1
|
Zhu Q, Hao H, Gao Y, Li N, Liu Z, Shu L, Wang Q, Zhang L. Dapagliflozin ameliorates kidney injury following limb ischemia-reperfusion via the AMPK/SIRT1/NLRP3 pathway. Ren Fail 2025; 47:2495111. [PMID: 40264429 PMCID: PMC12020146 DOI: 10.1080/0886022x.2025.2495111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/20/2025] [Accepted: 04/08/2025] [Indexed: 04/24/2025] Open
Abstract
Limb ischemia-reperfusion (I/R) results in both localized tissue harm and injury to distant organs, particularly affecting the kidneys and leading to acute kidney injury. This study evaluates the renoprotective effect of dapagliflozin, a drug frequently prescribed for type 2 diabetes management, in relation to kidney injury caused by limb I/R. The extent of kidney injury was detected through serum marker testing in the rat model. Oxidative stress indicators and inflammatory factors were evaluated in rat and cellular models. Histological changes in the kidneys were examined using HE staining and electron microscopy. Cell pyroptosis was quantified using both TUNEL staining and flow cytometry. Cellular mitochondrial function was analyzed with JC-1 staining. AMPK/SIRT1/NLRP3 pathway-related proteins and their mRNAs were assessed via western blotting and RT-qPCR techniques. We showed that dapagliflozin reduced serum CRE, BUN, NGAL and KIM-1 levels and improved renal pathology in rat. Additionally, dapagliflozin significantly raised the concentrations of GSH-Px and SOD, concurrently reduced MDA and ROS levels in vivo and in vitro. It also lowered the levels of IL-6 and TNF-α and reduced cell pyroptosis. Furthermore, it was observed that dapagliflozin elevated AMPK and SIRT1 expressions, while decreasing NLRP3, ASC, GSDMD, IL-1β, and caspase-1 expressions. Notably, these effects of dapagliflozin were diminished in the presence of AMPK siRNA. Taken together, dapagliflozin exhibits a significant protective effect against kidney injury resulting from limb I/R. This protective effect operates through the inhibition of pyroptosis by activating the AMPK/SIRT1/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Qiuxiao Zhu
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Huiyao Hao
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ya Gao
- Department of Geriatrics, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Na Li
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zibo Liu
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Linyi Shu
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Qian Wang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lihui Zhang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
2
|
Mao Z, Chen P, Ji Q, Zhao X, Zhong K, Zeng X. Hydrogen sulfide and ferroptosis inhibition underlies the dietary restriction-induced protection against cyclophosphamide cystitis. Front Pharmacol 2025; 16:1562852. [PMID: 40432898 PMCID: PMC12106363 DOI: 10.3389/fphar.2025.1562852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Dietary restriction (DR) has emerged as a potential therapeutic intervention for various pathological conditions. This study investigated the effects of DR on cyclophosphamide-induced cystitis in mice. Animals were subjected to controlled food restriction for 1 week prior to cyclophosphamide administration. We evaluated changes in body weight, bladder pathology, redox status, and ferroptotic parameters. DR significantly attenuated cyclophosphamide-induced cystitis severity, as evidenced by reduced bladder weight, decreased lipid peroxidation, and diminished ferroptotic markers in bladder tissue. Mechanistic investigations revealed that DR upregulated hepatic hydrogen sulfide (H2S)-synthesizing enzymes and enhanced H2S production. Inhibition of H2S-synthesizing enzymes with DL-propargylglycine (PAG) and aminooxyacetic acid (AOAA) exacerbated cyclophosphamide-induced cystitis, whereas administration of diallyl trisulfide (DATS), an H2S donor, markedly ameliorated bladder pathology. In vitro studies demonstrated that H2S donors, NaHS and DATS, protected against cyclophosphamide metabolite acrolein (ACR)-induced urothelial cell death by suppressing oxidative stress, as indicated by reduced p38 MAPK activation and protein carbonylation. These findings suggest that DR confers protection against cyclophosphamide-induced cystitis through the induction of endogenous H2S production and inhibition of ferroptosis. Our study provides additional evidence supporting the health-promoting effects of DR as well as novel mechanistic insights into the beneficial effects of DR. Given H2S has anti-inflammatory and anti-oxidative properties and that oxidative stress and ferroptosis underlie various diseases, our finding could have broader implications.
Collapse
Affiliation(s)
- Zhimin Mao
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, Jiangsu, China
| | | | | | | | | | | |
Collapse
|
3
|
Zhu M, Fan X, Zhang N, Wang H, Ma J, Yin X, Cai J, Cong L, Chen R, Fan J, Kong X, Geng B, Gong Y, Du C. Endothelial endogenous CSE/H 2S inhibits endothelial pyroptosis by activating sirtuin1 to attenuate LPS-induced acute lung injury. FASEB J 2025; 39:e70420. [PMID: 40028711 DOI: 10.1096/fj.202402042r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 01/30/2025] [Accepted: 02/17/2025] [Indexed: 03/05/2025]
Abstract
Endothelial pyroptosis, a pro-inflammatory programmed cell death, promotes endothelial inflammation and is a pivotal process in the initial stage of acute lung injury (ALI). Hydrogen sulfide (H2S), a gasotransmitter primarily dependent on cystathionine γ-lyase (CSE) in the cardiovascular and respiratory systems, plays a protective role during ALI. Nonetheless, the modulatory role and precise molecular mechanism of endothelial endogenous CSE/H2S in the pathogenesis of ALI remain elusive. Herein, we prepared an ALI mouse model using intratracheal administration of LPS (5 mg/kg), and lung injury was assessed by evaluating pulmonary edema, inflammatory response, and endothelial pyroptosis. In this model, H2S production from pulmonary tissues declined in a time-dependent manner, accompanied by a compensatory elevation of CSE protein levels. Treatment with the H2S donor (NaHS) attenuated pulmonary edema, inflammatory cell infiltration, endothelial pyroptosis, and reduced serum levels of tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). Meanwhile, the inducible deletion of CSE in endothelial cells exacerbated these changes. The blocking effect of CSE/H2S on endothelial pyroptosis (evidenced by caspase-11 activation and GSDMD-NT formation) was also confirmed in cultured pulmonary microvascular endothelial cells (PMECs). Mechanistically, H2S-mediated regulation of sirtuin-1 (SIRT1) expression and activation (via sulfhydration) contributed to the modulatory process. Collectively, we uncovered that endothelial endogenous CSE/H2S alleviates endothelial pyroptosis by activating SIRT1, thereby preventing LPS-induced acute lung injury.
Collapse
Affiliation(s)
- Min Zhu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Anesthesiology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Nan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Hui Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianshe Ma
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xianghong Yin
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junyan Cai
- Department of Cardiology, Zhongda Hospital Affiliated to Southeast University, Nanjing, Jiangsu, P.R. China
| | - Linjing Cong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ran Chen
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiaoxia Kong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bin Geng
- Hypertension Center, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Congkuo Du
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Zhang K, Wu G, Chen Y, Hu Q, Li Y, Jiang X, Gu C, Zhang N, Zhao F. Hydrogen sulfide alleviates endothelial glycocalyx damage and promotes placental angiogenesis in rats exposed to cigarette smoke. Nitric Oxide 2025; 154:115-127. [PMID: 39645161 DOI: 10.1016/j.niox.2024.12.002] [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: 10/20/2024] [Revised: 11/15/2024] [Accepted: 12/05/2024] [Indexed: 12/09/2024]
Abstract
Our previous study has shown that hydrogen sulfide (H2S) can attenuate cigarette smoke exposure (CSE)-induced placental injury in rats. This study investigated whether H2S alleviates CSE-induced endothelial glycocalyx (eGC) impairment and promotes placental angiogenesis in rats. Twenty-four pregnant rats were randomly divided into four groups: control, NaHS (a donor of H2S), CSE, and CSE + NaHS. On gestational day 21, rat placentas were collected to detect H2S levels and protein expression of the H2S-synthesizing enzymes, cystathionine beta synthase (CBS), cystathionine gamma-lyase (CGL), and 3-mercaptopyruvate sulfurtransferase (3-MST), using a C-7Az fluorescent probe, H2S testing kit, and western blotting, respectively. Transmission electron microscopy and double immunofluorescence staining were performed to observe the placental eGC alterations. Placental angiogenesis, vascular endothelial proliferation and apoptosis, and protein expression levels of the PI3K/AKT/mTOR signaling pathway were assessed in rat placentas. The results showed that the administration of NaHS markedly attenuated the reduction in H2S levels and the decrease in CBS, CGL, and 3-MST expression caused by CSE in rat placentas. Notably, NaHS treatment distinctly alleviated eGC damage and facilitated placental angiogenesis in CSE-treated rats. NaHS administration effectively promoted placental vascular endothelial proliferation and suppressed endothelial apoptosis in CSE-treated rats. Furthermore, NaHS treatment markedly elevated the phosphorylation of PI3K, AKT, and mTOR in the placenta of CSE-treated rats. Taken together, these results indicate that exogenous administration of H2S can alleviate CSE-induced eGC damage and promote placental angiogenesis in CSE-treated rats, suggesting that H2S may be a novel therapeutic agent for the treatment of CSE-associated vascular disease.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Geng Wu
- Department of Biochemistry and Molecular Biology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yonglan Chen
- Department of Biochemistry and Molecular Biology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Qunying Hu
- Department of Biochemistry and Molecular Biology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yuanyuan Li
- Department of Biochemistry and Molecular Biology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xinyue Jiang
- Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Chunfu Gu
- Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Na Zhang
- Department of Biochemistry and Molecular Biology, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Fusheng Zhao
- Department of Histology and Embryology, Mudanjiang Medical University, Mudanjiang, 157011, China.
| |
Collapse
|
5
|
Yu Z, Xie S. Loureirin B improves H/R-induced hepatic ischemia-reperfusion injury by downregulating ALOX5 to regulate mitochondrial homeostasis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7561-7571. [PMID: 38662194 DOI: 10.1007/s00210-024-03079-7] [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: 03/07/2024] [Accepted: 04/01/2024] [Indexed: 04/26/2024]
Abstract
This study was conceived to explore the role and the mechanism of Loureirin B (LB) in hepatic IRI. The viability of LB-treated AML-12 cells was assessed using CCK-8 assay and inflammatory cytokines were detected using ELISA. The activities of ROS and oxidative stress markers MDA, SOD, and GSH-Px were detected using DCFH-DA and corresponding assay kits. The cell apoptosis and caspase3 activity were estimated with flow cytometry and caspase3 assay kits. The expressions of arachidonate 5-lipoxygenase (ALOX5) and apoptosis- and mitochondrial dynamics-related proteins were detected using western blot. The interaction between LB and ALOX5 was analyzed with molecular docking. The transfection efficacy of oe-ALOX5 was examined with RT-qPCR and western blot. Mitochondrial membrane potential was detected with JC-1 staining and immunofluorescence (IF) assay was employed to estimate mitochondrial fusion and fission. The present work found that LB revived the viability, inhibited inflammatory response, suppressed oxidative stress, repressed the apoptosis, and maintained mitochondrial homeostasis in H/R-induced AML-12 cells, which were all reversed by ALOX5 overexpression. Collectively, LB regulated mitochondrial homeostasis by downregulating ALOX5, thereby improving hepatic IRI.
Collapse
Affiliation(s)
- Zhaolong Yu
- Third Department of Internal Medicine, Yiwu Second People's Hospital, No. 1, Jiangbei Road, Fotang Town, Yiwu, 322000, Zhejiang, China.
| | - Shunying Xie
- Department of Emergency Medicine, Yiwu Second People's Hospital, Yiwu, Zhejiang, China
| |
Collapse
|
6
|
Liu Y, Pan R, Ouyang Y, Gu W, Xiao T, Yang H, Tang L, Wang H, Xiang B, Chen P. Pyroptosis in health and disease: mechanisms, regulation and clinical perspective. Signal Transduct Target Ther 2024; 9:245. [PMID: 39300122 DOI: 10.1038/s41392-024-01958-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024] Open
Abstract
Pyroptosis is a type of programmed cell death characterized by cell swelling and osmotic lysis, resulting in cytomembrane rupture and release of immunostimulatory components, which play a role in several pathological processes. Significant cellular responses to various stimuli involve the formation of inflammasomes, maturation of inflammatory caspases, and caspase-mediated cleavage of gasdermin. The function of pyroptosis in disease is complex but not a simple angelic or demonic role. While inflammatory diseases such as sepsis are associated with uncontrollable pyroptosis, the potent immune response induced by pyroptosis can be exploited as a therapeutic target for anti-tumor therapy. Thus, a comprehensive review of the role of pyroptosis in disease is crucial for further research and clinical translation from bench to bedside. In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development history, molecular mechanisms including canonical, non-canonical, caspase 3/8, and granzyme-mediated pathways, and its regulatory function in health and multiple diseases. Moreover, this review also provides updates on promising therapeutic strategies by applying novel small molecule inhibitors and traditional medicines to regulate pyroptosis. The present dilemmas and future directions in the landscape of pyroptosis are also discussed from a clinical perspective, providing clues for scientists to develop novel drugs targeting pyroptosis.
Collapse
Affiliation(s)
- Yifan Liu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
- Department of Oncology, Xiangya Hospital, Central South University, 87th Xiangya road, Changsha, 410008, Hunan province, China
| | - Renjie Pan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Yuzhen Ouyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
- Department of Neurology, Xiangya Hospital, Central South University, 87th Xiangya road, Changsha, 410008, Hunan province, China
| | - Wangning Gu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Tengfei Xiao
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Hongmin Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Ling Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China
| | - Hui Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| | - Bo Xiang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| | - Pan Chen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410013, China.
| |
Collapse
|
7
|
Wang Y, Zhang J, Yang Z, Li C, Zhang C, Sun S, Jiao Z, Che G, Gao H, Liu J, Li J. Ocotillol-Type Pseudoginsenoside-F11 Alleviates Lipopolysaccharide-Induced Acute Kidney Injury through Regulation of Macrophage Function by Suppressing the NF-κB/NLRP3/IL-1β Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20496-20512. [PMID: 39239930 DOI: 10.1021/acs.jafc.4c05185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Acute kidney injury (AKI) is characterized by a sudden decline in renal function. The inflammatory response is the fundamental pathologic alteration throughout AKI, regardless of the various causal factors. Macrophages are the main immune cells involved in the inflammatory microenvironment in AKI. Consequently, targeting macrophages might become a novel strategy for the treatment of AKI. In this study, we demonstrated that pseudoginsenoside-F11 (PF11), a distinctive component of Panax quinquefolius L., regulated macrophage function and protected renal tubular epithelial cells TCMK-1 from lipopolysaccharide (LPS) in vitro. PF11 also alleviated renal injuries in an LPS-induced AKI mouse model, decreased the levels of inflammatory cytokines, reduced macrophage inflammatory infiltration, and promoted the polarization of M1 macrophages to M2c macrophages with suppression of the nuclear factor-κB/NOD-like receptor thermal protein domain-associated protein 3/interleukin-1β (NF-κB/NLRP3/IL-1β) signaling pathway. To further investigate whether this nephroprotective effect of PF11 is mediated by macrophages, we performed macrophage depletion by injection of clodronate liposomes in mice. Macrophage depletion abolished PF11's ability to protect against LPS-induced kidney damage with downregulating the NF-κB/NLRP3/IL-1β signaling pathway. In summary, this is the first study providing data on the efficacy and mechanism of PF11 in the treatment of AKI by regulating macrophage function.
Collapse
Affiliation(s)
- Yaru Wang
- Innovation Center of New Drug Preclinical Pharmacology Evaluation of Jilin Province, Department of pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Jinyu Zhang
- Innovation Center of New Drug Preclinical Pharmacology Evaluation of Jilin Province, Department of pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Zhuo Yang
- The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130062, China
| | - Changcheng Li
- Innovation Center of New Drug Preclinical Pharmacology Evaluation of Jilin Province, Department of pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Chenming Zhang
- China-Japan Union Hospital of Jilin University, Changchun, Jilin 130022, China
| | - Shengkai Sun
- The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130062, China
| | - Ziyan Jiao
- The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130062, China
| | - Guanghua Che
- The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130062, China
| | - Hang Gao
- Innovation Center of New Drug Preclinical Pharmacology Evaluation of Jilin Province, Department of pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| | - Jinping Liu
- Research Center of Natural Drug, School of Pharmaceutical Sciences of Jilin University, Changchun, Jilin 130012, China
| | - Jing Li
- Innovation Center of New Drug Preclinical Pharmacology Evaluation of Jilin Province, Department of pharmacology, College of Basic Medicine of Jilin University, Changchun, Jilin 130012, China
| |
Collapse
|
8
|
Nelson P, Dugbartey GJ, McFarlane L, McLeod P, Major S, Jiang J, O'Neil C, Haig A, Sener A. Effect of Sodium Thiosulfate Pre-Treatment on Renal Ischemia-Reperfusion Injury in Kidney Transplantation. Int J Mol Sci 2024; 25:9529. [PMID: 39273476 PMCID: PMC11395123 DOI: 10.3390/ijms25179529] [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: 08/08/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/15/2024] Open
Abstract
We recently reported in a rat model of kidney transplantation that the addition of sodium thiosulfate (STS) to organ preservation solution improved renal graft quality and prolonged recipient survival. The present study investigates whether STS pre-treatment would produce a similar effect. In vitro, rat kidney epithelial cells were treated with 150 μM STS before and/or during exposure to hypoxia followed by reoxygenation. In vivo, donor rats were treated with PBS or 2.4 mg/kg STS 30 min before donor kidneys were procured and stored in UW or UW+150 μM STS solution at 4 °C for 24 h. Renal grafts were then transplanted into bilaterally nephrectomised recipient rats which were then sacrificed on post-operative day 3. STS pre-treatment significantly reduced cell death compared to untreated and other treated cells in vitro (p < 0.05), which corresponded with our in vivo result (p < 0.05). However, no significant differences were observed in other parameters of tissue injury. Our results suggest that STS pre-treatment may improve renal graft function after transplantation.
Collapse
Affiliation(s)
- Pierce Nelson
- Department of Microbiology & Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
| | - George J Dugbartey
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
- Multi-Organ Transplant Program, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
- London Health Sciences Center, Department of Surgery, Western University, London, ON N6A 5A5, Canada
- Department of Pharmacology & Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Accra P.O. Box LG43, Ghana
| | - Liam McFarlane
- Department of Microbiology & Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
| | - Patrick McLeod
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
| | - Sally Major
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
| | - Jifu Jiang
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
| | - Caroline O'Neil
- The Molecular Pathology Core, Robarts Research Institute, London, ON N6A 5A5, Canada
| | - Aaron Haig
- Department of Pathology and Laboratory Medicine, Western University, London, ON N6A 5A5, Canada
| | - Alp Sener
- Department of Microbiology & Immunology, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
- Matthew Mailing Center for Translational Transplant Studies, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
- Multi-Organ Transplant Program, London Health Sciences Center, Western University, London, ON N6A 5A5, Canada
- London Health Sciences Center, Department of Surgery, Western University, London, ON N6A 5A5, Canada
| |
Collapse
|
9
|
Lou S, Jiang ZL, Zhu YW, Zhang RY, Wang Y, Chu T, Liu YF, Zhang YX, Zhang CH, Su YK, Liu HX, Ji XY, Wu DD. Exploring the impact of hydrogen sulfide on hematologic malignancies: A review. Cell Signal 2024; 120:111236. [PMID: 38810860 DOI: 10.1016/j.cellsig.2024.111236] [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: 04/27/2024] [Revised: 05/22/2024] [Accepted: 05/24/2024] [Indexed: 05/31/2024]
Abstract
Hydrogen sulfide (H2S) is one of the three most crucial gaseous messengers in the body. The discovery of H2S donors, coupled with its endogenous synthesis capability, has sparked hope for the treatment of hematologic malignancies. In the last decade, the investigation into the impact of H2S has expanded, particularly within the fields of cardiovascular function, inflammation, infection, and neuromodulation. Hematologic malignancies refer to a diverse group of cancers originating from abnormal proliferation and differentiation of blood-forming cells, including leukemia, lymphoma, and myeloma. In this review, we delve deeply into the complex interrelation between H2S and hematologic malignancies. In addition, we comprehensively elucidate the intricate molecular mechanisms by which both H2S and its donors intricately modulate the progression of tumor growth. Furthermore, we systematically examine their impact on pivotal aspects, encompassing the proliferation, invasion, and migration capacities of hematologic malignancies. Therefore, this review may contribute novel insights to our understanding of the prospective therapeutic significance of H2S and its donors within the realm of hematologic malignancies.
Collapse
Affiliation(s)
- Shang Lou
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Zhi-Liang Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Rui-Yu Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Chuan-Hao Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Kun Su
- School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Hong-Xia Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan 450064, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan 475004, China; School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, China.
| |
Collapse
|
10
|
Kapisiz A, Kaya C, Eryilmaz S, Karabulut R, Turkyilmaz Z, Inan MA, Gulbahar O, Sonmez K. Protective effects of lupeol in rats with renal ischemia‑reperfusion injury. Exp Ther Med 2024; 28:313. [PMID: 38911048 PMCID: PMC11190881 DOI: 10.3892/etm.2024.12602] [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: 12/13/2023] [Accepted: 05/02/2024] [Indexed: 06/25/2024] Open
Abstract
Acute kidney injury (AKI) caused by ischemia and, exogenous or endogenous nephrotoxic agents poses a serious health issue. AKI is seen in 1% of all hospital admissions, 2-5% of hospitalizations and 67% of intensive care unit (ICU) patients. The in-hospital mortality rates for AKI is 40-50, and >50% for ICU patients. Ischemia-reperfusion (I/R) injury in the kidney can activate inflammatory responses and oxidative stress, resulting in AKI. The common endpoint in acute tubular necrosis is a cellular insult secondary to ischemia or direct toxins, which results in effacement of brush border, cell death and decreased function of tubular cells. The aim of the present study was to assess if the reported antioxidant and anti-inflammatory agent lupeol can exert any effects against renal I/R damage. In total, 24 Wistar Albino rats were randomly assigned into four groups of 6, namely Sham, lupeol, ischemia and therapy groups. In the lupeol group, intraperitoneal administration of 100 mg/kg lupeol was given 1 h before laparotomy, whilst only laparotomy was conducted in the sham group. The renal arteries of both kidneys were clamped for 45 min, 1 h after either intraperitoneal saline injection (in the ischemia group) or 100 mg/kg lupeol application (in the therapy group). The blood samples and renal tissues of all rats were collected after 24 h. In blood samples, blood urea nitrogen (BUN) was measured by the urease enzymatic method, and creatinine was measured by the kinetic Jaffe method. Using ELISA method, TNF-α and IL-6 levels were measured in the blood samples, whereas malondialdehyde (MDA), glutathione (GSH), caspase-3 levels were measured in kidney tissues. In addition, kidney histopathological analysis was performed by evaluating the degree of degeneration, tubular dilatation, interstitial lymphocyte infiltration, protein cylinders, necrosis and loss of brush borders. It was determined that renal damage occurred due to higher BUN, creatinine, MDA, TNF-α and caspase-3 values observed in the kidney tissues and blood samples of rats in ischemia group compared with the Sham group. Compared with those in the ischemia group, rats in the therapy group exhibited increased levels of GSH and reduced levels of BUN, TNF-α, MDA. Furthermore, the ischemia group also had reduced histopathological damage scores. Although differences in creatinine, IL-6 and caspase-3 levels were not statistically significant, they were markedly reduced in the treatment group. Taken together, these findings suggest that lupeol can prevent kidney damage as mainly evidenced by the reduced histopathological damage scores, decreased levels of oxidative stress and reduced levels of inflammatory markers. These properties may allow lupeol to be used in the treatment of AKI.
Collapse
Affiliation(s)
- Alparslan Kapisiz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Cem Kaya
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Sibel Eryilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Ramazan Karabulut
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Zafer Turkyilmaz
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Mehmet Arda Inan
- Department of Pathology, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Ozlem Gulbahar
- Department of Biochemistry, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| | - Kaan Sonmez
- Department of Pediatric Surgery, Gazi University Faculty of Medicine, Yenimahalle, 06500 Ankara, Turkey
| |
Collapse
|
11
|
Sun X, Wu S, Mao C, Qu Y, Xu Z, Xie Y, Jiang D, Song Y. Therapeutic Potential of Hydrogen Sulfide in Ischemia and Reperfusion Injury. Biomolecules 2024; 14:740. [PMID: 39062455 PMCID: PMC11274451 DOI: 10.3390/biom14070740] [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: 05/22/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury, a prevalent pathological condition in medical practice, presents significant treatment challenges. Hydrogen sulfide (H2S), acknowledged as the third gas signaling molecule, profoundly impacts various physiological and pathophysiological processes. Extensive research has demonstrated that H2S can mitigate I/R damage across multiple organs and tissues. This review investigates the protective effects of H2S in preventing I/R damage in the heart, brain, liver, kidney, intestines, lungs, stomach, spinal cord, testes, eyes, and other tissues. H2S provides protection against I/R damage by alleviating inflammation and endoplasmic reticulum stress; inhibiting apoptosis, oxidative stress, and mitochondrial autophagy and dysfunction; and regulating microRNAs. Significant advancements in understanding the mechanisms by which H2S reduces I/R damage have led to the development and synthesis of H2S-releasing agents such as diallyl trisulfide-loaded mesoporous silica nanoparticles (DATS-MSN), AP39, zofenopril, and ATB-344, offering a new therapeutic avenue for I/R injury.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Siyu Wu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Caiyun Mao
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Ying Qu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Zihang Xu
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| | - Ying Xie
- Department of Synopsis of the Golden Chamber, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Deyou Jiang
- Department of Synopsis of the Golden Chamber, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China;
| | - Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, China; (S.W.); (C.M.); (Y.Q.); (Z.X.)
| |
Collapse
|
12
|
Lv S, Zhao X, Ma C, Zhao D, Sun T, Fu W, Wei Y, Li W. Advancements in the study of acute lung injury resulting from intestinal ischemia/reperfusion. Front Med (Lausanne) 2024; 11:1399744. [PMID: 38933104 PMCID: PMC11199783 DOI: 10.3389/fmed.2024.1399744] [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: 03/12/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Intestinal ischemia/reperfusion is a prevalent pathological process that can result in intestinal dysfunction, bacterial translocation, energy metabolism disturbances, and subsequent harm to distal tissues and organs via the circulatory system. Acute lung injury frequently arises as a complication of intestinal ischemia/reperfusion, exhibiting early onset and a grim prognosis. Without appropriate preventative measures and efficacious interventions, this condition may progress to acute respiratory distress syndrome and elevate mortality rates. Nonetheless, the precise mechanisms and efficacious treatments remain elusive. This paper synthesizes recent research models and pertinent injury evaluation criteria within the realm of acute lung injury induced by intestinal ischemia/reperfusion. The objective is to investigate the roles of pathophysiological mechanisms like oxidative stress, inflammatory response, apoptosis, ferroptosis, and pyroptosis; and to assess the strengths and limitations of current therapeutic approaches for acute lung injury stemming from intestinal ischemia/reperfusion. The goal is to elucidate potential targets for enhancing recovery rates, identify suitable treatment modalities, and offer insights for translating fundamental research into clinical applications.
Collapse
Affiliation(s)
- Shihua Lv
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xudong Zhao
- Department of Hepatopancreatobiliary, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Can Ma
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Dengming Zhao
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tian Sun
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenchao Fu
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuting Wei
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Wenzhi Li
- Key Laboratory of Anesthesia and Intensive Care Research, Harbin, China
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
13
|
Li C, Yu Y, Zhu S, Hu Y, Ling X, Xu L, Zhang H, Guo K. The emerging role of regulated cell death in ischemia and reperfusion-induced acute kidney injury: current evidence and future perspectives. Cell Death Discov 2024; 10:216. [PMID: 38704372 PMCID: PMC11069531 DOI: 10.1038/s41420-024-01979-4] [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: 12/25/2023] [Revised: 04/14/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024] Open
Abstract
Renal ischemia‒reperfusion injury (IRI) is one of the main causes of acute kidney injury (AKI), which is a potentially life-threatening condition with a high mortality rate. IRI is a complex process involving multiple underlying mechanisms and pathways of cell injury and dysfunction. Additionally, various types of cell death have been linked to IRI, including necroptosis, apoptosis, pyroptosis, and ferroptosis. These processes operate differently and to varying degrees in different patients, but each plays a role in the various pathological conditions of AKI. Advances in understanding the underlying pathophysiology will lead to the development of new therapeutic approaches that hold promise for improving outcomes for patients with AKI. This review provides an overview of the recent research on the molecular mechanisms and pathways underlying IRI-AKI, with a focus on regulated cell death (RCD) forms such as necroptosis, pyroptosis, and ferroptosis. Overall, targeting RCD shows promise as a potential approach to treating IRI-AKI.
Collapse
Affiliation(s)
- Chenning Li
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Yan Hu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Xiaomin Ling
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Liying Xu
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
14
|
Yang K, Liu J, He T, Dong W. Caffeine and neonatal acute kidney injury. Pediatr Nephrol 2024; 39:1355-1367. [PMID: 37665410 DOI: 10.1007/s00467-023-06122-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 08/02/2023] [Accepted: 08/02/2023] [Indexed: 09/05/2023]
Abstract
Acute kidney injury is one of the most threatening diseases in neonates, with complex pathogenesis and limited treatment options. Caffeine is a commonly used central nervous system stimulant for treating apnea in preterm infants. There is compelling evidence that caffeine may have potential benefits for preventing neonatal acute kidney injury, but comprehensive reports are lacking in this area. Hence, this review aims to provide a summary of clinical data on the potential benefits of caffeine in improving neonatal acute kidney injury. Additionally, it delves into the molecular mechanisms underlying caffeine's effects on acute kidney injury, with a focus on various aspects such as oxidative stress, adenosine receptors, mitochondrial dysfunction, endoplasmic reticulum stress, inflammasome, autophagy, p53, and gut microbiota. The ultimate goal of this review is to provide information for healthcare professionals regarding the link between caffeine and neonatal acute kidney injury and to identify gaps in our current understanding.
Collapse
Affiliation(s)
- Kun Yang
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Jinjing Liu
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Ting He
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China
| | - Wenbin Dong
- Division of Neonatology, Department of Pediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Perinatology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Birth Defects, Luzhou, 646000, China.
| |
Collapse
|
15
|
Flori L, Benedetti G, Calderone V, Testai L. Hydrogen Sulfide and Irisin, Potential Allies in Ensuring Cardiovascular Health. Antioxidants (Basel) 2024; 13:543. [PMID: 38790648 PMCID: PMC11118251 DOI: 10.3390/antiox13050543] [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: 03/13/2024] [Revised: 04/19/2024] [Accepted: 04/27/2024] [Indexed: 05/26/2024] Open
Abstract
Irisin is a myokine secreted under the influence of physical activity and exposure to low temperatures and through different exogenous stimuli by the cleavage of its precursor, fibronectin type III domain-containing protein 5 (FNDC5). It is mainly known for maintaining of metabolic homeostasis, promoting the browning of white adipose tissue, the thermogenesis process, and glucose homeostasis. Growing experimental evidence suggests the possible central role of irisin in the regulation of cardiometabolic pathophysiological processes. On the other side, hydrogen sulfide (H2S) is well recognized as a pleiotropic gasotransmitter that regulates several homeostatic balances and physiological functions and takes part in the pathogenesis of cardiometabolic diseases. Through the S-persulfidation of cysteine protein residues, H2S is capable of interacting with crucial signaling pathways, exerting beneficial effects in regulating glucose and lipid homeostasis as well. H2S and irisin seem to be intertwined; indeed, recently, H2S was found to regulate irisin secretion by activating the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α)/FNDC5/irisin signaling pathway, and they share several mechanisms of action. Their involvement in metabolic diseases is confirmed by the detection of their lower circulating levels in obese and diabetic subjects. Along with the importance of metabolic disorders, these modulators exert favorable effects against cardiovascular diseases, preventing incidents of hypertension, atherosclerosis, heart failure, myocardial infarction, and ischemia-reperfusion injury. This review, for the first time, aims to explore the role of H2S and irisin and their possible crosstalk in cardiovascular diseases, pointing out the main effects exerted through the common molecular pathways involved.
Collapse
Affiliation(s)
- Lorenzo Flori
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (L.F.); (G.B.); (V.C.)
| | - Giada Benedetti
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (L.F.); (G.B.); (V.C.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (L.F.); (G.B.); (V.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56120 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56120 Pisa, Italy
| | - Lara Testai
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56120 Pisa, Italy; (L.F.); (G.B.); (V.C.)
- Interdepartmental Research Center Nutrafood “Nutraceuticals and Food for Health”, University of Pisa, 56120 Pisa, Italy
- Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, 56120 Pisa, Italy
| |
Collapse
|
16
|
Zhao J, Yang T, Yi J, Hu H, Lai Q, Nie L, Liu M, Chu C, Yang J. AP39 through AMPK-ULK1-FUNDC1 pathway regulates mitophagy, inhibits pyroptosis, and improves doxorubicin-induced myocardial fibrosis. iScience 2024; 27:109321. [PMID: 38558936 PMCID: PMC10981016 DOI: 10.1016/j.isci.2024.109321] [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: 08/08/2023] [Revised: 01/10/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Doxorubicin induces myocardial injury and fibrosis. Still, no effective interventions are available. AP39 is an H2S donor that explicitly targets mitochondria. This study investigated whether AP39 could improve doxorubicin-induced myocardial fibrosis. Doxorubicin induced significant myocardial fibrosis while suppressing mitophagy-related proteins and elevating pyroptosis-related proteins. Conversely, AP39 reverses these effects, enhancing mitophagy and inhibiting pyroptosis. In vitro experiments revealed that AP39 inhibited H9c2 cardiomyocyte pyroptosis, improved doxorubicin-induced impairment of mitophagy, reduced ROS levels, ameliorated the mitochondrial membrane potential, and upregulated AMPK-ULK1-FUNDC1 expression. In contrast, AMPK inhibitor (dorsomorphin) and ULK1 inhibitor (SBI-0206965) reversed AP39 antagonism of doxorubicin-induced FUNDC1-mediated impairment of mitophagy and secondary cardiomyocyte pyroptosis. These results suggest that mitochondria-targeted H2S can antagonize doxorubicin-induced pyroptosis and impaired mitophagy in cardiomyocytes via AMPK-ULK1-FUNDC1 and ameliorated myocardial fibrosis and remodeling.
Collapse
Affiliation(s)
- Junxiong Zhao
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Yang
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jiali Yi
- Department of Cardiology, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Hongmin Hu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Qi Lai
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| |
Collapse
|
17
|
Dilxat T, Shi Q, Chen X, Liu X. Garlic oil supplementation blocks inflammatory pyroptosis-related acute lung injury by suppressing the NF-κB/NLRP3 signaling pathway via H 2S generation. Aging (Albany NY) 2024; 16:6521-6536. [PMID: 38613798 PMCID: PMC11042940 DOI: 10.18632/aging.205721] [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: 10/19/2023] [Accepted: 03/09/2024] [Indexed: 04/15/2024]
Abstract
Acute lung injury (ALI) is a major cause of acute respiratory failure with a high morbidity and mortality rate, and effective therapeutic strategies for ALI remain limited. Inflammatory response is considered crucial for the pathogenesis of ALI. Garlic, a globally used cooking spice, reportedly exhibits excellent anti-inflammatory bioactivity. However, protective effects of garlic against ALI have never been reported. This study aimed to investigate the protective effects of garlic oil (GO) supplementation on lipopolysaccharide (LPS)-induced ALI models. Hematoxylin and eosin staining, pathology scores, lung myeloperoxidase (MPO) activity measurement, lung wet/dry (W/D) ratio detection, and bronchoalveolar lavage fluid (BALF) analysis were performed to investigate ALI histopathology. Real-time polymerase chain reaction, western blotting, and enzyme-linked immunosorbent assay were conducted to evaluate the expression levels of inflammatory factors, nuclear factor-κB (NF-κB), NLRP3, pyroptosis-related proteins, and H2S-producing enzymes. GO attenuated LPS-induced pulmonary pathological changes, lung W/D ratio, MPO activity, and inflammatory cytokines in the lungs and BALF. Additionally, GO suppressed LPS-induced NF-κB activation, NLRP3 inflammasome expression, and inflammatory-related pyroptosis. Mechanistically, GO promoted increased H2S production in lung tissues by enhancing the conversion of GO-rich polysulfide compounds or by increasing the expression of H2S-producing enzymes in vivo. Inhibition of endogenous or exogenous H2S production reversed the protective effects of GO on ALI and eliminated the inhibitory effects of GO on NF-κB, NLRP3, and pyroptotic signaling pathways. Overall, these findings indicate that GO has a critical anti-inflammatory effect and protects against LPS-induced ALI by suppressing the NF-κB/NLRP3 signaling pathway via H2S generation.
Collapse
Affiliation(s)
- Tursunay Dilxat
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| | - Qiang Shi
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| | - Xiaofan Chen
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| | - Xuxin Liu
- Xinjiang Agricultural Vocational Technological College, Changji 831100, Xinjiang, China
| |
Collapse
|
18
|
Xiong J, Zhao J. Pyroptosis: The Determinator of Cell Death and Fate in Acute Kidney Injury. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:118-131. [PMID: 38751798 PMCID: PMC11095617 DOI: 10.1159/000535894] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/15/2023] [Indexed: 05/18/2024]
Abstract
Background Acute kidney injury (AKI) is kidney damage that leads to a rapid decline in function. AKI primarily occurs when the tubular epithelium is damaged, causing swelling, loss of brush margin, and eventual apoptosis. Research has shown that tubular epithelial cell damage in AKI is linked to cell cycle arrest, autophagy, and regulation of cell death. Summary Pyroptosis, a type of programmed cell death triggered by inflammation, is believed to play a role in the pathophysiology of AKI. Cumulative evidence has shown that pyroptosis is the main cause of tubular cell death in AKI. Thus, targeted intervention of pyroptosis may be a promising therapeutic approach for AKI. This review delves deep into the cutting-edge research surrounding pyroptosis in the context of AKI, shedding light on its intricate mechanisms and potential implications for clinical practice. Additionally, we explore the exciting realm of potential preclinical treatment options for AKI, aiming to pave the way for future therapeutic advancements. Key Messages Pyroptosis, a highly regulated form of cell death, plays a crucial role in determining the fate of cells during the development of AKI. This intricate process involves the activation of inflammasomes, which are multi-protein complexes that initiate pyroptotic cell death. By understanding the mechanisms underlying pyroptosis, researchers aim to gain insights into the pathogenesis of AKI and potentially identify new therapeutic targets for this condition.
Collapse
Affiliation(s)
- Jiachuan Xiong
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| |
Collapse
|
19
|
Cao C, Yu P, Chu C, Wang Z, Xu W, Cheng F, Zhao H, Qiu Z. Magnesium hydride attenuates intestinal barrier injury during hemorrhage shock by regulating neutrophil extracellular trap formation via the ROS/MAPK/PAD4 pathway. Int Immunopharmacol 2024; 130:111688. [PMID: 38394886 DOI: 10.1016/j.intimp.2024.111688] [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: 12/15/2023] [Revised: 01/31/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024]
Abstract
Magnesium hydride (MgH2) is a hydrogen storage material that is known for its high capacity and safety and is capable of releasing hydrogen in a controlled manner when administered orally. This release of hydrogen has been associated with a range of biological effects, including anti-inflammatory properties, antioxidant activity, and protection of the intestinal barrier. Previous research has shown that neutrophil extracellular traps (NETs) play a role in the dysfunction of the intestinal barrier in conditions such as sepsis and critical illnesses. However, it remains unclear as to whether MgH2 can protect the intestinal barrier by inhibiting NET formation, and the underlying mechanisms have yet to be elucidated. A rat model of hemorrhagic shock was created, and pretreatment or posttreatment procedures with MgH2 were performed. After 24 h, samples from the small intestine and blood were collected for analysis. In vitro, human neutrophils were incubated with either phorbol-12-myristate-13-acetate (PMA) or MgH2. Reactive oxygen species generation and the expression of key proteins were assessed. The results demonstrated that MgH2 administration led to a decrease in inflammatory cytokines in the serum and mitigated distant organ dysfunction in rats with HS. Furthermore, MgH2 treatment reversed histopathological damage in the intestines, improved intestinal permeability, and enhanced the expression of tight junction proteins (TJPs) during HS. Additionally, MgH2 treatment was found to suppress NET formation in the intestines. In vitro pretreatment with MgH2 alleviated intestinal monolayer barrier disruption that was induced by NETs. Mechanistically, MgH2 pretreatment reduced ROS production and NET formation, inhibited the activation of ERK and p38, and suppressed the expression of the PAD4 protein. These findings indicated that MgH2 may inhibit NET formation in a ROS/MAPK/PAD4-dependent manner, which reduces NET-related intestinal barrier damage, thus offering a novel protective role in preventing intestinal barrier dysfunction during HS.
Collapse
Affiliation(s)
- Changkui Cao
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China; Department of Emergency, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Pan Yu
- Department of Burn and Plastic Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chengnan Chu
- Department of Emergency, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Zhenjie Wang
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Weiqi Xu
- Department of Emergency, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Feng Cheng
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Heng Zhao
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China
| | - Zhaolei Qiu
- Department of Emergency Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu City, Anhui Province, China.
| |
Collapse
|
20
|
Zhang Y, Zhao H, Fu X, Wang K, Yang J, Zhang X, Wang H. The role of hydrogen sulfide regulation of pyroptosis in different pathological processes. Eur J Med Chem 2024; 268:116254. [PMID: 38377826 DOI: 10.1016/j.ejmech.2024.116254] [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: 10/03/2023] [Revised: 01/31/2024] [Accepted: 02/15/2024] [Indexed: 02/22/2024]
Abstract
Pyroptosis is one kind of programmed cell death in which the cell membrane ruptures and subsequently releases cell contents and pro-inflammatory cytokines including IL-1β and IL-18. Pyroptosis is caused by many types of pathological stimuli, such as hyperglycemia (HG), oxidative stress, and inflammation, and is mediated by gasdermin (GSDM) protein family. Increasing evidence indicates that pyroptosis plays an important role in multiple diseases, such as cancer, kidney diseases, inflammatory diseases, and cardiovascular diseases. Therefore, the regulation of pyroptosis is crucial for the occurrence, development, and treatment of many diseases. Hydrogen sulfide (H2S) is a biologically active gasotransmitter following carbon monoxide (CO) and nitrogen oxide (NO) in mammalian tissues. So far, three enzymes, including 3-mercaptopyruvate sulphurtransferase (3-MST), cystathionine γ- Lyase (CSE), and Cystine β-synthesis enzyme (CBS), have been found to catalyze the production of endogenous H2S in mammals. H2S has been reported to have multiple biological functions including anti-inflammation, anti-oxidative stress, anti-apoptosis and so on. Hence, H2S is involved in various physiological and pathological processes. In recent years, many studies have demonstrated that H2S plays a critical role by regulating pyroptosis in various pathological processes, such as ischemia-reperfusion injury, alcoholic liver disease, and diabetes cardiomyopathy. However, the relevant mechanism has not been completely understood. Therefore, elucidating the mechanism by which H2S regulates pyroptosis in diseases will help understand the pathogenesis of multiple diseases and provide important new avenues for the treatment of many diseases. Here, we reviewed the progress of H2S regulation of pyroptosis in different pathological processes, and analyzed the molecular mechanism in detail to provide a theoretical reference for future related research.
Collapse
Affiliation(s)
- Yanting Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China; School of Clinical Medicine, Henan University, Kaifeng, Henan, 475004, China
| | - Huijie Zhao
- Institute of Chronic Disease Risks Assessment, Henan University, Jinming Avenue, Kaifeng, 475004, China
| | - Xiaodi Fu
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Kexiao Wang
- School of Clinical Medicine, Henan University, Kaifeng, Henan, 475004, China
| | - Jiahao Yang
- School of Clinical Medicine, Henan University, Kaifeng, Henan, 475004, China
| | | | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| |
Collapse
|
21
|
Shentu Y, Chen M, Wang H, Du X, Zhang W, Xie G, Zhou S, Ding L, Zhu Y, Zhu M, Zhang N, Du C, Ma J, Chen R, Yang J, Fan X, Gong Y, Zhang H, Fan J. Hydrogen sulfide ameliorates lipopolysaccharide-induced anxiety-like behavior by inhibiting checkpoint kinase 1 activation in the hippocampus of mice. Exp Neurol 2024; 371:114586. [PMID: 37898396 DOI: 10.1016/j.expneurol.2023.114586] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/19/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
Hydrogen sulfide (H2S), an endogenous gasotransmitter, exhibits the anxiolytic roles through its anti-inflammatory effects, although its underlying mechanisms remain largely elusive. Emerging evidence has documented that cell cycle checkpoint kinase 1 (Chk1)-regulated DNA damage plays an important role in the neurodegenerative diseases; however, there are few relevant reports on the research of Chk1 in neuropsychiatric diseases. Here, we aimed to investigate the regulatory role of H2S on Chk1 in lipopolysaccharide (LPS)-induced anxiety-like behavior focusing on inflammasome activation in the hippocampus. Cystathionine γ-lyase (CSE, a H2S-producing enzyme) knockout (CSE-/-) mice displayed anxiety-like behavior and activation of inflammasome-mediated inflammatory responses, manifesting by the increase levels of interleukin-1β (IL-1β), IL-6, and ionized calcium-binding adaptor molecule-1 (Iba-1, microglia marker) expression in the hippocampus. Importantly, expression of p-Chk1 and γ-H2AX (DNA damage marker) levels were also increased in the hippocampus of CSE-/- mice. LPS treatment decreased the expression of CSE and CBS while increased p-Chk1 and γ-H2AX levels and inflammasome-activated neuroinflammation in the hippocampus of mice. Moreover, p-Chk1 and γ-H2AX protein levels and cellular immunoactivity were significantly increased while CSE and CBS were markedly decreased in cultured BV2 cells followed by LPS treatment. Treatment of mice with GYY4137, a donor of H2S, inhibited LPS-induced increased in p-Chk1 and γ-H2AX levels, mitigated inflammasome activation and inflammatory responses as well as amelioration of anxiety-like behavior. Notably, SB-218078, a selective Chk1 inhibitor treatment attenuated the effect of LPS on inflammasome activation and inflammatory responses and the induction of anxiety-like behavior. Finally, STAT3 knockdown with AAV-STAT3 shRNA alleviated LPS-induced anxiety-like behavior and inhibited inflammasome activation in the hippocampus, and blockade of NLRP3 with MCC950 attenuated neuroinflammation induction and ameliorated LPS-induced anxiety-like behavior. Overall, this study indicates that downregulation of Chk1 activity by H2S activation may be considered as a valid strategy for preventing the progression of LPS-induced anxiety-like behavior.
Collapse
Affiliation(s)
- Yangping Shentu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Mengfan Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Hui Wang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xiaotong Du
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Institute of Cixi Biomedical Research, Wenzhou Medical University, Cixi, Zhejiang 315302, China
| | - Wenjing Zhang
- Renji College, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Guizhen Xie
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Shaoyan Zhou
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lu Ding
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yun Zhu
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Min Zhu
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Nan Zhang
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Congkuo Du
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jianshe Ma
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Ran Chen
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jinge Yang
- Department of Medical Technology, Jiangxi Medical College, Shangrao, Jiangxi 334709, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Hongyu Zhang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Institute of Cixi Biomedical Research, Wenzhou Medical University, Cixi, Zhejiang 315302, China.
| | - Junming Fan
- Institute of Hypoxia Medicine, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Institute of Cixi Biomedical Research, Wenzhou Medical University, Cixi, Zhejiang 315302, China.
| |
Collapse
|
22
|
Wang B, Chen SM, Yang SQ, Jiang JM, Zhang P, Zou W, Tang XQ. GDF11 mediates H 2S to prevent chronic stress-induced cognitive impairment by reducing hippocampal NLRP3/caspase-1-dependent pyroptosis. J Affect Disord 2024; 344:600-611. [PMID: 37827256 DOI: 10.1016/j.jad.2023.10.040] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/17/2023] [Accepted: 10/08/2023] [Indexed: 10/14/2023]
Abstract
BACKGROUND We previously revealed that hydrogen sulfide (H2S) attenuates chronic stress-induced cognitive impairment, but the underlying mechanism needs to be further clarified. Growth differentiation factor 11 (GDF11) plays an important regulatory role in cognitive function and that hippocampal NLRP3/caspase-1-mediated pyroptosis contributes to the pathogenesis of cognitive impairment. Hence, this research aimed to explore whether promoting GDF11 levels and suppressing hippocampal NLRP3/caspase-1-mediated pyroptosis mediate H2S to alleviate chronic stress-induced cognitive impairment. METHODS Sprague-Dawley rats were subjected to unpredictable chronic mild stress lasting four weeks to establish an animal model of chronic stress-induced cognitive impairment. Behavioral performance was assessed by the Y-maze test and the novel object recognition test. The expression levels of proteins were analyzed by Western blot analysis. The levels of IL-1β and IL-18 in the hippocampus were measured by ELISA. RESULTS NaHS upregulated the expression of GDF11 in the hippocampus of chronic unpredictable mild stress (CUMS)-exposed rats. Silencing GDF11 blocked NaHS-improved cognitive impairment in CUMS-exposed rats, according to the Y-maze test and the novel object recognition test. Furthermore, NaHS mitigated NLRP3/caspase-1-mediated pyroptosis in the hippocampus of CUMS-exposed rats and this effect was reversed by silencing GDF11. Moreover, overexpression of GDF11 alleviated CUMS-induced cognitive impairment and NLRP3/caspase-1-mediated hippocampal pyroptosis. CONCLUSIONS GDF11 mediates H2S to attenuate chronic stress-induced cognitive impairment via inhibiting hippocampal NLRP3/caspase-1-mediated pyroptosis.
Collapse
Affiliation(s)
- Bo Wang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China; The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Si-Min Chen
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - San-Qiao Yang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Jia-Mei Jiang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Ping Zhang
- The Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Wei Zou
- The Affiliated Nanhua Hospital, Department of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China.
| | - Xiao-Qing Tang
- The First Affiliated Hospital, Institute of Neurology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China; Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Institute of Neuroscience, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China; The Second Affiliated Hospital, Institute of Cerebral Disease, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China.
| |
Collapse
|
23
|
Zhang N, Zhou Z, Huang Y, Wang G, Tang Z, Lu J, Wang C, Ni X. Reduced hydrogen sulfide production contributes to adrenal insufficiency induced by hypoxia via modulation of NLRP3 inflammasome activation. Redox Rep 2023; 28:2163354. [PMID: 36661247 PMCID: PMC9869992 DOI: 10.1080/13510002.2022.2163354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Objective: Adrenocortical responsiveness is critical for maintaining glucocorticoids production and homeostasis during stress. We sought to investigate adrenocortical responsiveness during hypoxia in mice and the mechanisms responsible for the regulation of adrenal responsiveness.Methods: (1) Adult male WT mice were randomly divided into four groups: normoxia, hypoxia (24h), hypoxia (72h), hypoxia (72h) + GYY4137(hydrogen sulfide (H2S) donor, 133mmol/kg/day); (2) WT mice were randomly divided into four groups: sham, adrenalectomy (ADX), sham+hypoxia, ADX+hypoxia; (3) Cse-/- mice were randomly divided into two groups: Cse-/-, Cse-/- +GYY4137.Results: The circulatory level of corticosteroid induced by ACTH stimulation was significantly reduced in the mice with hypoxia compared with control mice. The mortality rate induced by lipopolysaccharide (LPS) increased during hypoxia. Cystathionine-γ-lyase (CSE) expression was significantly reduced in adrenal glands during hypoxia. GYY4137 treatment significantly increased adrenal responsiveness and attenuated NLRP3 inflammasome activation in mice treated by hypoxia and Cse-/- mice. Furthermore, The sulfhydrated level of PSMA7 in adrenal gland was decreased in the mice with hypoxia and Cse-/- mice. PSMA7 was S-sulfhydrated at cysteine 70. Blockage of S-sulfhydration of PSMA7 increased NLRP3 expression in adrenocortical cells.Conclusion: Reduced H2S production mediated hypo-adrenocortical responsiveness and NLRP3 inflammasome activation via PAMA7 S-sulfhydration during hypoxia.
Collapse
Affiliation(s)
- Ningning Zhang
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China,Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Zhan Zhou
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China
| | - Yan Huang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Gang Wang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China
| | - Zhengshan Tang
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China
| | - Jianqiang Lu
- The Key Laboratory of Exercise and Health Sciences of Ministry of Education, School of Kinesiology, Shanghai University of Sport, Shanghai, People’s Republic of China
| | - Changnan Wang
- Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China, Changnan Wang Department of Physiology, Navy Medical University, Shanghai200433, People’s Republic of China; Xin Ni
| | - Xin Ni
- National Clinical Research Center for Geriatric Disorders, Central South University Xiangya Hospital, Changsha, People’s Republic of China,International Collaborative Research Center for Medical Metabolomics, Central South University Xiangya Hospital, Changsha, People’s Republic of China,Department of Physiology, Navy Medical University, Shanghai, People’s Republic of China, Changnan Wang Department of Physiology, Navy Medical University, Shanghai200433, People’s Republic of China; Xin Ni
| |
Collapse
|
24
|
Zhang K, Li M, Yin K, Wang M, Dong Q, Miao Z, Guan Y, Wu Q, Zhou Y. Hyperoside mediates protection from diabetes kidney disease by regulating ROS-ERK signaling pathway and pyroptosis. Phytother Res 2023; 37:5871-5882. [PMID: 37646382 DOI: 10.1002/ptr.7993] [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: 05/15/2023] [Revised: 07/28/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
Renal tubular injury is a key factor in the progression of diabetic kidney disease to end-stage renal disease. Hyperoside, a natural flavonol glycoside in various plants, is a potentially effective drug for the clinical treatment of diabetic kidney disease. However, the specific mechanisms remain unknown. Therefore, this study will explore the effect and mechanism of hyperoside on renal tubulointerstitium in diabetic kidney disease. db/db mouse (C57BL/KsJ) is a model of type 2 diabetes resulting from Leptin receptor point mutations, with the appearance of diabetic kidney disease. Therefore, db/db mice were used for in vivo experimental studies. In vitro, human renal tubular epithelial cells were incubated with bovine serum albumin to simulate the injury of renal tubular epithelial cells caused by excessive albumin in primary urine. The experimental results showed that hyperoside could improve kidney function and reduce kidney tissue damage in mice, and could inhibit oxidative stress, extracellularly regulated protein kinases 1/2 signaling activation, and pyroptosis in human renal tubular epithelial cells. Therefore, hyperoside inhibited oxidative stress by regulating the activation of the extracellularly regulated protein kinases 1/2/mitogen-activated protein kinase signaling pathway, thereby alleviating proteinuria-induced pyroptosis in renal tubular epithelial cells. This study provides novel evidence that could facilitate the clinical application of hyperoside in diabetic kidney disease treatment.
Collapse
Affiliation(s)
- Kejia Zhang
- Public Experimental Research Center, Xuzhou Medical University, Xuzhou, China
| | - MiaoMiao Li
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou, China
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Kaiwen Yin
- The Second Clinical Medical School, Xuzhou Medical University, Xuzhou, China
| | - Minjie Wang
- The Second Clinical Medical School, Xuzhou Medical University, Xuzhou, China
| | - Qiuchi Dong
- The Second Clinical Medical School, Xuzhou Medical University, Xuzhou, China
| | - Zilan Miao
- The Second Clinical Medical School, Xuzhou Medical University, Xuzhou, China
| | - Yubo Guan
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou, China
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| | - Qi Wu
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Yao Zhou
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou, China
- Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
25
|
Zhou Z, Li Q. The Role of Pyroptosis in the Pathogenesis of Kidney Diseases. KIDNEY DISEASES (BASEL, SWITZERLAND) 2023; 9:443-458. [PMID: 38089443 PMCID: PMC10712988 DOI: 10.1159/000531642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 06/12/2023] [Indexed: 01/21/2025]
Abstract
BACKGROUND Recently, in addition to apoptosis and necrosis, several other forms of cell death have been discovered, such as necroptosis, autophagy, pyroptosis, and ferroptosis. These cell death modalities play diverse roles in kidney diseases. Pyroptosis is a newly described type of proinflammatory programmed necrosis. Further exploring pyroptosis is helpful to slow the progression of kidney diseases and reduce their complications. SUMMARY Pyroptosis is mainly mediated by the cleavage of gasdermin D (GSDMD) along with downstream inflammasome activation. Activated caspase-1 induces the release of cytokines by cleaving GSDMD. Inflammation is a major pathogenic mechanism for kidney diseases. Increasing evidence corroborated that pyroptosis was closely related to the progression of renal diseases, including acute kidney injury, renal fibrosis, diabetic nephropathy, and kidney cancer. In this paper, we reviewed the role and the therapeutic treatment of pyroptosis in renal diseases. KEY MESSAGES The better understanding of the progress and new intervention approaches of pyroptosis in kidney diseases may pave the way for new therapeutic opportunities in clinical practice.
Collapse
Affiliation(s)
- Zhuanli Zhou
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qin Li
- Department of Clinical Pharmacy, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
26
|
Hunter CE, Mesfin FM, Manohar K, Liu J, Shelley WC, Brokaw JP, Pecoraro AR, Hosfield BD, Markel TA. Hydrogen Sulfide Improves Outcomes in a Murine Model of Necrotizing Enterocolitis via the Cys440 Residue on Endothelial Nitric Oxide Synthase. J Pediatr Surg 2023; 58:2391-2398. [PMID: 37684170 PMCID: PMC10841167 DOI: 10.1016/j.jpedsurg.2023.08.006] [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: 05/16/2023] [Revised: 07/31/2023] [Accepted: 08/13/2023] [Indexed: 09/10/2023]
Abstract
BACKGROUND Hydrogen sulfide (H2S) has been shown to improve outcomes in a murine model of necrotizing enterocolitis (NEC). There is evidence in humans that H2S relies on endothelial nitric oxide synthase (eNOS) to exert its protective effects, potentially through the persulfidation of eNOS at the Cysteine 443 residue. We obtained a novel mouse strain with a mutation at this residue (eNOSC440G) and hypothesized that this locus would be critical for GYY4137 (an H2S donor) to exert its protective effects. METHODS Necrotizing enterocolitis was induced in 5-day old wild type (WT) and eNOSC440G mice using intermittent exposure to hypoxia and hypothermia in addition to gavage formula feeds. On postnatal day 9, mice were humanely euthanized. Data collected included daily weights, clinical sickness scores, histologic lung injury, intestinal injury (macroscopically and histologically), and intestinal perfusion. During the NEC model, pups received daily intraperitoneal injections of either GYY4137 (50 mg/kg) or PBS (vehicle). Data were tested for normality and compared using t-test or Mann-Whitney, and a p-value <0.05 was considered significant. RESULTS In WT mice, the administration of GYY4137 significantly improved clinical sickness scores, attenuated intestinal and lung injury, and improved mesenteric perfusion compared to vehicle (p < 0.05). In eNOSC440G mice, the treatment and vehicle groups had similar clinical sickness scores, intestinal and lung injury scores, and intestinal perfusion. CONCLUSIONS GYY4137 administration improves clinical outcomes, attenuates intestinal and lung injury, and improves perfusion in a murine model of necrotizing enterocolitis. The beneficial effects of GYY4137 are dependent on the Cys440 residue of eNOS.
Collapse
Affiliation(s)
- Chelsea E Hunter
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fikir M Mesfin
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Krishna Manohar
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jianyun Liu
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - John P Brokaw
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anthony R Pecoraro
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Brian D Hosfield
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Troy A Markel
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA; Riley Hospital for Children at Indiana University Health, Indianapolis, IN, USA.
| |
Collapse
|
27
|
Ramalingam V. NLRP3 inhibitors: Unleashing their therapeutic potential against inflammatory diseases. Biochem Pharmacol 2023; 218:115915. [PMID: 37949323 DOI: 10.1016/j.bcp.2023.115915] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The NOD-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome has been linked to the release of pro-inflammatory cytokines and is essential for innate defence against infection and danger signals. These secreted cytokines improve the inflammatory response caused by tissue damage and associated inflammation. Consequently, the development of NLRP3 inflammasome inhibitors are viable option for the treatment of diverse inflammatory disorders. The significant anti-inflammatory effects of the NLRP3 inhibitors have severe side effects. Hence, the application of NLRP3 inhibitors against inflammatory disease has not yet been understood and most of the developed inhibitors are unsuccessful in clinical trials. The processes behind the NLRP3 complex, priming, and activation are the main emphasis of this review, which also covers therapeutical inhibitors of the NLRP3 inflammasome and potential therapeutic strategies for directing the NLRP3 inflammasome towards clinical development.
Collapse
Affiliation(s)
- Vaikundamoorthy Ramalingam
- Department of Natural Products and Medicinal Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
28
|
Li F, Zhang Y, Ruan H, He Y, Zhan L, Chen S, Wang T, Qiu J, Guo Z, Wang D, He X. Addition of a liver to the normothermic perfusion circuit reduces renal pro-inflammatory factors. Artif Organs 2023; 47:1732-1741. [PMID: 37553847 DOI: 10.1111/aor.14626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/25/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND Normothermic machine perfusion (NMP) provides a novel platform to preserve isolated organs in an artificial condition. Our study aimed to explore the interaction between the liver and kidney at an ex vivo organ level by adding a liver to the kidney NMP circuit. METHODS Porcine kidney and liver obtained from abattoir were subjected to 9 h NMP after suffering 30-min warm ischemia time and 90-min cold ischemia time. The liver-kidney NMP group (n = 5) and the single-kidney NMP group (n = 5) were designed. During the NMP, perfusion parameters, blood gas analysis, and tissue samples were compared. RESULTS The perfusate of both groups remained stable, and continuous urine production was observed during NMP. In the liver-kidney NMP group, the lactate level was low, while blood urea nitrogen increased and glucose levels decreased. After the NMP, the renal tissue in the liver-kidney group exhibited fewer histological changes such as tubular epithelium vacuolization, along with reduced expression of IL-6, IL-8, IL-1β, NLRP3, and GSDMD. CONCLUSIONS Our results indicated that the expression of renal pro-inflammatory factors was reduced in the liver-kidney NMP system.
Collapse
Affiliation(s)
- Fangcong Li
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Yimin Zhang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hehuan Ruan
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Yu He
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Liqiang Zhan
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Shirui Chen
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Tielong Wang
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Jiang Qiu
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Zhiyong Guo
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Dongping Wang
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Xiaoshun He
- Organ Transplant Centre, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant, Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| |
Collapse
|
29
|
Zhang L, Rao J, Liu X, Wang X, Wang C, Fu S, Xiao J. Attenuation of Sepsis-Induced Acute Kidney Injury by Exogenous H 2S via Inhibition of Ferroptosis. Molecules 2023; 28:4770. [PMID: 37375325 DOI: 10.3390/molecules28124770] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Sepsis-associated acute kidney injury (SA-AKI) results in significant morbidity and mortality, and ferroptosis may play a role in its pathogenesis. Our aim was to examine the effect of exogenous H2S (GYY4137) on ferroptosis and AKI in in vivo and in vitro models of sepsis and explore the possible mechanism involved. Sepsis was induced by cecal ligation and puncture (CLP) in male C57BL/6 mice, which were randomly divided into the sham, CLP, and CLP + GYY4137 group. The indicators of SA-AKI were most prominent at 24 h after CLP, and analysis of the protein expression of ferroptosis indicators showed that ferroptosis was also exacerbated at 24 h after CLP. Moreover, the level of the endogenous H2S synthase CSE (Cystathionine-γ-lyase) and endogenous H2S significantly decreased after CLP. Treatment with GYY4137 reversed or attenuated all these changes. In the in vitro experiments, LPS was used to simulate SA-AKI in mouse renal glomerular endothelial cells (MRGECs). Measurement of ferroptosis-related markers and products of mitochondrial oxidative stress showed that GYY4137 could attenuate ferroptosis and regulate mitochondrial oxidative stress. These findings imply that GYY4137 alleviates SA-AKI by inhibiting ferroptosis triggered by excessive mitochondrial oxidative stress. Thus, GYY4137 may be an effective drug for the clinical treatment of SA-AKI.
Collapse
Affiliation(s)
- Li Zhang
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Jin Rao
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Xuwen Liu
- School of Medicine, Guangxi University, Nanning 530004, China
| | - Xuefu Wang
- School of Health Sciences and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Changnan Wang
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Shangxi Fu
- Department of Urology, Kidney Transplantation Center, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Jian Xiao
- School of Medicine, Guangxi University, Nanning 530004, China
- Department of Cardiothoracic Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| |
Collapse
|
30
|
Chen J, Luo W, Hu C, Ren M, Xu H, Xu X, Li W, Chen Y, Shao J, Xiao Z, Lv X, Liang G. Tanshinone IIA analogue 15a inhibits NLRP3-mediated inflammation by activating mitophagy in macrophages to alleviate acute tubular necrosis. Int Immunopharmacol 2023; 118:110065. [PMID: 37004347 DOI: 10.1016/j.intimp.2023.110065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/13/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
BACKGROUND Acute tubular necrosis (ATN) is a common type of acute renal failure. Recent studies have shown that NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome-mediated pyroptosis in macrophages plays a crucial role in the progression of ATN. Previously, we synthesized an anti-inflammatory compound 15a based on Tanshinone IIA (Tan IIA). In the present study, we found that compound 15a exhibited a greater inhibitory effect on NLRP3-mediated pyroptosis than Tan IIA in vitro. METHODS C57BL/6 and NLRP3-knockout (NLRP3-KO) mice were intraperitoneally injected with LPS or folic acid (FA) to develop ATN. In vitro, bone marrow-derived macrophages (BMDMs) were treated with LPS for 3 h and then treated with ATP for 0.5 h. RESULTS We explored the mechanism by which compound 15a inhibited NLRP3 inflammasome in BMDMs as well as its renal protective effect against ATN in mice. We found that compound 15a exhibited a protective effect on mitochondria and reduced the production of mitochondrial reactive oxygen species (mtROS). Moreover, we revealed that compound 15a remarkably reduced the production of mtROS by promoting mitophagy, which resulted in the inhibition of NLRP3 inflammasome to alleviates ATN in mice. CONCLUSION In summary, compound 15a inhibited NLRP3-mediated inflammation by activating mitophagy in macrophages to alleviate ATN. Our results identified compound 15a as a promising candidate for the treatment of NLRP3-driven ATN.
Collapse
|
31
|
Emre Aydıngöz S, Teimoori A, Orhan HG, Efe OE, Kibaroğlu S, Erdem ŞR. Effect of hydrogen sulfide on ischemia-reperfusion injury of kidney: A systematic review and meta-analysis of in vivo animal studies. Eur J Pharmacol 2023; 943:175564. [PMID: 36736943 DOI: 10.1016/j.ejphar.2023.175564] [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: 11/14/2022] [Revised: 01/14/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Hydrogen sulfide (H2S) has been shown to be effective against kidney ischemia-reperfusion injury (IRI) in animal studies. We aimed to evaluate the current evidence from in vivo animal studies for the protective effects of H2S against kidney IRI by systematically reviewing the literature and performing a meta-analysis. Based on the preregistered protocol (PROSPERO: CRD42021295469); PubMed, Medline, Embase, Web of Science, and Scopus were searched to identify in vivo animal studies evaluating the effect of H2S against kidney IRI. Standardized mean difference (SMD) with 95% confidence interval (CI) was calculated and pooled using random-effects meta-analysis. Twenty-two articles complied with eligibility criteria, from which the creatinine levels of 152 control animals and 182 animals treated with H2S from 27 individual experiments were pooled. H2S treatment significantly decreased serum creatinine (SMD = -1.82 [95% CI -1.12, -2.51], p < 0.0001), blood urea nitrogen (-2.50 [-1.46, -3.54], p < 0.0001), tissue malondialdehyde (-2.59 [-3.30, -1.88], p < 0.0001), tunel positive cells (-3.16 [-4.38, -1.94], p < 0.0001), and tubular damage score (-2.01 [-3.03, -0.99], p < 0.0001). There was a high heterogeneity across studies (I2 = 83.5% for serum creatinine level). In meta-regression analysis, the type of H2S donor and its application time accounted for 11.3% (p = 0.025) and 16.6% (p = 0.039) of heterogeneity, respectively. Accordingly, H2S protects the kidney against IRI only if it is given as GYY4137 before or during ischemia. Although H2S is a potential candidate against kidney IRI, further well-designed preclinical studies focusing on GYY4137 are warranted before clinical implication.
Collapse
Affiliation(s)
- Selda Emre Aydıngöz
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey.
| | - Arıyan Teimoori
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Halit Güner Orhan
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Oğuzhan Ekin Efe
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Seda Kibaroğlu
- Department of Pharmacology, Başkent University Institute of Health Sciences, Ankara, Turkey
| | - Ş Remzi Erdem
- Department of Medical Pharmacology, Başkent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
32
|
Li W, Yang K, Li B, Wang Y, Liu J, Chen D, Diao Y. Corilagin alleviates intestinal ischemia/reperfusion-induced intestinal and lung injury in mice via inhibiting NLRP3 inflammasome activation and pyroptosis. Front Pharmacol 2022; 13:1060104. [PMID: 36506567 PMCID: PMC9727192 DOI: 10.3389/fphar.2022.1060104] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Intestinal ischemia reperfusion (II/R) is a clinical emergency that frequently occurs in a variety of clinical conditions. Severe intestinal injury results in the release of cytotoxic substances and inflammatory mediators which can activate local inflammatory response and bacterial translocation. This triggers multi-organ failure, including lung injury, which is a common complication of II/R injury and contributes to the high mortality rate. Corilagin (Cor) is a natural ellagitannin found in a variety of plants. It has many biological and pharmacological properties, including antioxidant, anti-inflammatory and anti-apoptosis activities. However, no studies have evaluated the effects and molecular mechanisms of Cor in alleviating II/R-induced intestinal and lung damage. In this study, Cor was found to significantly alleviate II/R-induced pathological damage, inflammatory response, oxidative stress, NLRP3 inflammasome activation, and pyroptosis in intestinal and lung tissues both in vivo and in vitro. Further, Cor inhibited the NLRP3 inflammasome activation and pyroptosis in RAW264.7 and MLE-12 cells induced by LPS/nigericin and that in IEC-6 cells induced by nigericin, indicating an amelioration of Cor in II/R-induced intestinal and lung injury via inhibiting NLRP3 inflammasome activation and pyroptosis. Thus, Cor might be a potential therapeutic agent for II/R-induced inflammation and tissue injury.
Collapse
Affiliation(s)
- Wenlian Li
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Kejia Yang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Bin Li
- College of Pharmacy, Dalian Medical University, Dalian, China,Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China
| | - Yunxiang Wang
- College of Pharmacy, Dalian Medical University, Dalian, China
| | - Jing Liu
- College of Pharmacy, Dalian Medical University, Dalian, China,Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China,*Correspondence: Jing Liu, ; Yunpeng Diao,
| | - Dapeng Chen
- Comparative Medicine Department of Researching and Teaching, Dalian Medical University, Dalian, China
| | - Yunpeng Diao
- College of Pharmacy, Dalian Medical University, Dalian, China,Dalian Anti-Infective Traditional Chinese Medicine Development Engineering Technology Research Center, Dalian, China,*Correspondence: Jing Liu, ; Yunpeng Diao,
| |
Collapse
|
33
|
Luo S, Yang M, Han Y, Zhao H, Jiang N, Li L, Chen W, Li C, Yang J, Liu Y, Liu C, Zhao C, Sun L. β-Hydroxybutyrate against Cisplatin-Induced acute kidney injury via inhibiting NLRP3 inflammasome and oxidative stress. Int Immunopharmacol 2022; 111:109101. [PMID: 35940076 DOI: 10.1016/j.intimp.2022.109101] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 02/07/2023]
Abstract
Cisplatin, as a commonly used anticancer drug, can easily lead to acute kidney injury (AKI), and has received more and more attention in clinical practice. β-hydroxybutyric acid (BHB) is a metabolite in the body and acts as an inhibitor of oxidative stress and NLRP3 inflammasome, reducing inflammatory responses and apoptosis. However, the role of BHB in cisplatin-induced AKI is currently not fully elucidated. In this study, C57BL/6 male mice were randomly divided into normal control group, cisplatin-induced AKI group and AKI with BHB treatment group. Compared to the control, cisplatin-treated mice exhibited high level of serum creatinine, blood urea nitrogen and severe tubular injury, which accompanied with significantly increased expression level of NLRP3, IL-1β, IL-18, BAX, cleaved-caspase 3, as well as aggravated oxidative stress and renal tubular cell apoptosis. However, these changes were significantly improved in that of BHB treatment. In vitro, our study showed that the expression of cleaved-caspase3, IL-1β and IL-18 were significantly increased in human proximal tubular epithelial cell line (HK-2) treated with cisplatin compared with the control group, while decreased in cells treated with BHB. Furthermore, a significantly increased expression of cGAS and STING in HK-2 cells treated with cisplatin were found, whereas notably decreased in cells treated with BHB. This data indicates that BHB protects against cisplatin-induced AKI and renal tubular damage mediated by NLRP3 inflammasome and cGAS-STING pathway.
Collapse
Affiliation(s)
- Shilu Luo
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Ming Yang
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yachun Han
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Hao Zhao
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Na Jiang
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Li Li
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Wei Chen
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chenrui Li
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Jinfei Yang
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yan Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chongbin Liu
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Chanyue Zhao
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, the Second Xiangya Hospital, Central South University, Changsha, Hunan, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China.
| |
Collapse
|
34
|
Zhao H, Yang Y, Si X, Liu H, Wang H. The Role of Pyroptosis and Autophagy in Ischemia Reperfusion Injury. Biomolecules 2022; 12:biom12071010. [PMID: 35883566 PMCID: PMC9313059 DOI: 10.3390/biom12071010] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 02/07/2023] Open
Abstract
Pyroptosis is a process of programmed cell death mediated by gasdermin (GSDM) found in recent years. In the process of pyroptosis, caspase-1 or caspase-11/4/5 is activated, which cleaves gasdermin D and separates its N-terminal pore-forming domain (PFD). The oligomers of PFD bind to the cell membrane and form macropores on the membrane, resulting in cell swelling and membrane rupture. Increasing evidence indicates that pyroptosis is involved in many diseases, including ischemia reperfusion injury. Autophagy is a highly conserved catabolic process in eukaryotic cells. It plays an important role in the survival and maintenance of cells by degrading organelles, proteins, and macromolecules in the cytoplasm and recycling degradation products. Increasing evidence shows that dysfunctional autophagy participates in many diseases. Recently, autophagy and pyroptosis have been reported to play a vital role in the process of ischemia/reperfusion injury, but the related mechanisms are not completely clear. Therefore, this article reviews the role of autophagy and pyroptosis in ischemia–reperfusion injury and analyzes the related mechanisms to provide a basis for future research.
Collapse
Affiliation(s)
- Huijie Zhao
- Institute of Chronic Disease Risks Assessment, Henan University, Jinming Avenue, Kaifeng 475004, China;
| | - Yihan Yang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
| | - Xinya Si
- School of Stomatology, Henan University, Kaifeng 475004, China;
| | - Huiyang Liu
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
| | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng 475004, China; (Y.Y.); (H.L.)
- Correspondence:
| |
Collapse
|
35
|
Chen M, Rong R, Xia X. Spotlight on pyroptosis: role in pathogenesis and therapeutic potential of ocular diseases. J Neuroinflammation 2022; 19:183. [PMID: 35836195 PMCID: PMC9281180 DOI: 10.1186/s12974-022-02547-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/05/2022] [Indexed: 11/10/2022] Open
Abstract
Pyroptosis is a programmed cell death characterized by swift plasma membrane disruption and subsequent release of cellular contents and pro-inflammatory mediators (cytokines), including IL‐1β and IL‐18. It differs from other types of programmed cell death such as apoptosis, autophagy, necroptosis, ferroptosis, and NETosis in terms of its morphology and mechanism. As a recently discovered form of cell death, pyroptosis has been demonstrated to be involved in the progression of multiple diseases. Recent studies have also suggested that pyroptosis is linked to various ocular diseases. In this review, we systematically summarized and discussed recent scientific discoveries of the involvement of pyroptosis in common ocular diseases, including diabetic retinopathy, age-related macular degeneration, AIDS-related human cytomegalovirus retinitis, glaucoma, dry eye disease, keratitis, uveitis, and cataract. We also organized new and emerging evidence suggesting that pyroptosis signaling pathways may be potential therapeutic targets in ocular diseases, hoping to provide a summary of overall intervention strategies and relevant multi-dimensional evaluations for various ocular diseases, as well as offer valuable ideas for further research and development from the perspective of pyroptosis.
Collapse
Affiliation(s)
- Meini Chen
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Hunan Key Laboratory of Ophthalmology, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Changsha, 410008, Hunan, People's Republic of China
| | - Rong Rong
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.,Hunan Key Laboratory of Ophthalmology, Changsha, 410008, Hunan, People's Republic of China.,National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Changsha, 410008, Hunan, People's Republic of China
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China. .,Hunan Key Laboratory of Ophthalmology, Changsha, 410008, Hunan, People's Republic of China. .,National Clinical Research Center for Geriatric Diseases (Xiangya Hospital), Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
36
|
A novel fluorescent probe for real-time imaging of thionitrous acid under inflammatory and oxidative conditions. Redox Biol 2022; 54:102372. [PMID: 35728302 PMCID: PMC9214870 DOI: 10.1016/j.redox.2022.102372] [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: 04/30/2022] [Revised: 06/07/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Thionitrous acid (HSNO), a crosstalk intermediate of two crucial gasotransmitters nitric oxide and hydrogen sulfide, plays a critical role in redox regulation of cellular signaling and functions. However, real-time and facile detection of HSNO with high selectivity and sensitivity remains highly challenging. Herein we report a novel fluorescent probe (SNP-1) for HSNO detection. SNP-1 has a simple molecular structure, but showing strong fluorescence, a low detection limit, a broad linear detection range (from nanomolar to micromolar concentrations), ultrasensitivity, and high selectivity for HSNO in both aqueous media and cells. Benefiting from these unique features, SNP-1 could effectively visualize changes of HSNO levels in mouse models of acute ulcerative colitis and renal ischemia/reperfusion injury. Moreover, the good correlation between colonic HSNO levels and disease activity index demonstrated that HSNO is a promising new diagnostic agent for acute ulcerative colitis. Therefore, SNP-1 can serve as a useful fluorescent probe for precision detection of HSNO in various biological systems, thereby facilitating mechanistic studies, therapeutic assessment, and high-content drug screening for corresponding diseases. HSNO was the preferred intermediate to study crosstalk between H2S and NO. HSNO displayed translational potential for diagnosis and assessment of diseases. SNP-1 displayed excellent fluorescence performance for HSNO detection. SNP-1 could effectively image HSNO in cells and mouse models.
Collapse
|
37
|
Juan C, Zhu Y, Chen Y, Mao Y, Zhou Y, Zhu W, Wang X, Wang Q. Knocking down ETS Proto-oncogene 1 (ETS1) alleviates the pyroptosis of renal tubular epithelial cells in patients with acute kidney injury by regulating the NLR family pyrin domain containing 3 (NLRP3) transcription. Bioengineered 2022; 13:12927-12940. [PMID: 35611792 PMCID: PMC9275905 DOI: 10.1080/21655979.2022.2079242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Acute kidney injury (AKI) has a high mortality rate, but its pathogenesis remains unclear Lipopolysaccharide (LPS)-mediated renal tubular epithelial pyroptosis is involved in the pathogenesis of AKI. NLR family of pyrin domains containing 3 (NLRP3) plays an important role in pyroptosis. To further understand the transcriptional regulation mechanism of NLRP3, the peripheral blood of patients with AKI was analyzed in this study, showing that the levels of NLRP3 and cell pyroptosis in patients with AKI were significantly higher than those in normal controls. Furthermore, elevated levels of NLRP3 and cell pyroptosis were found in renal tubular epithelial cells after LPS treatment. Transcription factor ETS Proto-Oncogene 1 (ETS1) could bind to the upstream promoter transcription site of NLRP3 to transactivate NLRP3 in renal tubular epithelial cells. The cell pyroptosis level also decreased by knocking down ETS1. It is concluded that knocking down of ETS1 may reduce the renal tubular epithelial pyroptosis by regulating the transcription of NLRP3, thus relieving AKI. ETS1 is expected to be a molecular target for the treatment of AKI.
Collapse
Affiliation(s)
- Chenxia Juan
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ye Zhu
- Department of Nephrology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yan Chen
- Department of Nephrology, Jiangsu Province Geriatric Hospital, Jiangsu Province Official Hospital, Nanjing, Jiangsu, China
| | - Yan Mao
- Department of Pediatrics, the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yan Zhou
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Weiwei Zhu
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xufang Wang
- Department of Nephrology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Qian Wang
- Department of Pediatrics, Shanghai General Hospital, Shanghai, Minhang, China
| |
Collapse
|
38
|
Xu J, Zhao X, Jiang X, He L, Wu X, Wang J, Chen Q, Li Y, Zhang M. Tubastatin A Improves Post-Resuscitation Myocardial Dysfunction by Inhibiting NLRP3-Mediated Pyroptosis Through Enhancing Transcription Factor EB Signaling. J Am Heart Assoc 2022; 11:e024205. [PMID: 35322683 PMCID: PMC9075499 DOI: 10.1161/jaha.121.024205] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Background Myocardial dysfunction is the leading cause of early death following successful cardiopulmonary resuscitation (CPR) in people with cardiac arrest (CA), which is potentially driven by cell pyroptosis mediated by NOD‐like receptor pyrin domain 3 (NLRP3) inflammasome. Recently, histone deacetylase 6 (HDAC6) inhibition was shown to exert effective myocardial protection against regional ischemia/reperfusion injury. In this study, we investigated whether tubastatin A, a specific histone deacetylase 6 inhibitor, could improve postresuscitation myocardial dysfunction through the inhibition of NLRP3‐mediated cell pyroptosis and its modulation mechanism. Methods and Results Healthy male white domestic swine were used to establish the model of CA/CPR in vivo, and the H9c2 cardiomyocyte hypoxia/reoxygenation model was used to simulate the CA/CPR process in vitro. Consequently, tubastatin A inhibited NLRP3 inflammasome activation, decreased proinflammatory cytokines production and cell pyroptosis, and increased cell survival after hypoxia/reoxygenation in H9c2 cardiomyocytes in vitro. In addition, tubastatin A increased the acetylated levels of transcription factor EB and its translocation to the nucleus, and its protective effect above was partly abrogated by transcription factor EB short interfering RNA after hypoxia/reoxygenation in H9c2 cardiomyocytes. Similarly, tubastatin A promoted cardiac transcription factor EB nuclear translocation, inhibited NLRP3‐mediated cell pyroptosis, and mitigated myocardial dysfunction after CA/CPR in swine. Conclusions The inhibition of histone deacetylase 6 activity by tubastatin A limited NLRP3 inflammasome activation and cell pyroptosis probably through the enhancement of transcription factor EB signaling, and therefore improved myocardial dysfunction after CA/CPR.
Collapse
Affiliation(s)
- Jiefeng Xu
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province Hangzhou China.,Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine Hangzhou China
| | - Xue Zhao
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Department of Emergency Medicine Affiliated Hangzhou First People's Hospital Zhejiang University School of Medicine Hangzhou China
| | - Xiangkang Jiang
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province Hangzhou China.,Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine Hangzhou China
| | - Lu He
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province Hangzhou China.,Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine Hangzhou China
| | - Xinjie Wu
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Department of Emergency Medicine The First Hospital of Ninghai Ningbo China
| | | | - Qijiang Chen
- Department of Intensive Care Medicine The First Hospital of Ninghai Ningbo China
| | - Yulin Li
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province Hangzhou China.,Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine Hangzhou China
| | - Mao Zhang
- Department of Emergency Medicine Second Affiliated Hospital Zhejiang University School of Medicine Hangzhou China.,Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province Hangzhou China.,Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine Hangzhou China
| |
Collapse
|