Building medical microrobots from the body's own cells may circumvent the biocompatibility concern and hence presents more potential in clinical applications to improve the possibility of escaping from the host defense mechanism. More importantly, live cells can enable therapeutically relevant functions with significantly higher efficiency than synthetic systems. Here, live immune cell-derived microrobots from macrophages, i.e., immunobots, which can be remotely steered with externally applied magnetic fields and directed toward anti-tumorigenic (M1) phenotypes, are presented. Macrophages engulf the engineered magnetic decoy bacteria, composed of 0.5 µm diameter silica Janus particles with one side coated with anisotropic FePt magnetic nanofilm and the other side coated with bacterial lipopolysaccharide (LPS). This study demonstrates the torque-based surface rolling locomotion of the immunobots along assigned trajectories inside blood plasma, over a layer of endothelial cells, and under physiologically relevant flow rates. The immunobots secrete signature M1 cytokines, IL-12 p40, TNF-α, and IL-6, and M1 cell markers, CD80 and iNOS, via toll-like receptor 4 (TLR4)-mediated stimulation with bacterial LPS. The immunobots exhibit anticancer activity against urinary bladder cancer cells. This study further demonstrates such immunobots from freshly isolated primary bone marrow-derived macrophages since patient-derivable macrophages may have a strong clinical potential for future cell therapies in cancer.
Collapse
Affiliation(s)
- Nihal Olcay Dogan
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8092, Switzerland
| | - Hakan Ceylan
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Eylül Suadiye
- Materials Central Scientific Facility, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Devin Sheehan
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Asli Aydin
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Immihan Ceren Yasa
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Anna-Maria Wild
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Gunther Richter
- Materials Central Scientific Facility, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569, Stuttgart, Germany
- Institute for Biomedical Engineering, ETH Zurich, Zurich, 8092, Switzerland
- School of Medicine and College of Engineering, Koç University, Istanbul, 34450, Turkey
| |
Collapse
|
29
|
The Regulating Effect of CII-3 and Its Active Components from Periplaneta americana on M1/M2 Macrophage Polarization. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27144416. [PMID: 35889289 PMCID: PMC9323847 DOI: 10.3390/molecules27144416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/26/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
CII-3 is the effective part of Periplaneta americana for application in oncotherapy. This study investigated its main chemical components for macrophage polarization regulation activity. Compounds were separated and purified, and their structures were elucidated based on NMR and HR-ESI-MS analyses. After inducing the M1 and M2 phenotype macrophages, CII-3 and testing components were added and co-incubated to evaluate their effects on the relevant markers of macrophages. Then, gradient concentrations of CII-3 and active monomers were further investigated for their effects on M2 macrophages. The effects were detected by RT-PCR, ELISA, flow cytometry, and immunofluorescence. Twelve compounds were identified from CII-3. CII-3 and pericanaside (5) had no obvious effect on M1 macrophages, while they significantly reduced the expression levels of M2 macrophage markers. Specifically, they significantly reduced the levels of TGF-β and IL-10 and the mRNA expression levels of ARG-1 and CD206 in the M2 phenotypes of RAW264.7 and Ana-1 macrophages. The conditioned medium of CII-3 and pericanaside (5) could inhibit the migration capacity of CT26.WT tumor cells. Macrophage M1/M2 polarization is a dynamic equilibrium, and the M2 phenotype, which can promote the growth of tumor cells, is relatively highly expressed in the tumor microenvironment. CII-3 and pericanaside could significantly reduce the phenotype of M2-type macrophages, indicating that the anti-tumor activity of CII-3 could be related to the inhibitory effect on M2 polarization, and pericanaside was one of the active components.
Collapse
|
30
|
Intratumoral injection of schwannoma with attenuated Salmonella typhimurium induces antitumor immunity and controls tumor growth. Proc Natl Acad Sci U S A 2022; 119:e2202719119. [PMID: 35675425 PMCID: PMC9214496 DOI: 10.1073/pnas.2202719119] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Schwannomas are slow-growing benign neoplasms that develop throughout the body causing pain, sensory/motor dysfunction, and death. Because bacterial immunotherapy has been used in the treatment of some malignant neoplasms, we evaluated attenuated Salmonella typhimurium strains as immunotherapies for benign murine schwannomas. Several bacterial strains were tested, including VNP20009, a highly attenuated strain that was previously shown to be safe in human subjects with advanced malignant neoplasms, and a VNP20009 mutant that was altered in motility and other properties that included adherence and invasion of cultured mammalian cells. VNP20009 controlled tumor growth in two murine schwannoma models and induced changes in cytokine and immune effector cell profiles that were consistent with induction of enhanced innate and adaptive host immune responses compared with controls. Intratumoral (i.t.) injection of S. typhimurium led to tumor cell apoptosis, decreased tumor angiogenesis, and lower growth of the injected schwannoma tumors. Invasive VNP20009 was significantly more efficacious than was a noninvasive derivative in controlling the growth of injected tumors. Bacterial treatment apparently induced systemic antitumor immunity in that the growth of rechallenge schwannomas implanted following primary bacterial treatment was also reduced. Checkpoint programmed death-1 (PD-1) blockade induced by systemic administration of anti-PD-1 antibodies controlled tumor growth to the same degree as i.t. injection of S. typhimurium, and together, these two therapies had an additive effect on suppressing schwannoma growth. These experiments represent validation of a bacterial therapy for a benign neoplasm and support development of S. typhimurium VNP20009, potentially in combination with PD-1 inhibition, as a schwannoma immunotherapy.
Collapse
|
31
|
Yang D, Yang L, Cai J, Li H, Xing Z, Hou Y. Phosphoinositide 3-kinase/Akt and its related signaling pathways in the regulation of tumor-associated macrophages polarization. Mol Cell Biochem 2022; 477:2469-2480. [PMID: 35590082 DOI: 10.1007/s11010-022-04461-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 04/28/2022] [Indexed: 12/24/2022]
Abstract
Tumor-associated macrophages (TAMs) are a type of functionally plastic immune cell population in tumor microenvironment (TME) and mainly polarized into two phenotypes: M2 and M1-like TAMs. The M2-like TAMs could stimulate tumor growth and metastasis, tissue remodeling and immune-suppression, whereas M1-like TAMs could initiate immune response to dampen tumor progression. TAMs with different polarization phenotypes can produce various kinds of cytokines, chemokines and growth factors to regulate immunity and inflammatory responses. It is an effective method to treat cancer through ameliorating TME and modulating TAMs by converting M2 into M1-like phenotype. However, intracellular signaling mechanisms underlying TAMs polarization are largely undefined. Phosphoinositide 3-kinase (PI3K)/Akt is an important signaling pathway participating in M2-like TAMs polarization, survival, growth, proliferation, differentiation, apoptosis and cytoskeleton rearrangement. In the present review, we analyzed the mechanism of TAMs polarization focusing on PI3K/Akt and its downstream mitogen‑activated protein kinase (MAPK) as well as nuclear factor kappa B (NF-κB) signaling pathways, thus provides the first evidence of intracellular targets for cancer immunotherapy.
Collapse
Affiliation(s)
- Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Jialing Cai
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Huaxin Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, 150001, Heilongjiang, China
| | - Zheng Xing
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beijing Advanced Innovation Centre for Biomedical Engineering, Beihang University, Beijing, 100191, China
| | - Ying Hou
- Institute of Basic and Translational Medicine, Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Shaanxi Key Laboratory of Brain Disorders, Xi'an Medical University, Xi'an, 710021, Shaanxi, China.
| |
Collapse
|
32
|
Abstract
Mitogen-activated protein kinase (MAPK)-activated protein kinases (MAPKAPKs) are defined by their exclusive activation by MAPKs. They can be activated by classical and atypical MAPKs that have been stimulated by mitogens and various stresses. Genetic deletions of MAPKAPKs and availability of highly specific small-molecule inhibitors have continuously increased our functional understanding of these kinases. MAPKAPKs cooperate in the regulation of gene expression at the level of transcription; RNA processing, export, and stability; and protein synthesis. The diversity of stimuli for MAPK activation, the cross talk between the different MAPKs and MAPKAPKs, and the specific substrate pattern of MAPKAPKs orchestrate immediate-early and inflammatory responses in space and time and ensure proper control of cell growth, differentiation, and cell behavior. Hence, MAPKAPKs are promising targets for cancer therapy and treatments for conditions of acute and chronic inflammation, such as cytokine storms and rheumatoid arthritis. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Natalia Ronkina
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany;
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany;
| |
Collapse
|
33
|
Inman KS, Liu Y, Scotti Buzhardt ML, Leitges M, Krishna M, Crawford HC, Fields AP, Murray NR. Prkci Regulates Autophagy and Pancreatic Tumorigenesis in Mice. Cancers (Basel) 2022; 14:796. [PMID: 35159064 PMCID: PMC8834021 DOI: 10.3390/cancers14030796] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
Protein kinase C iota (PKCι) functions as a bonafide human oncogene in lung and ovarian cancer and is required for KrasG12D-mediated lung cancer initiation and progression. PKCι expression is required for pancreatic cancer cell growth and maintenance of the transformed phenotype; however, nothing is known about the role of PKCι in pancreas development or pancreatic tumorigenesis. In this study, we investigated the effect of pancreas-specific ablation of PKCι expression on pancreatic cellular homeostasis, susceptibility to pancreatitis, and KrasG12D-mediated pancreatic cancer development. Knockout of pancreatic Prkci significantly increased pancreatic immune cell infiltration, acinar cell DNA damage, and apoptosis, but reduced sensitivity to caerulein-induced pancreatitis. Prkci-ablated pancreatic acinar cells exhibited P62 aggregation and a loss of autophagic vesicles. Loss of pancreatic Prkci promoted KrasG12D-mediated pancreatic intraepithelial neoplasia formation but blocked progression to adenocarcinoma, consistent with disruption of autophagy. Our results reveal a novel promotive role for PKCι in pancreatic epithelial cell autophagy and pancreatic cancer progression.
Collapse
Affiliation(s)
- Kristin S. Inman
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
- Environmental Health Perspectives/National Institute of Environmental Health Sciences, Durham, NC 27709, USA
| | - Yi Liu
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
| | - Michele L. Scotti Buzhardt
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
- Neogenomics Laboratories, Clinical Division, Charlotte, NC 28104, USA
| | - Michael Leitges
- Department of BioMedical Sciences, Faculty of Medicine, Memorial University, St. John’s, NL A1M 2V7, Canada;
| | - Murli Krishna
- Department of Pathology/Lab Medicine, Mayo Clinic, Jacksonville, FL 32224, USA;
| | - Howard C. Crawford
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Detroit, MI 48202, USA
| | - Alan P. Fields
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
| | - Nicole R. Murray
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA; (K.S.I.); (Y.L.); (M.L.S.B.); (H.C.C.); (A.P.F.)
| |
Collapse
|
34
|
Mesencephalic astrocyte-derived neurotrophic factor reprograms macrophages to ameliorate acetaminophen-induced acute liver injury via p38 MAPK pathway. Cell Death Dis 2022; 13:100. [PMID: 35110525 PMCID: PMC8810950 DOI: 10.1038/s41419-022-04555-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 12/31/2022]
Abstract
Acetaminophen (APAP)-induced liver injury (AILI) is the most frequent cause of acute liver failure; but the underlying mechanisms still remain obscure. Macrophages and endoplasmic reticulum (ER) stress play an important role in the pathogenesis of AILI. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a newly identified 18-kDa soluble protein, whose expression and secretion are stimulated by ER stress. To investigate the role of myeloid cell MANF in the pathogenesis of AILI, we assayed serum and liver samples from AILI model mice and patients with drug-induced liver injury (DILI). We demonstrated that the levels of MANF were elevated in patients with DILI and in mice with AILI. Moreover, myeloid-specific MANF knockout mice were generated and used. It was observed that a delayed liver recovery from myeloid-specific MANF gene knockout mice following APAP overdose compared to that from wild-type mice. MANF deficiency in myeloid cells resulted in increased infiltrating monocyte-derived macrophages (MoMFs) but reduced restorative Ly6Clow macrophages after APAP treatment. MANF supplementation increased restorative Ly6Clow macrophages and subsequently alleviated liver injury. Moreover, MANF could enhance IL-10 expression and phagocytosis in macrophages via p38 MAPK pathway. Altogether, MANF seems to be a critical immune modulator in promoting liver repair via reducing and reprogramming MoMFs. MANF perhaps promoted the phenotype conversion of pro-inflammatory MoMFs to pro-restorative Ly6Clow MoMFs via p38 MAPK pathway, particularly through enhancing IL-10 and phagocytosis.
Collapse
|
35
|
Zeng Q, Ma X, Song Y, Chen Q, Jiao Q, Zhou L. Targeting regulated cell death in tumor nanomedicines. Am J Cancer Res 2022; 12:817-841. [PMID: 34976215 PMCID: PMC8692918 DOI: 10.7150/thno.67932] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/21/2021] [Indexed: 12/17/2022] Open
Abstract
Nanomedicines hold great potential in anticancer therapy by modulating the biodistribution of nanomaterials and initiating targeted oxidative stress damage, but they are also limited by the inherent self-protection mechanism and the evolutionary treatment resistance of cancer cells. New emerging explorations of regulated cell death (RCD), including processes related to autophagy, ferroptosis, pyroptosis, and necroptosis, substantially contribute to the augmented therapeutic efficiency of tumors by increasing the sensitivity of cancer cells to apoptosis. Herein, paradigmatic studies of RCD-mediated synergistic tumor nanotherapeutics are introduced, such as regulating autophagy-enhanced photodynamic therapy (PDT), targeting ferroptosis-sensitized sonodynamic therapy (SDT), inducing necroptosis-augmented photothermal therapy (PTT), and initiating pyroptosis-collaborative chemodynamic therapy (CDT), and the coordination mechanisms are discussed in detail. Multiangle analyses addressing the present challenges and upcoming prospects of RCD-based nanomedicines have also been highlighted and prospected for their further strengthening and the broadening of their application scope. It is believed that up-and-coming coadjutant therapeutic methodologies based on RCDs will considerably impact precision nanomedicine for cancer.
Collapse
|
36
|
Jia CL, Yang F, Li R. Prognostic Model Construction and Immune Microenvironment Analysis of Breast Cancer Based on Ferroptosis-Related lncRNAs. Int J Gen Med 2021; 14:9817-9831. [PMID: 34949938 PMCID: PMC8691199 DOI: 10.2147/ijgm.s342783] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose To construct a prognostic model of breast cancer using ferroptosis-related lncRNAs and explore novel therapeutic targets. Materials and Methods A prognostic characteristic model based on differential expression of ferroptosis-related lncRNAs in breast cancer was established based on TCGA data. Results Eleven ferroptosis-related lncRNAs associated with breast cancer prognosis were identified. Kaplan–Meier analysis suggested that high-risk lncRNA signatures correspond to a poor prognosis. The AUC of the signature lncRNAs was 0.682, demonstrating that it is accurate in predicting BC prognosis. GSEA showed that ferroptosis-related lncRNAs in high-risk individuals are mainly enriched in cell cycle, cell adhesion and tumor pathways. Immunity and gene expression analysis revealed that APC co-inhibition, check-point, HLA, inflammation-promoting and T cell co-stimulation among others were significantly different between the high-and low-risk group. Three immune checkpoints were highly expressed in the high-risk group. Conclusion Ferroptosis-related lncRNAs can be used as a prognostic feature to construct a prognostic model of breast cancer, based on which early detection markers, therapeutic targets and anti-tumor immune microenvironment can be studied, and clinical treatment can also be instructive.
Collapse
Affiliation(s)
- Cong Li Jia
- Institute of Plastic Surgery, Weifang Medical College, Weifang, Shandong, People's Republic of China
| | - Fu Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People's Republic of China
| | - Ruining Li
- Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu, People's Republic of China
| |
Collapse
|
37
|
Small SH, Tang EJ, Ragland RL, Ruzankina Y, Schoppy DW, Mandal RS, Glineburg MR, Ustelenca Z, Powell DJ, Simpkins F, Johnson FB, Brown EJ. Induction of IL19 expression through JNK and cGAS-STING modulates DNA damage-induced cytokine production. Sci Signal 2021; 14:eaba2611. [PMID: 34932373 PMCID: PMC9218922 DOI: 10.1126/scisignal.aba2611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2025]
Abstract
Cytokine production is a critical component of cell-extrinsic responses to DNA damage and cellular senescence. Here, we demonstrated that expression of the gene encoding interleukin-19 (IL-19) was enhanced by DNA damage through pathways mediated by c-Jun amino-terminal kinase (JNK) and cGAS-STING and that IL19 expression was required for the subsequent production of the cytokines IL-1, IL-6, and IL-8. IL19 expression was stimulated by diverse cellular stresses, including inhibition of the DNA replication checkpoint kinase ATR (ataxia telangiectasia and Rad3-related protein), oncogene expression, replicative exhaustion, oxidative stress, and DNA double-strand breaks. Unlike the production of IL-6 and IL-8, IL19 expression was not affected by abrogation of signaling by the IL-1 receptor (IL-1R) or the mitogen-activated protein kinase p38. Instead, the DNA damage–induced production of IL-1, IL-6, and IL-8 was substantially reduced by suppression of IL19 expression. The signaling pathways required to stimulate IL19 expression selectively depended on the type of DNA-damaging agent. Reactive oxygen species and the ASK1-JNK pathway were critical for responses to ionizing radiation (IR), whereas the cGAS-STING pathway stimulated IL19 expression in response to either IR or ATR inhibition. Whereas induction of IL1, IL6, and IL8 by IR depended on IL19 expression, the cGAS-STING–dependent induction of the immune checkpoint gene PDL1 after IR and ATR inhibition was independent of IL19. Together, these results suggest that IL-19 production by diverse pathways forms a distinct cytokine regulatory arm of the response to DNA damage.
Collapse
Affiliation(s)
- Sara H. Small
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E. Jessica Tang
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ryan L. Ragland
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yaroslava Ruzankina
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David W. Schoppy
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rahul S. Mandal
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - M. Rebecca Glineburg
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zgjim Ustelenca
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J. Powell
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Ovarian Cancer Research Center, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fiona Simpkins
- Penn Ovarian Cancer Research Center, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - F. Bradley Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric J. Brown
- Department of Cancer Biology and the Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
38
|
Suarez-Lopez L, Shui B, Brubaker DK, Hill M, Bergendorf A, Changelian PS, Laguna A, Starchenko A, Lauffenburger DA, Haigis KM. Cross-species transcriptomic signatures predict response to MK2 inhibition in mouse models of chronic inflammation. iScience 2021; 24:103406. [PMID: 34849469 PMCID: PMC8609096 DOI: 10.1016/j.isci.2021.103406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/28/2021] [Accepted: 11/03/2021] [Indexed: 11/30/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are genetically complex and exhibit significant inter-patient heterogeneity in disease presentation and therapeutic response. Here, we show that mouse models of IBD exhibit variable responses to inhibition of MK2, a pro-inflammatory serine/threonine kinase, and that MK2 inhibition suppresses inflammation by targeting inflammatory monocytes and neutrophils in murine models. Using a computational approach (TransComp-R) that allows for cross-species comparison of transcriptomic features, we identified an IBD patient subgroup that is predicted to respond to MK2 inhibition, and an independent preclinical model of chronic intestinal inflammation predicted to be non-responsive, which we validated experimentally. Thus, cross-species mouse-human translation approaches can help to identify patient subpopulations in which to deploy new therapies. MK2 kinase inhibition shows variable efficacy in different IBD mouse models TCT and TNFΔARE mice express distinct inflammatory and MK2-responsive genes “Response to MK2i” signature is enriched in monocytes and neutrophils Cross-species modeling identifies patient groups potentially responsive to MK2i
Collapse
Affiliation(s)
- Lucia Suarez-Lopez
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Bing Shui
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Douglas K. Brubaker
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47906, USA
- Regenstrief Center for Healthcare Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Marza Hill
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Alexander Bergendorf
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47906, USA
| | - Paul S. Changelian
- Aclaris Therapeutics, Inc., 4320 Forest Park Avenue, St. Louis, MO 63108, USA
| | - Aisha Laguna
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Alina Starchenko
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Douglas A. Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Kevin M. Haigis
- Department of Cancer Biology, Dana Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Brigham & Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
- Corresponding author
| |
Collapse
|
39
|
The Colorectal Cancer Tumor Microenvironment and Its Impact on Liver and Lung Metastasis. Cancers (Basel) 2021; 13:cancers13246206. [PMID: 34944826 PMCID: PMC8699466 DOI: 10.3390/cancers13246206] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/02/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Colorectal cancer (CRC) is the third most common cancer worldwide. Metastasis to secondary organs, such as the liver and lungs, is a key driver of CRC-related mortality. The tumor microenvironment, which consists of the primary cancer cells, as well as associated support and immune cells, significantly affects the behavior of CRC cells at the primary tumor site, as well as in metastatic lesions. In this paper, we review the role of the individual components of the tumor microenvironment on tumor progression, immune evasion, and metastasis, and we discuss the implications of these components on antitumor therapies. Abstract Colorectal cancer (CRC) is the third most common malignancy and the second most common cause of cancer-related mortality worldwide. A total of 20% of CRC patients present with distant metastases, most frequently to the liver and lung. In the primary tumor, as well as at each metastatic site, the cellular components of the tumor microenvironment (TME) contribute to tumor engraftment and metastasis. These include immune cells (macrophages, neutrophils, T lymphocytes, and dendritic cells) and stromal cells (cancer-associated fibroblasts and endothelial cells). In this review, we highlight how the TME influences tumor progression and invasion at the primary site and its function in fostering metastatic niches in the liver and lungs. We also discuss emerging clinical strategies to target the CRC TME.
Collapse
|
40
|
Chen IT, Chen HC, Lo YH, Lai PY, Hsieh FY, Wu YH, Shih HM, Lai MZ. Promyelocytic leukemia protein targets MK2 to promote cytotoxicity. EMBO Rep 2021; 22:e52254. [PMID: 34633746 PMCID: PMC8647022 DOI: 10.15252/embr.202052254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 09/19/2021] [Accepted: 09/27/2021] [Indexed: 11/09/2022] Open
Abstract
Promyelocytic leukemia protein (PML) is a tumor suppressor possessing multiple modes of action, including induction of apoptosis. We unexpectedly find that PML promotes necroptosis in addition to apoptosis, with Pml-/- macrophages being more resistant to TNF-mediated necroptosis than wild-type counterparts and PML-deficient mice displaying resistance to TNF-induced systemic inflammatory response syndrome. Reduced necroptosis in PML-deficient cells is associated with attenuated receptor-interacting protein kinase 1 (RIPK1) activation, as revealed by reduced RIPK1[S166] phosphorylation, and attenuated RIPK1-RIPK3-MLKL necrosome complex formation. We show that PML deficiency leads to enhanced TNF-induced MAPK-activated kinase 2 (MK2) activation and elevated RIPK1[S321] phosphorylation, which suppresses necrosome formation. MK2 inhibitor treatment or MK2 knockout abrogates resistance to cell death induction in PML-null cells and mice. PML binds MK2 and p38 MAPK, thereby inhibiting p38-MK2 interaction and MK2 activation. Moreover, PML participates in autocrine production of TNF induced by cellular inhibitors of apoptosis 1 (cIAP1)/cIAP2 degradation, since PML-knockout attenuates autocrine TNF. Thus, by targeting MK2 activation and autocrine TNF, PML promotes necroptosis and apoptosis, representing a novel tumor-suppressive activity for PML.
Collapse
Affiliation(s)
- I-Ting Chen
- Institute of Molecular Biology, Taipei, Taiwan
| | | | - Yu-Hsun Lo
- Institute of Molecular Biology, Taipei, Taiwan
| | | | - Fu-Yi Hsieh
- Institute of Molecular Biology, Taipei, Taiwan
| | | | - Hsiu-Ming Shih
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
41
|
Shafiq M, Lone ZR, Bharati P, Singh H, Jagavelu K, Verma NK, Ghosh JK, Gaestel M, Hanif K. Involvement of mitogen-activated protein kinase (MAPK)-activated protein kinase 2 (MK2) in endothelial dysfunction associated with pulmonary hypertension. Life Sci 2021; 286:120075. [PMID: 34678260 DOI: 10.1016/j.lfs.2021.120075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/07/2021] [Accepted: 10/18/2021] [Indexed: 01/08/2023]
Abstract
AIMS Increased proliferation, inflammation, and endothelial microparticle (EMP) generation in the pulmonary vasculature lead to endothelial dysfunction in pulmonary hypertension (PH). Interestingly, MK2, a downstream of p38MAPK, is a central regulator of inflammation, proliferation, and EMP generation in cardiovascular diseases. However, the role of MK2 in pulmonary endothelial dysfunction remains unexplored. MAIN METHODS The Human Pulmonary Artery Endothelial cells (HPAECs) were exposed to hypoxia (1% O2) for 72 h, and MK2 inhibition was achieved by siRNA treatment. Western blotting, qualitative RT-PCR, immunocytochemistry, flow cytometry and enzyme-linked immunoassays were conducted to study pathological alterations and molecular mechanisms. Neoangiogenesis was studied using cell migration and tubule formation assays. For in vivo study, Male Sprague Dawley rats and MK2 knock-out mice with littermate control were treated with monocrotaline (MCT) 60 mg/kg and 600 mg/kg, respectively (s.c. once in rat and weekly in mice) to induce PH. MMI-0100 (40 μg/kg, i.p. daily for 35 days), was administered in rats to inhibit MK2. KEY FINDINGS MK2 inhibition significantly decreased inflammation, cell proliferation, apoptosis resistance, and improved mitochondrial functions in hypoxic HPAECs. Hypoxia promoted cell migration, VEGF expression, and angiogenesis in HPAECs, which were also reversed by MK2 siRNA. MK2 inhibition decreased EMP generation and increased the expression of p-eNOS in hypoxic HPAECs, a marker of endothelial function. Furthermore, MK2 deficiency and inhibition both reduced the EMP generation in mice and rats, respectively. SIGNIFICANCE These findings proved that MK2 is involved in endothelial dysfunction, and its inhibition may be beneficial for endothelial function in PH.
Collapse
Affiliation(s)
- Mohammad Shafiq
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Zahid Rasool Lone
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pragya Bharati
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Himalaya Singh
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Kumaravelu Jagavelu
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India
| | - Neeraj Kumar Verma
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jimut Kanti Ghosh
- Division of Molecular and Structural Biology, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Matthias Gaestel
- Institute for Zellbiochemie, Medizinische Hochschule Hannover (MHH), OE 4310, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh, India.
| |
Collapse
|
42
|
Sriram G, Milling LE, Chen JK, Kong YW, Joughin BA, Abraham W, Swartwout S, Handly ED, Irvine DJ, Yaffe MB. The injury response to DNA damage in live tumor cells promotes antitumor immunity. Sci Signal 2021; 14:eabc4764. [PMID: 34665642 PMCID: PMC8791539 DOI: 10.1126/scisignal.abc4764] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although immune checkpoint blockade (ICB) has strong clinical benefit for treating some tumor types, it fails in others, indicating a need for additional modalities to enhance the ICB effect. Here, we identified one such modality by using DNA damage to create a live, injured tumor cell adjuvant. Using an optimized ex vivo coculture system, we found that treating tumor cells with specific concentrations of etoposide, mitoxantrone, or doxorubicin markedly enhanced dendritic cell–mediated T cell activation. These immune-enhancing effects of DNA damage did not correlate with immunogenic cell death markers or with the extent of apoptosis or necroptosis; instead, these effects were mediated by live injured cells with activation of the DNA-PK, ATR, NF-κB, p38 MAPK, and RIPK1 signaling pathways. In mice, intratumoral injection of ex vivo etoposide–treated tumor cells in combination with systemic ICB (by anti-PD-1 and anti-CTLA4 antibodies) increased the number of intratumoral CD103+ dendritic cells and circulating tumor-antigen–specific CD8+ T cells, decreased tumor growth, and improved survival. These effects were absent in Batf3−/− mice and in mice in which the DNA-damaging drug was injected directly into the tumor, due to DNA damage in the immune cells. The combination treatment induced complete tumor regression in a subset of mice that were then able to reject tumor rechallenge, indicating that the injured cell adjuvant treatment induced durable antitumor immunological memory. These results provide a strategy for enhancing the efficacy of immune checkpoint inhibition in tumor types that do not respond to this treatment modality by itself.
Collapse
Affiliation(s)
- Ganapathy Sriram
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Lauren E. Milling
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Jung-Kuei Chen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Yi Wen Kong
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Brian A. Joughin
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Wuhbet Abraham
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Susanne Swartwout
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Erika D. Handly
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Darrell J. Irvine
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Michael B. Yaffe
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142
- Center for Precision Cancer Medicine, Massachusetts Institute of Technology, Cambridge, MA 02139
- David. H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
- Divisions of Acute Care Surgery, Trauma, and Surgical Critical Care and Surgical Oncology, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
43
|
Kubota K, Shimizu A, Notake T, Masuo H, Hosoda K, Yasukawa K, Hayashi H, Umemura K, Kamachi A, Goto T, Tomida H, Yamazaki S, Soejima Y. Preoperative Peripheral Blood Lymphocyte-to-Monocyte Ratio Predicts Long-Term Outcome for Patients with Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol 2021; 29:1437-1448. [PMID: 34664139 DOI: 10.1245/s10434-021-10848-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/10/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Although various biomarkers are useful in predicting cancer prognosis, the most effective preoperative systemic biomarkers for pancreatic ductal adenocarcinoma (PDAC) have not been established. This study aimed to evaluate whether the lymphocyte-to-monocyte ratio (LMR) can predict the long-term outcomes for patients who were to undergo surgical resection of PDAC. METHODS The study involved 170 patients with PDAC who underwent resection. Multivariate analysis was performed to identify the independent prognostic factors for overall survival (OS) and disease-free survival (DFS) among clinicopathologic, surgical, and seven systemic biomarker-related factors including LMR. Subgroup analysis of PDAC located in the body and tail of the pancreas (B/T PDAC) was performed (n = 60) to eliminate the influence of preoperative cholangitis and surgical procedure. Furthermore, OS according to the postoperative course of the LMR value group was investigated. RESULTS A low LMR (<3.3) was the only independent predictive factor for OS (hazard ratio [HR], 3.52; p < 0.001) and DFS (HR, 3.31; p < 0.001) among the systemic biomarkers. Subgroup analysis of the B/T PDAC also showed that low the LMR was the independent predictive factor for OS (HR, 3.24; p = 0.002) and DFS (HR, 4.42; p = 0.003). The PDAC that maintained a high LMR from before surgery to 1 year after surgery showed good long-term outcomes (median OS, 8.5 years; 5-year survival rate, 61.8 %). CONCLUSIONS Preoperative LMR was an independent predictor of OS and DFS after surgery for PDAC. Maintaining a high LMR through the pre- and postoperative courses might improve the prognosis for patients with PDAC.
Collapse
Affiliation(s)
- Koji Kubota
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Akira Shimizu
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan.
| | - Tsuyoshi Notake
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hitoshi Masuo
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Kiyotaka Hosoda
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Koya Yasukawa
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hikaru Hayashi
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Kentaro Umemura
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Atsushi Kamachi
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Takamune Goto
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Hidenori Tomida
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Shiori Yamazaki
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| | - Yuji Soejima
- Division of Gastroenterological, Hepato-Billiary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto, Nagano, Japan
| |
Collapse
|
44
|
Olmsted-Davis E, Mejia J, Salisbury E, Gugala Z, Davis AR. A Population of M2 Macrophages Associated With Bone Formation. Front Immunol 2021; 12:686769. [PMID: 34712222 PMCID: PMC8547272 DOI: 10.3389/fimmu.2021.686769] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
We previously identified transient brown adipocyte-like cells associated with heterotopic ossification (HO). These ancillary cells support new vessel synthesis essential to bone formation. Recent studies have shown that the M2 macrophage contributes to tissue regeneration in a similar way. To further define the phenotype of these brown adipocyte-like cells they were isolated and characterized by single-cell RNAseq (scRNAseq). Analysis of the transcriptome and the presence of surface markers specific for macrophages suggest that these cells are M2 macrophages. To validate these findings, clodronate liposomes were delivered to the tissues during HO, and the results showed both a significant reduction in these macrophages as well as bone formation. These cells were isolated and shown in culture to polarize towards either M1 or M2 similar to other macrophages. To confirm that these are M2 macrophages, mice received lipopolysacheride (LPS), which induces proinflammation and M1 macrophages. The results showed a significant decrease in this specific population and bone formation, suggesting an essential role for M2 macrophages in the production of bone. To determine if these macrophages are specific to HO, we isolated these cells using fluorescence-activated cell sorting (FACS) from a bone defect model and subjected them to scRNAseq. Surprisingly, the macrophage populations overlapped between the two groups (HO-derived versus callus) suggesting that they may be essential ancillary cells for bone formation in general and not selective to HO. Of further note, their unique metabolism and lipogenic properties suggest the potential for unique cross talk between these cells and the newly forming bone.
Collapse
Affiliation(s)
- Elizabeth Olmsted-Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States,Department of Pediatrics – Section Hematology/Oncology, Baylor College of Medicine, Houston, TX, United States,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Julio Mejia
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States
| | - Elizabeth Salisbury
- Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, United States
| | - Zbigniew Gugala
- Department of Orthopedic Surgery and Rehabilitation, University of Texas Medical Branch, Galveston, TX, United States
| | - Alan R. Davis
- Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children’s Hospital and Houston Methodist Hospital, Houston, TX, United States,Department of Pediatrics – Section Hematology/Oncology, Baylor College of Medicine, Houston, TX, United States,Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States,*Correspondence: Alan R. Davis,
| |
Collapse
|
45
|
Wang W, Liu Y, Liu Z, Li S, Deng C, Yang X, Deng Q, Sun Y, Zhang Y, Ma Z, Li W, Liu Y, Zhou X, Li T, Zhu J, Wang J, Dai K. Evaluation of Interleukin-4-Loaded Sodium Alginate-Chitosan Microspheres for Their Support of Microvascularization in Engineered Tissues. ACS Biomater Sci Eng 2021; 7:4946-4958. [PMID: 34525809 DOI: 10.1021/acsbiomaterials.1c00882] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Defects in the formation of microvascular networks, which provide oxygen and nutrients to cells, are the main reason for the engraftment failure of clinically applicable engineered tissues. Inflammatory responses and immunomodulation can promote the vascularization of the engineered tissues. We developed a capillary construct composed of a gelatin methacrylate-based cell-laden hydrogel framework complexed with interleukin-4 (IL-4)-loaded alginate-chitosan (AC) microspheres and endothelial progenitor cells (EPCs) and RAW264.7 macrophages as model cells. The AC microspheres maintained and guided the EPCs through electrostatic adhesion, facilitating the formation of microvascular networks. The IL-4-loaded microspheres promoted the polarization of the macrophages into the M2 type, leading to a reduction in pro-inflammatory factors and enhancement of the vascularization. Hematoxylin and eosin staining and immunohistochemical analysis revealed that, without IL-4 or AC microspheres, the scaffold was less effective in angiogenesis. We provide an alternative and promising approach for constructing vascularized tissues.
Collapse
Affiliation(s)
- Wenhao Wang
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.,Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yuehua Liu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, 7 Kangfu Road, Zhengzhou 450052, People's Republic of China
| | - Zifan Liu
- School of Biological Science and Medical Engineering, BUAA, 37 Xueyuan Road, Haidian District, Beijing 100191, China
| | - Shuai Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Changxu Deng
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaoxiao Yang
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Qian Deng
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Yeye Sun
- Hunan Polytechnic of Environment and Biology, Hengyang 422000, China
| | - Yuxin Zhang
- Department of Rehabilitation Medicine, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Zhenjiang Ma
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wentao Li
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Yang Liu
- Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Xiaojun Zhou
- College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai 201620, China
| | - Tao Li
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Junfeng Zhu
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jinwu Wang
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.,Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Kerong Dai
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.,Shanghai Key Laboratory of Orthopaedic Implants, Department of Orthopaedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
46
|
Morgan D, Berggren KL, Spiess CD, Smith HM, Tejwani A, Weir SJ, Lominska CE, Thomas SM, Gan GN. Mitogen-activated protein kinase-activated protein kinase-2 (MK2) and its role in cell survival, inflammatory signaling, and migration in promoting cancer. Mol Carcinog 2021; 61:173-199. [PMID: 34559922 DOI: 10.1002/mc.23348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022]
Abstract
Cancer and the immune system share an intimate relationship. Chronic inflammation increases the risk of cancer occurrence and can also drive inflammatory mediators into the tumor microenvironment enhancing tumor growth and survival. The p38 MAPK pathway is activated both acutely and chronically by stress, inflammatory chemokines, chronic inflammatory conditions, and cancer. These properties have led to extensive efforts to find effective drugs targeting p38, which have been unsuccessful. The immediate downstream serine/threonine kinase and substrate of p38 MAPK, mitogen-activated-protein-kinase-activated-protein-kinase-2 (MK2) protects cells against stressors by regulating the DNA damage response, transcription, protein and messenger RNA stability, and motility. The phosphorylation of downstream substrates by MK2 increases inflammatory cytokine production, drives an immune response, and contributes to wound healing. By binding directly to p38 MAPK, MK2 is responsible for the export of p38 MAPK from the nucleus which gives MK2 properties that make it unique among the large number of p38 MAPK substrates. Many of the substrates of both p38 MAPK and MK2 are separated between the cytosol and nucleus and interfering with MK2 and altering this intracellular translocation has implications for the actions of both p38 MAPK and MK2. The inhibition of MK2 has shown promise in combination with both chemotherapy and radiotherapy as a method for controlling cancer growth and metastasis in a variety of cancers. Whereas the current data are encouraging the field requires the development of selective and well tolerated drugs to target MK2 and a better understanding of its effects for effective clinical use.
Collapse
Affiliation(s)
- Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kiersten L Berggren
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, UNM School of Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Colby D Spiess
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hannah M Smith
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ajay Tejwani
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Scott J Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Christopher E Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sufi M Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
47
|
Abstract
INTRODUCTION Chemokines and their cognate receptors play a major role in modulating inflammatory responses. Depending on their ligand binding, chemokine receptors can stimulate both immune activating and inhibitory signaling pathways. The CC chemokine receptor 5 (CCR5) promotes immune responses by recruiting immune cells to the sites of inflammation/tumor, and is involved in stimulating tumor cell proliferation, invasion and migration through various mechanisms. Moreover, CCR5 also contributes to an immune-suppressive tumor microenvironment by recruiting regulatory T cells and myeloid-derived suppressor cells facilitating tumor development and progression. In summary, cells expressing CCR5 modulate immune response and tumor progression. Expression of CCR5 is increased in various malignancies and associated with poor outcome. Experimental data show promising efficacy signals with CCR5 antagonists in preclinical tumor models. Therefore, CCR5 has been recognized as a potential therapeutic target for cancer. AREAS COVERED In this review, we focus on the role of CCR5 in cancer progression and discuss its impact and potential as a therapeutic target for cancer. EXPERT OPINION Beyond immune-checkpoint inhibitors, potentially synergistic immune-modulatory drugs such as CCR5 antagonists are a promising approach to enlarge our treatment armamentarium against cancer.
Collapse
Affiliation(s)
- Hossein Hemmatazad
- Department of Radiation Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
48
|
Xu SS, Li H, Li TJ, Li S, Xia HY, Long J, Wu CT, Wang WQ, Zhang WH, Gao HL, Han X, Ye LY, Lin X, Xu HX, Yu XJ, Liu L. Neutrophil Extracellular Traps and Macrophage Extracellular Traps Predict Postoperative Recurrence in Resectable Nonfunctional Pancreatic Neuroendocrine Tumors. Front Immunol 2021; 12:577517. [PMID: 34084158 PMCID: PMC8168461 DOI: 10.3389/fimmu.2021.577517] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
Background Extracellular traps (ETs) and tumor-infiltrating immune cells can contribute to disease progression. The clinical significance of tumor-infiltrating neutrophils and macrophages and related extracellular traps in pancreatic neuroendocrine tumors (pNETs) has not been fully elucidated. This study aimed to explore the prognostic value of tumor infiltration and ET formation by neutrophils and macrophages in pNETs. Methods A total of 135 patients with radical resection of nonfunctional pNETs were analyzed retrospectively. Immunohistochemistry and immunofluorescence were utilized to stain tumor tissue sections. The recurrence-free survival (RFS) of subgroups determined by Kaplan-Meier analysis was compared with the log-rank test. Univariate and multivariate Cox regression analyses were used to identify independent prognostic factors. A nomogram was established to predict 3-year RFS. Results Patients with high tumor-infiltrating neutrophils or macrophages or positive expression of neutrophils ETs or macrophage ETs displayed worse RFS (all p<0.05). Moreover, univariate and multivariate Cox regression analyses showed that neutrophil and macrophage infiltration and ETs were independent prognostic factors for RFS (all p<0.05). A combined parameter including WHO grade, TNM stage, tumor-infiltrating neutrophils and macrophages, and neutrophil and macrophage ETs had the highest C-index (0.866) and lowest Akaike information criteria (326.557). The calibration plot of nomogram composed of the combined parameter exhibited excellent prognostic values for 3-year RFS. Conclusions Infiltration and ETs by neutrophils and macrophages can be used as biological indicators of patient prognosis, suggesting the treatment potential for targeting those in nonfunctional pNETs.
Collapse
Affiliation(s)
- Shuai-Shuai Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Hao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Tian-Jiao Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shuo Li
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Huan-Yu Xia
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Long
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chun-Tao Wu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wen-Quan Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wu-Hu Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - He-Li Gao
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Han
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Long-Yun Ye
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xuan Lin
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Hua-Xiang Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xian-Jun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Shanghai Pancreatic Cancer Institute, Shanghai, China.,Pancreatic Cancer Institute, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Ozcan L, Kasikara C, Yurdagul A, Kuriakose G, Hubbard B, Serrano-Wu MH, Tabas I. Allosteric MAPKAPK2 inhibitors improve plaque stability in advanced atherosclerosis. PLoS One 2021; 16:e0246600. [PMID: 33983975 PMCID: PMC8118275 DOI: 10.1371/journal.pone.0246600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/20/2021] [Indexed: 11/19/2022] Open
Abstract
Atherosclerotic vascular disease resulting from unstable plaques is the leading cause of morbidity and mortality in subjects with type 2 diabetes (T2D), and thus a major therapeutic goal is to discover T2D drugs that can also promote atherosclerotic plaque stability. Genetic or pharmacologic inhibition of mitogen-activated protein kinase-activated protein kinase-2 (MAPKAPK2 or MK2) in obese mice improves glucose homeostasis and enhances insulin sensitivity. We developed two novel orally active small-molecule inhibitors of MK2, TBX-1 and TBX-2, and tested their effects on metabolism and atherosclerosis in high-fat Western diet (WD)-fed Ldlr-/- mice. Ldlr-/- mice were first fed the WD to allow atherosclerotic lesions to become established, and the mice were then treated with TBX-1 or TBX-2. Both compounds improved glucose metabolism and lowered plasma cholesterol and triglyceride, without an effect on body weight. Most importantly, the compounds decreased lesion area, lessened plaque necrosis, and increased fibrous cap thickness in the aortic root lesions of the mice. Thus, in a preclinical model of high-fat feeding and established atherosclerosis, MK2 inhibitors improved metabolism and also enhanced atherosclerotic plaque stability, suggesting potential for further clinical development to address the epidemic of T2D associated with atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Lale Ozcan
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Canan Kasikara
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Arif Yurdagul
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - George Kuriakose
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
| | - Brian Hubbard
- Tabomedex Biosciences, Boxford, Massachusetts, United States of America
| | | | - Ira Tabas
- Department of Medicine, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York, United States of America
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, United States of America
| |
Collapse
|
50
|
Zhao L, Shen J, Jia K, Shi F, Hao Q, Gao F. MicroRNA-24-3p Inhibits Microglia Inflammation by Regulating MK2 Following Spinal Cord Injury. Neurochem Res 2021; 46:843-852. [PMID: 33439430 DOI: 10.1007/s11064-020-03211-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 06/30/2020] [Accepted: 12/18/2020] [Indexed: 11/27/2022]
Abstract
Spinal cord injury (SCI) is a functional impairment of the spinal cord caused by external forces, accompanied by limb movement disorders and permanent paralysis, which seriously lowers the life quality of SCI patients. Secondary injury caused by inflammation attenuated the therapeutic effects of SCI. Therefore, the exploration of biomarkers associated with the inflammatory response following SCI might provide novel therapy strategy against SCI.SCI rat model was established as previously reported and evaluated by BBB score. The expression of microRNA-24-3p (miR-24-3p) and MAPK-activated protein kinase 2 (MK2) in spinal cord tissues of SCI rats and HAPI cells was analyzed by qRT-PCR. Protein expression of MK2, ionized calcium-binding adapter molecule-1 (Iba-1), tumor necrosis factor-alpha (TNF-α), and interleukin-1β (IL-1β) was assessed by western blot assay. The release of inflammatory cytokines TNF-α and IL-1β was measured by enzyme-linked immunosorbent assay (ELISA). The interaction between miR-24-3p and MK2 was examined by the luciferase reporter system. Basso-Beattie-Bresnahan (BBB) score dramatically reduced in rats following SCI compared with sham rats. Moreover, the expression of miR-24-3p was down-regulated, while MK2 was up-regulated in the spinal cord tissues of SCI rats and LPS-induced microglia cells compared with the corresponding control group. Luciferase reporter system confirmed the interaction between miR-24-3p and MK2. In addition, miR-24-3p upregulation or MK2 knockdown attenuated LPS induced activation of microglial cells and expression of inflammatory cytokine TNF-α and IL-1β. Besides, we discovered that miR-24-3p regulated inflammation of highly aggressively proliferating immortalized (HAPI) cells by targeting MK2.In our study, we clarified that miR-24-3p repressed inflammation of microglia cells following SCI by regulating MK2, thereby providing promising biomarkers for SCI therapy.
Collapse
Affiliation(s)
- Lin Zhao
- Department of Human Anatomy, Medical College of Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Juan Shen
- Department of Human Anatomy, Medical College of Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Kunpeng Jia
- Department of Pediatrics, Affiliated Hospital of Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Fangfang Shi
- Department of Human Anatomy, Medical College of Yan'an University, Yan'an, 716000, Shaanxi, China
| | - Qin Hao
- Department of Nursing, Medical College of Yan'an University, Guanghua Road, Baota District, Yan'an, 716000, Shaanxi, China.
| | - Feng Gao
- Department of Physiology, Medical College of Yan'an University, Guanghua Road, Baota District, Yan'an, 716000, Shaanxi, China.
| |
Collapse
|