1
|
Zou YT, Li JY, Chai JY, Hu YS, Zhang WJ, Zhang Q. The impact of the P2X7 receptor on the tumor immune microenvironment and its effects on tumor progression. Biochem Biophys Res Commun 2024; 707:149513. [PMID: 38508051 DOI: 10.1016/j.bbrc.2024.149513] [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: 12/04/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 03/22/2024]
Abstract
Cancer is a significant global health concern, and finding effective methods to treat it has been a focus of scientific research. It has been discovered that the growth, invasion, and metastasis of tumors are closely related to the environment in which they exist, known as the tumor microenvironment (TME). The immune response interacting with the tumor occurring within the TME constitutes the tumor immune microenvironment, and the immune response can lead to anti-tumor and pro-tumor outcomes and has shown tremendous potential in immunotherapy. A channel called the P2X7 receptor (P2X7R) has been identified within the TME. It is an ion channel present in various immune cells and tumor cells, and its activation can lead to inflammation, immune responses, angiogenesis, immunogenic cell death, and promotion of tumor development. This article provides an overview of the structure, function, and pharmacological characteristics of P2X7R. We described the concept and components of tumor immune microenvironment and the influence immune components has on tumors. We also outlined the impact of P2X7R regulation and how it affects the development of tumors and summarized the effects of drugs targeting P2X7R on tumor progression, both past and current, assisting researchers in treating tumors using P2X7R as a target.
Collapse
Affiliation(s)
- Yu-Ting Zou
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Jin-Yuan Li
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Jun-Yi Chai
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Yu-Shan Hu
- The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| | - Wen-Jun Zhang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China; The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China.
| | - Qiao Zhang
- Orthopedics Department, The Second Affiliated Hospital, Nanchang University, Nanchang City, Jiangxi province, 343000, China
| |
Collapse
|
2
|
Song J, Qin BF, Feng QY, Zhang JJ, Zhao GY, Luo Z, Sun HM. Albiflorin ameliorates thioacetamide-induced hepatic fibrosis: The involvement of NURR1-mediated inflammatory signaling cascades in hepatic stellate cells activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 276:116334. [PMID: 38626607 DOI: 10.1016/j.ecoenv.2024.116334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/11/2024] [Accepted: 04/12/2024] [Indexed: 04/18/2024]
Abstract
Thioacetamide (TAA) within the liver generates hepatotoxic metabolites that can be induce hepatic fibrosis, similar to the clinical pathological features of chronic human liver disease. The potential protective effect of Albiflorin (ALB), a monoterpenoid glycoside found in Paeonia lactiflora Pall, against hepatic fibrosis was investigated. The mouse hepatic fibrosis model was induced with an intraperitoneal injection of TAA. Hepatic stellate cells (HSCs) were subjected to treatment with transforming growth factor-beta (TGF-β), while lipopolysaccharide/adenosine triphosphate (LPS/ATP) was added to stimulate mouse peritoneal macrophages (MPMs), leading to the acquisition of conditioned medium. For TAA-treated mice, ALB reduced ALT, AST, HYP levels in serum or liver. The administration of ALB reduced histopathological abnormalities, and significantly regulated the expressions of nuclear receptor-related 1 protein (NURR1) and the P2X purinoceptor 7 receptor (P2×7r) in liver. ALB could suppress HSCs epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) deposition, and pro-inflammatory factor level. ALB also remarkably up-regulated NURR1, inhibited P2×7r signaling pathway, and worked as working as C-DIM12, a NURR1 agonist. Moreover, deficiency of NURR1 in activated HSCs and Kupffer cells weakened the regulatory effect of ALB on P2×7r inhibition. NURR1-mediated inhibition of inflammatory contributed to the regulation of ALB ameliorates TAA-induced hepatic fibrosis, especially based on involving in the crosstalk of HSCs-macrophage. Therefore, ALB plays a significant part in the mitigation of TAA-induced hepatotoxicity this highlights the potential of ALB as a protective intervention for hepatic fibrosis.
Collapse
Affiliation(s)
- Jian Song
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China
| | - Bo-Feng Qin
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China
| | - Qi-Yuan Feng
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China
| | - Jin-Jin Zhang
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China
| | - Gui-Yun Zhao
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China.
| | - Zheng Luo
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China.
| | - Hai-Ming Sun
- College of Pharmacy, Beihua University, Jilin, Jilin Province 132013, China.
| |
Collapse
|
3
|
Ohba Y, Adachi K, Furukawa T, Nishimaru T, Sakurai K, Masuo R, Inami T, Orita T, Akai S, Adachi T, Usui K, Hamada Y, Mori M, Kurimoto T, Wakashima T, Akiyama Y, Miyazaki S, Noji S. Discovery of Novel NLRP3 Inflammasome Inhibitors Composed of an Oxazole Scaffold Bearing an Acylsulfamide. ACS Med Chem Lett 2023; 14:1833-1838. [PMID: 38116417 PMCID: PMC10726461 DOI: 10.1021/acsmedchemlett.3c00433] [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: 09/25/2023] [Revised: 10/22/2023] [Accepted: 11/07/2023] [Indexed: 12/21/2023] Open
Abstract
The NLRP3 inflammasome plays an important role in the defense mechanism of the innate immune system and has recently attracted much attention as a drug target for various inflammatory disorders. Among the strategies for generating the novel chemotype in current drug discovery, scaffold hopping and bioisosteric replacement are known to be attractive approaches. As the results of our medicinal chemistry campaign, which involved exploration of core motifs using a ring closing approach, a five-membered oxazole-based scaffold was identified, and subsequent implementation of bioisosteric replacement led to discovery of a novel chemical class of NLRP3 inflammasome inhibitor bearing the acylsulfamide group. Further optimization of aniline and sulfamide moieties to improve potency in human whole blood assay led to the identification of the orally bioactive compound 32 in the LPS challenge model. Furthermore, compound 32 attenuated kidney injury in adriamycin-induced glomerulonephritis in mice. These investigations indicated that the NLRP3 inhibitor could be a potential therapeutic agent for glomerulonephritis.
Collapse
Affiliation(s)
- Yusuke Ohba
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Kaoru Adachi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Takayuki Furukawa
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Tatsuya Nishimaru
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Kentaro Sakurai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Ritsuki Masuo
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Tasuku Inami
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Takuya Orita
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Shota Akai
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Tsuyoshi Adachi
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Kenji Usui
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Yuji Hamada
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Mutsuki Mori
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Takafumi Kurimoto
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Takeshi Wakashima
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Yoshiyuki Akiyama
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Susumu Miyazaki
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| | - Satoru Noji
- Central Pharmaceutical Research Institute, Japan Tobacco Inc., 1-1, Murasaki-cho, Takatsuki Osaka 569-1125, Japan
| |
Collapse
|
4
|
Yufang W, Mingfang L, Nan H, Tingting W. Quercetin-targeted AKT1 regulates the Raf/MEK/ERK signaling pathway to protect against doxorubicin-induced nephropathy in mice. Tissue Cell 2023; 85:102229. [PMID: 37812949 DOI: 10.1016/j.tice.2023.102229] [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: 06/07/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Doxorubicin is an anthracycline antitumor agent commonly used in clinical practice, which has some nephrotoxicity and is often used to establish mouse models of kidney injury for basic medical research. This study will investigate the protective effect of quercetin on renal function in doxorubicin-induced nephropathy mice. METHODS C57BL/6 mice were divided into control, model, and quercetin low-, and high-dose groups. Serum and urine were collected to analyze markers of kidney function. H&E staining was used to detect pathological changes in renal tissues. Transmission electron microscopy was performed to observe the ultrastructural changes in renal tissues. Immunohistochemistry was performed to detect the changes of Ang II. RT-qPCR was performed to detect the changes of cytokines. ELISA was used to detect changes in serum inflammatory factors. Molecular docking was performed to verify the targeting relationship between quercetin and AKT1. Western blot was performed to detect Bax, Bcl-2, Cyt-c, AKT1, Raf, MEK, and ERK proteins. RESULTS Quercetin could induce the recovery of kidney function in kidney-injured mice; H&E results showed that kidney tissue damage and tissue fibrosis were reduced in kidney-injured mice under quercetin. The mitochondrial swollen structure was destroyed by doxorubicin, while the mitochondrial structure was restored under quercetin. The levels of abnormal apoptotic proteins Bax and Bcl-2 were regulated to normal by quercetin. The high expression of Ang II caused by doxorubicin was down-regulated by quercetin. Abnormal inflammatory factors caused by doxorubicin were reversed by quercetin. Western blot experiments showed that quercetin regulated the protein levels of AKT1 and Raf/MEK/ERK and inhibited the detrimental effects of doxorubicin. CONCLUSION Quercetin may mitigate doxorubicin-induced kidney injury in mice by regulating renal cell inflammatory factors and Raf/MEK/ERK signaling pathway through AKT1 to promote recovery of renal function.
Collapse
Affiliation(s)
- Wang Yufang
- Department of Medical Laboratory Diagnosis Lecturer, Quanzhou Medical College, Quanzhou, Fujian Province, China.
| | - Liu Mingfang
- The Second Attached Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Huang Nan
- Department of Medical Laboratory Diagnosis Lecturer, Quanzhou Medical College, Quanzhou, Fujian Province, China
| | - Wang Tingting
- Department of Medical Laboratory Diagnosis Lecturer, Quanzhou Medical College, Quanzhou, Fujian Province, China
| |
Collapse
|
5
|
Liu Y, Lei H, Zhang W, Xing Q, Liu R, Wu S, Liu Z, Yan Q, Li W, Liu X, Hu Y. Pyroptosis in renal inflammation and fibrosis: current knowledge and clinical significance. Cell Death Dis 2023; 14:472. [PMID: 37500614 PMCID: PMC10374588 DOI: 10.1038/s41419-023-06005-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/29/2023]
Abstract
Pyroptosis is a novel inflammatory form of regulated cell death (RCD), characterized by cell swelling, membrane rupture, and pro-inflammatory effects. It is recognized as a potent inflammatory response required for maintaining organismal homeostasis. However, excessive and persistent pyroptosis contributes to severe inflammatory responses and accelerates the progression of numerous inflammation-related disorders. In pyroptosis, activated inflammasomes cleave gasdermins (GSDMs) and generate membrane holes, releasing interleukin (IL)-1β/18, ultimately causing pyroptotic cell death. Mechanistically, pyroptosis is categorized into caspase-1-mediated classical pyroptotic pathway and caspase-4/5/11-mediated non-classical pyroptotic pathway. Renal fibrosis is a kidney disease characterized by the loss of structural and functional units, the proliferation of fibroblasts and myofibroblasts, and extracellular matrix (ECM) accumulation, which leads to interstitial fibrosis of the kidney tubules. Histologically, renal fibrosis is the terminal stage of chronic inflammatory kidney disease. Although there is a multitude of newly discovered information regarding pyroptosis, the regulatory roles of pyroptosis involved in renal fibrosis still need to be fully comprehended, and how to improve clinical outcomes remains obscure. Hence, this review systematically summarizes the novel findings regarding the role of pyroptosis in the pathogenesis of renal fibrosis and discusses potential biomarkers and drugs for anti-fibrotic therapeutic strategies.
Collapse
Affiliation(s)
- Ya Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Haibo Lei
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wenyou Zhang
- Department of Pharmacy, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qichang Xing
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Renzhu Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Shiwei Wu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Zheng Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Qingzi Yan
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Wencan Li
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China
| | - Xiang Liu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| | - Yixiang Hu
- Molecular Pharmacology Laboratory, Department of Clinical Pharmacy, Xiangtan Center Hospital, Xiangtan, 411100, China.
- Honghao Zhou Research Institute, Xiangtan Center Hospital, Xiangtan, 411100, China.
| |
Collapse
|