1
|
Peng Z, Huang X, Pan Y, Li W, Hu H, Chen X, Zhang Z, Hu J, Qi Y, Chen W, Cui X, Liu H, Liang W, Ding G, Chen Z. USP22 promotes angiotensin II-induced podocyte injury by deubiquitinating and stabilizing HMGB1. Cell Signal 2025; 131:111771. [PMID: 40154587 DOI: 10.1016/j.cellsig.2025.111771] [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: 01/03/2025] [Revised: 03/13/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Chronic kidney disease (CKD) remains a significant global health burden, with hypertensive nephropathy (HN) as one of its primary causes. Podocyte injury is a key factor in the progression of CKD. However, the molecular mechanisms underlying angiotensin II-induced podocyte injury remain incompletely understood. Ubiquitin-specific protease 22 (USP22) has been reported to facilitate a range of cellular processes, including cell proliferation and apoptosis. However, the role of USP22 in HN pathogenesis is unclear. METHODS The expression of USP22 was assessed in kidney samples from hypertensive nephropathy patients, angiotensin II-induced hypertensive nephropathy mouse models, and cultured podocytes treated with angiotensin II. Podocyte-specific USP22 knockout mice were used to investigate the effects of USP22 deletion on podocyte injury and inflammation. RESULTS USP22 expression was significantly upregulated in kidneys of HN patients, angiotensin II-induced mouse models, and cultured podocytes. Podocyte-specific deletion of USP22 markedly reduced angiotensin II-induced podocyte injury and inflammatory responses. Furthermore, we identified high-mobility group box protein 1 (HMGB1) as a protein that interacts with USP22. USP22 deubiquitinated and stabilized HMGB1 through K48-linked ubiquitination. Downregulation of USP22 expression improved kidney function and pathological changes in HN by promoting HMGB1 degradation. CONCLUSION This study identifies USP22 as a key regulator of angiotensin II-induced podocyte injury and inflammation through its interaction with HMGB1. Our findings revealed that following glomerular injury, damage and shedding of tubular cells also occurred. Targeting the USP22-HMGB1 axis offers a promising therapeutic strategy for treating hypertensive nephropathy and other types of CKD.
Collapse
Affiliation(s)
- Zhuan Peng
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxiao Huang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yangbin Pan
- Division of Nephrology, Shanghai Pudong Hospital, Fudan University, Pudong Medical Center, Shanghai, China
| | - Weiwei Li
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongtu Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinghua Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Zongwei Zhang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Jijia Hu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Yue Qi
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjie Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofei Cui
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hongyan Liu
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Wei Liang
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China
| | - Guohua Ding
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| | - Zhaowei Chen
- Division of Nephrology, Renmin Hospital of Wuhan University, Wuhan, China; Nephrology and Urology Research Institute of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Zhao X, Wang L. mTOR/p70S6K signaling pathway promotes fibrillin-1 expression in AKI-to-CKD transition post CA/CPR. Cell Signal 2025; 128:111624. [PMID: 39880104 DOI: 10.1016/j.cellsig.2025.111624] [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: 07/22/2024] [Revised: 01/08/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
The possible involvement of mTOR/p70S6K signaling in mediating Fibrillin-1 expression during the transition from acute kidney injury (AKI) to chronic kidney disease (CKD) after cardiac arrest and cardiopulmonary resuscitation (CA/CPR). A CA/CPR AKI model was established using male C57BL/6 mice aged 8-12 weeks. The expression of Fibrillin-1 and activation of the mTOR/p70S6K signaling pathway in kidney tissues were assessed at different time points. Rapamycin, administered intraperitoneally, inhibited the mTOR/p70S6K signaling pathway in CA/CPR AKI mice. Tissue immunofluorescence and immunohistochemistry were used to detect the injury, fibrosis, and inflammatory cell infiltration in renal tissues. The expression level of Fibrillin-1 and components of the mTOR/p70S6K signaling pathway, while ELISA quantified levels of inflammatory factors in renal tissues. Results showed that Fibrillin-1 expression progressively increased alongside enhanced mTOR/p70S6K signaling in the renal tissues of CA/CPR AKI mice. Inhibition of mTOR/p70S6K signaling by rapamycin reduced Fibrillin-1 expression, collagen deposition, and α-SMA levels, alleviating renal injury and decreasing macrophage and T cell infiltration, as well as inflammatory factor production. Conversely, combining rapamycin with Fibrillin-1 overexpression exacerbated renal injury and increased inflammatory factor production. Activation of the mTOR/p70S6K pathway upregulates Fibrillin-1 expression, potentially facilitating the progression from AKI to CKD in CA/CPR mice.
Collapse
Affiliation(s)
- Xiaohui Zhao
- School of Basic Medicine, Jiamusi University, Jiamusi 154007, PR China
| | - Limin Wang
- School of Basic Medicine, Jiamusi University, Jiamusi 154007, PR China.
| |
Collapse
|
3
|
Wang Y, Yang Y, Meng T, Liu S, Liu J, Liu D, Laxman B, Chen S, Dong Z. β-caryophyllene reduces inflammation to protect against ischemic stroke by suppressing HMGB1 signaling. Mol Med 2025; 31:112. [PMID: 40128654 PMCID: PMC11931857 DOI: 10.1186/s10020-025-01171-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 03/15/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Ischemic stroke is characterized by high mortality and high disability rates and accounts for the vast majority of current stroke cases. Reperfusion after surgical treatment can cause serious secondary damage to ischemic stroke patients, but there are still no specific drugs for the clinical treatment of ischemic stroke. Inflammation plays a critical role in ischemia and reperfusion injury, highlighting the urgent need for new anti-inflammatory targets and therapeutic agents. High-mobility group box-1 (HMGB1) is highly expressed in both neuronal cell bodies and axons and has been found to have late proinflammatory effects; thus, the role of HMGB1 in stroke has recently become a hot research topic in critical care medicine. An increase in HMGB1 expression leads to the aggravation of inflammatory reactions after ischemic stroke. B-caryophyllene (BCP) is a natural drug with anti-inflammatory effects. However, whether HMGB1 is involved in the anti-inflammatory mechanism of BCP is still unknown. We aimed to investigate the relationship between HMGB1 and BCP in in vivo and in vitro ischemic stroke models. METHODS A middle cerebral artery embolism model was established in mice by thread thrombus, and primary neurons were subjected to oxygen‒glucose deprivation and reoxygenation (OGD/R) in vitro. In vitro, the HMGB1 DNA overexpression virus(GV-HMGB1)or the HMGB1 DNA silencing virus(RNAi-HMGB1)was injected into the lateral ventricles of mice.. RESULTS HMGB1 expression increases after ischemic stroke and further affects the expression of TLR4, RAGE and other related inflammatory factors, thus reducing the inflammatory response and ultimately protecting against injury. These results confirmed the effect of HMGB1 on TLR4/RAGE signaling and the subsequent regulation of inflammation, oxidative stress and apoptosis. Furthermore, BCP potentially alleviates ischemic brain damage by suppressing HMGB1/TLR4/RAGE signaling, reducing the expression of IL-1β/IL-6/TNF-α, and inhibiting neuronal death and the inflammatory response. CONCLUSION These data indicate that BCP exerts a protective effect against ischemic stroke-induced inflammatory injury by regulating the HMGB1/TLR4/RAGE signaling pathway, which provides new insights into the mechanisms of this therapeutic candidate for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Yuchun Wang
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, 400016, China.
| | - Yang Yang
- Department of Pharmacy, Chongqing Health Center for Women and Children, Women and Children's Hospital of Chongqing Medical University, Chongqing, 401147, China
| | - Tuo Meng
- Children's Hospital of Chongqing Medical University, Department of Child Health in Children's Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Shengwei Liu
- Department of Pharmacy, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, China
| | - Jingdong Liu
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, 400016, China
| | - Daohang Liu
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, 400016, China
| | - Bharati Laxman
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, 400016, China
| | - Sha Chen
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, 400016, China
| | - Zhi Dong
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
4
|
Huang M, Li J, Bai J, Du X, Xu J. NAT10 induces mitochondrial dysfunction in lung epithelial cells by acetylating HMGB1 to exacerbate Pseudomonas aeruginosa-induced acute lung injury. Microb Pathog 2025; 200:107364. [PMID: 39909291 DOI: 10.1016/j.micpath.2025.107364] [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: 07/24/2024] [Revised: 01/25/2025] [Accepted: 02/01/2025] [Indexed: 02/07/2025]
Abstract
BACKGROUND Pseudomonas aeruginosa (PA) is a major pathogen that causes pneumonia and acute lung injury (ALI). Dysregulated NAT10 expression is associated with inflammatory and infectious diseases, but its role in PA-induced ALI remains unclear. METHODS A mouse pneumonia model was established by intratracheal injection of PA, and lentivirus-mediated NAT10 interference and HMGB1 overexpression vectors were administered via the tail vein. Lung mechanics, protein content, total cell counts, neutrophil counts, inflammatory factor levels in bronchoalveolar lavage fluid (BALF), and lung bacterial load were assessed 24 h after PA injection. HE staining was performed to evaluate lung structural damage. Intracellular oxidative stress levels in mouse lung epithelial cells (TC-1 cells) were measured by detecting ROS and MDA levels. Mitochondrial function was analyzed by testing the mitochondrial membrane potential, cytoplasmic accumulation of cytochrome C, mtDNA copy number, and ATP production. An N4-acetylcytidine (ac4C)-RNA immunoprecipitation assay was conducted to assess the ac4C level of HMGB1 mRNA. RESULTS NAT10 deficiency hindered PA infection-induced increases in immune cell infiltration, inflammatory factor levels, bacterial load, and ultimately lung structural and functional damage. However, upregulation of HMGB1 effectively antagonized the protective effects of NAT10 silencing in vivo. NAT10 knockdown suppressed PA-induced oxidative stress, mitochondrial dysfunction, and apoptosis in vitro. Whereas, HMGB1 overexpression reversed the inhibitory effects of NAT10 downregulation on PA-induced TC-1 cell injury. Mechanistically, as an acetyltransferase, NAT10 enhanced HMGB1 mRNA stability and protein expression by promoting HMGB1 mRNA ac4C modification. CONCLUSION NAT10 facilitated mitochondrial dysfunction in lung epithelial cells and exacerbated PA-induced ALI by promoting the N4-acetylcytidine of HMGB1 mRNA.
Collapse
Affiliation(s)
- Miaoyi Huang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China; Department of Respiratory Medicine, Xi'an Central Hospital, Xi'an, China
| | - Jianying Li
- Department of Respiratory Medicine, Xi'an Chest Hospital, Xi'an, China
| | - Jie Bai
- Department of Emergency Medicine, Xi'an Central Hospital, Xi'an, China
| | - Xusheng Du
- Department of Geratology Medicine, Xi'an Central Hospital, Xi'an, China
| | - Jiru Xu
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
5
|
Chen L, Ye Z, Yang S, Xie J, Li H, Zhou X, Cheng F. RNA-Binding Protein Lgals3 , Ferroptosis, and AKI. J Am Soc Nephrol 2025:00001751-990000000-00559. [PMID: 39928414 DOI: 10.1681/asn.0000000638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/31/2025] [Indexed: 02/12/2025] Open
Abstract
Key Points
LGALS3 was increased in the process of AKI.The inhibition of LGALS3 alleviated kidney injury in vivo and in vitro.
LGALS3 contributed to kidney injury by binding to the 3′untranslated region of Nr4a1 through AAUAAA, leading to the activation of ferroptosis.
Background
AKI is a syndrome characterized by a precipitous decline in kidney function, posing a significant threat to patient survival. The role of RNA-binding protein in AKI remains insufficiently understood, and we found an important RNA-binding protein, LGALS3, that may mediate the progress of AKI.
Methods
Lgals3
−/− mice, Nr4a1
−/− mice, and cross-linking immunoprecipitation and high-throughput sequencing were performed to examine the role of Lgals3 in AKI and the targeted binding proteins.
Results
Lgals3 expression was notably elevated in vivo and in vitro AKI models. The inhibition of Lgals3 mitigated kidney injury in both in vivo and in vitro AKI models. Conversely, kidney-specific overexpression of Lgals3 exacerbated kidney damage. Mechanistically, Lgals3 bound to the 3′-untranslated region of Nr4a1 through AAUAAA, resulting in upregulation of Nr4a1 and subsequent enhancement of Bap1 transcription, facilitating ferroptosis in AKI. Moreover, knockout of Nr4a1 or inhibition of the region of AAUAAA by antisense oligonucleotide conferred protection against Lgals3-induced ferroptosis in AKI models.
Conclusions
LGALS3 contributed to kidney injury by binding to the 3′untranslated region of Nr4a1 through AAUAAA, leading to the activation of ferroptosis.
Collapse
Affiliation(s)
- Lijia Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
6
|
Hoenig MP, Brooks CR, Hoorn EJ, Hall AM. Biology of the proximal tubule in body homeostasis and kidney disease. Nephrol Dial Transplant 2025; 40:234-243. [PMID: 39066502 PMCID: PMC11852287 DOI: 10.1093/ndt/gfae177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Indexed: 07/28/2024] Open
Abstract
The proximal tubule (PT) is known as the workhorse of the kidney, for both the range and magnitude of the functions that it performs. It is not only responsible for reabsorbing most solutes and proteins filtered by glomeruli, but also for secreting non-filtered substances including drugs and uremic toxins. The PT therefore plays a pivotal role in kidney physiology and body homeostasis. Moreover, it is the major site of damage in acute kidney injury and nephrotoxicity. In this review, we will provide an introduction to the cell biology of the PT and explore how it is adapted to the execution of a myriad of different functions and how these can differ between males and females. We will then discuss how the PT regulates phosphate, glucose and acid-base balance, and the consequences of alterations in PT function for bone and cardiovascular health. Finally, we explore why the PT is vulnerable to ischemic and toxic insults, and how acute injury in the PT can lead to maladaptive repair, chronic damage and kidney fibrosis. In summary, we will demonstrate that knowledge of the basic cell biology of the PT is critical for understanding kidney disease phenotypes and their associated systemic complications, and for developing new therapeutic strategies to prevent these.
Collapse
Affiliation(s)
- Melanie P Hoenig
- Division of Nephrology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Craig R Brooks
- Department of Medicine at Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ewout J Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Switzerland. Zurich Kidney Center, University of Zurich, Zürich, Switzerland
| |
Collapse
|
7
|
Li Z, Mao C, Zhao Y, Zhao Y, Yi H, Liu J, Liang J. The STING antagonist SN-011 ameliorates cisplatin induced acute kidney injury via suppression of STING/NF-κB-mediated inflammation. Int Immunopharmacol 2025; 146:113876. [PMID: 39709905 DOI: 10.1016/j.intimp.2024.113876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/17/2024] [Accepted: 12/14/2024] [Indexed: 12/24/2024]
Abstract
Acute kidney injury (AKI) is a critical clinical syndrome associated with both innate and adaptive immune responses and thus increases mortality. Nevertheless, specific therapeutics for AKI are scarce so far. Recent studies have revealed that knockout of STING alleviate AKI, suggesting that STING could be an attractive target for AKI therapy. SN-011, a promising STING inhibitor, has not been reported in studies of its anti-AKI activity. In this study, we sought to examine the effects of SN-011 on AKI and explore its underlying mechanism. Our findings indicate that SN-011 could modulate the NF-κB and MAPK pathways, suppress the expression of inflammatory factors, and decrease ROS release in the cisplatin-induced cell model. In addition, SN-011 blocked the nuclear translocation of NF-κB p65, further mitigating the inflammatory response. In vivo, SN-011 enhanced survival rates and alleviated renal dysfunction. According to gene set enrichment analysis of sequencing data from mouse kidneys, we further confirm that SN-011 modulates the NF-κB and MAPK pathways. Our study suggests that SN-011 could be an attractive anti-inflammatory agent for further anti-AKI research.
Collapse
Affiliation(s)
- Ziyang Li
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Can Mao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yixin Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yanbin Zhao
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Hanyu Yi
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Jin Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| | - Jinqiang Liang
- Key Laboratory of Tropical Biological Resources of Ministry of Education and One Health Institute, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
8
|
Yao R, Pan JS, He RB, Hou BB, Suo XG, Li GX, Xia KG, Hu DK, Mao XK, Li W, Hao ZY. Pectolinarigenin alleviates calcium oxalate-induced renal inflammation and oxidative stress by binding to HIF-1α. Int Immunopharmacol 2024; 143:113284. [PMID: 39378657 DOI: 10.1016/j.intimp.2024.113284] [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: 07/04/2024] [Revised: 09/02/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
Calcium oxalate (CaOx) crystals are the main constituents of renal crystals in humans and induce tubular lumen damage in renal tubules, leading to renal calcium deposition and kidney stone formation. Oxidative stress and inflammation play important roles in regulating calcium oxalate-induced injury. Here, we evaluated the efficacy in inhibiting oxidation and inflammation of pectinolinarigenin, a biologically active natural metabolite, in CaOx nephrocalcinosis and further explored its targets of action. First, we developed cellular and mouse models of calcium oxalate renal nephrocalcinosis and identified the onset of oxidative stress and inflammation according to experimental data. We found that pectolinarigenin inhibited this onset while reducing renal crystal deposition. Network pharmacology was subsequently utilized to screen for hypoxia-inducible factor-1α (HIF-1α), a regulator involved in the body's release and over-oxidation of inflammatory factors. Finally, molecular docking, cellular thermal shift assay, and other experiments to detect HIF-1α expression showed that pectolinarigenin directly combined with HIF-1α and prevented downstream reactive oxygen species activation and release. Our results indicate that pectolinarigenin can target and inhibit HIF-1α-mediated inflammatory responses and oxidative stress damage and be a novel drug for CaOx nephrocalcinosis treatment.
Collapse
Affiliation(s)
- Rui Yao
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Jia-Shan Pan
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Ruo-Bing He
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Bing-Bing Hou
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Xiao-Guo Suo
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Guo-Xiang Li
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Kai-Guo Xia
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - De-Kai Hu
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Xi-Ke Mao
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China
| | - Wei Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Zong-Yao Hao
- Department of Urology, the First Affiliated Hospital of Anhui Medical University, Hefei, China; Institute of Urology, Anhui Medical University, Hefei, China; Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, China.
| |
Collapse
|
9
|
Pan X, Zhu R, Peng J, Liu H, Pan W, Jin Y, Pei J, Zhang L. Molecular mechanisms and potential targets of lycopene for alleviating renal ischemia-reperfusion injury revealed by network pharmacology and animal experiments. Int Immunopharmacol 2024; 143:113421. [PMID: 39442187 DOI: 10.1016/j.intimp.2024.113421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/25/2024]
Abstract
OBJECTIVE Renal IRI is one of the leading causes of AKI. How to effectively mitigate renal IRI is important for the recovery of renal function. The regulatory mechanism of lycopene, a natural antioxidant, in renal IRI is currently unknown. Therefore, we utilized network pharmacology and animal experiments to explore the possible mechanisms and potential targets of lycopene for alleviating renal IRI. METHODS We obtained lycopene-regulated genes and renal IRI-related genes from the CTD database and GeneCards database, respectively. Subsequently, the two were intersected and the intersecting genes we defined as lycopene-regulated genes in renal IRI. Next, we explored their potential biological functions and mechanisms through enrichment analysis. Meanwhile, we constructed a rat renal IRI model and validated the protective effects of lycopene and related mechanisms. To further explore the Hub genes regulated by lycopene, we constructed a PPI protein interactions network and characterized the Hub genes using Cytoscape software. We also verified the expression of Hub genes using animal experiments and molecular docking techniques. Finally, we constructed TF-Hub gene and miRNA-Hub gene regulatory networks. RESULTS We obtained a total of 255 lycopene-regulated genes and 327 renal IRI-related genes. The enrichment analysis revealed that they were closely related to the regulation of oxidative stress as well as the regulation of inflammatory factors. At the same time, the MAPK signaling pathway was significantly enriched. Next, we found in animal experiments that lycopene significantly alleviated the level of oxidative stress and inflammation during renal IRI, and had a protective effect on kidney damage. Also, we found that this protective effect may be achieved by inhibiting the MAPK signaling pathway. Next, we identified a total of five Hub genes using Cytoscape software: TNF, AKT1, MAPK3, IL6 and CASP3. Both animal experiments and molecular docking techniques demonstrated that lycopene can effectively regulate the expression of Hub genes. Finally, our constructed TF-Hub gene and miRNA-Hub gene regulatory network provide a theoretical basis for further regulation of Hub genes in follow-up. CONCLUSIONS This study suggests that lycopene is a promising option in mitigating renal IRI. Lycopene may exert protective effects by inhibiting the MAPK signaling pathway.
Collapse
Affiliation(s)
- Xingyu Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Rong Zhu
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jinpu Peng
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Hongli Liu
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Wenqing Pan
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Yuhan Jin
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China
| | - Jun Pei
- Department of Pediatric Surgrey, Guizhou Provincial People's Hospital, Guiyang 550002, China.
| | - Li Zhang
- Department of Nursing, Affiliated Hospital of Zunyi Medical University, Zunyi 563100, China; Nursing School of Zunyi Medical University, Zunyi 563100, China.
| |
Collapse
|
10
|
Liu X, Zhan P, Zhang Y, Jin H, Wang Y, Yang Y, Wang Z, Wang X, Xu Q, Zhen J, Sun R, Sun J, Liu M, Yi F. Lysosomal-Associated Protein Transmembrane 5, Tubular Senescence, and Progression of CKD. J Am Soc Nephrol 2024; 35:1655-1670. [PMID: 39078711 PMCID: PMC11617486 DOI: 10.1681/asn.0000000000000446] [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: 02/26/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
Key Points Lysosomal-associated protein transmembrane 5 (LAPTM5) is increased in tubular epithelial cells in CKD. Conditional knockout of Laptm5 in tubules attenuates kidney fibrosis in mice with CKD. LAPTM5 contributes to tubular senescence by inhibiting WWP2-mediated ubiquitination of notch1 intracellular domain. Background Tubular senescence is a major determinant of CKD, and identification of potential therapeutic targets involved in senescent tubular epithelial cells has clinical importance. Lysosomal-associated protein transmembrane 5 (LAPTM5) is a key molecule related to T- and B-cell receptor expression and inflammation. However, the expression pattern of LAPTM5 in the kidney and the contribution of LAPTM5 to the development of CKD are unknown. Methods Laptm5 −/− mice and tubule specific–Laptm5 knockout mice were used to examine the role of LAPTM5 in tubular senescence by establishing different experimental mouse CKD models. Results LAPTM5 expression was significantly induced in the kidney, especially in proximal tubules and distal convoluted tubules, from mice with aristolochic acid nephropathy, bilateral ischemia/reperfusion injury–induced CKD, or unilateral ureter obstruction. Tubule-specific deletion of Laptm5 inhibited senescence of tubular epithelial cells and alleviated tubulointerstitial fibrosis in aged mice. Moreover, Laptm5 deficiency ameliorated kidney injury and tubular senescence in mice with CKD. Mechanistically, LAPTM5 inhibited ubiquitination of notch1 intracellular domain by mediating WWP2 lysosomal degradation and then leading to cellular senescence in tubular epithelial cells. We also observed a higher expression of LAPTM5 in tubules from patients with CKD, and the level of LAPTM5 was correlated with kidney fibrosis and tubular senescence in people with CKD. Conclusions LAPTM5 contributed to tubular senescence by regulating the WWP2/notch1 intracellular domain signaling pathway and exacerbated kidney injury during the progression of CKD.
Collapse
Affiliation(s)
- Xiaohan Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ping Zhan
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yang Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Huiying Jin
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Youzhao Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yujie Yang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ziying Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xiaojie Wang
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qianqian Xu
- Department of Organ Transplantation, Qilu Hospital of Shandong University, Jinan, China
| | - Junhui Zhen
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Rong Sun
- The Second Hospital of Shandong University, Jinan, China
| | - Jinpeng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Min Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Fan Yi
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, China
- National Key Laboratory for Innovation and Transformation of Luobing Theory, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China
| |
Collapse
|
11
|
Deng R, Yang H, Zhong W, Zhou J, Huang G, Zeng K. CITED2 Mediates Metabolic Reprogramming in Renal Tubular Epithelial Cells via the AKT Signaling Pathway to Induce Sepsis-Associated Acute Kidney Injury. J Inflamm Res 2024; 17:9485-9505. [PMID: 39600684 PMCID: PMC11590677 DOI: 10.2147/jir.s486596] [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: 07/11/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Background Sepsis-associated acute kidney injury (S-AKI) is a prevalent and severe clinical complication in intensive care units (ICUs) and is associated with high mortality and poor prognosis. The dysfunction of renal tubular epithelial cells (TECs), particularly through their metabolic reprogramming, plays a critical role in the onset and progression of S-AKI. CITED2 is shown to regulate a variety of cellular processes, but its specific impact on TECs metabolism and S-AKI pathogenesis remains unclear. The aim of this study was to investigate the role of CITED2 in the metabolic reprogramming of TECs and its effects on inflammation and kidney injury in S-AKI. Material and Methods The C57BL/6 mouse model of S-AKI was established using cecal ligation and puncture (CLP). We assessed the inflammatory responses, glucose metabolism and CITED2 expression in the kidneys of septic mice. Additionally, the effect of CITED2 on TECs metabolism and inflammation was evaluated using in vivo and in vitro models. CITED2 silencing and overexpression were employed to elucidate its regulatory role, focusing on the AKT signaling pathway. Results S-AKI causes structural and functional kidney damage, aggravated inflammatory responses, and dysregulated glucose metabolism, accompanied by increased expression of CITED2. CITED2 silencing attenuated TECs metabolic dysfunction and reduced inflammation, thereby protecting the kidney from injury. Conversely, CITED2 overexpression exacerbated TECs metabolic dysfunction, promoted inflammatory responses, and worsened kidney injury. Mechanistically, CITED2 regulates TEC metabolism through the AKT signaling pathway, promoting S-AKI-related inflammation and contributing to kidney injury. Conclusion CITED2 drives the metabolic reprogramming of TECs through the AKT signaling pathway, thereby aggravating the inflammatory response and leading to kidney injury, highlighting its critical role in S-AKI. Targeting CITED2 inhibition may represent a novel therapeutic approach for managing S-AKI.
Collapse
Affiliation(s)
- Ruiming Deng
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, 350004, People’s Republic of China
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Hang Yang
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, 350004, People’s Republic of China
| | - Weibo Zhong
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Juan Zhou
- Department of Thyroid and Breast Surgery, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Guiming Huang
- Department of Anesthesiology, Ganzhou People’s Hospital, Ganzhou City, Jiangxi Province, 341000, People’s Republic of China
| | - Kai Zeng
- Department of Anesthesiology, The First Affiliated Hospital of Fujian Medical University, Fuzhou City, Fujian Province, 350004, People’s Republic of China
| |
Collapse
|
12
|
Zhang C, Gerzanich V, Cruz-Cosme R, Zhang J, Tsymbalyuk O, Tosun C, Sallapalli BT, Liu D, Keledjian K, Papadimitriou JC, Drachenberg CB, Nasr M, Zhang Y, Tang Q, Simard JM, Zhao RY. SARS-CoV-2 ORF3a induces COVID-19-associated kidney injury through HMGB1-mediated cytokine production. mBio 2024; 15:e0230824. [PMID: 39345136 PMCID: PMC11559048 DOI: 10.1128/mbio.02308-24] [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: 07/31/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024] Open
Abstract
The primary challenge posed by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is COVID-19-related mortality, often exacerbated by additional medical complications, such as COVID-19-associated kidney injuries (CAKIs). Up to half of COVID-19 patients experience kidney complications, with those facing acute respiratory failure and kidney injury having the worst overall prognosis. Despite the significant impact of CAKI on COVID-19-related mortality and its enduring effects in long COVID, the underlying causes and molecular mechanisms of CAKI remain elusive. In this study, we identified a functional relationship between the expression of the SARS-CoV-2 ORF3a protein and inflammation-driven apoptotic death of renal tubular epithelial cells in patients with CAKI. We demonstrate in vitro that ORF3a independently induces renal cell-specific apoptotic cell death, as evidenced by the elevation of kidney injury molecule-1 (KIM-1) and the activation of NF-kB-mediated proinflammatory cytokine (TNFα and IL-6) production. By examining kidney tissues of SARS-CoV-2-infected K18-ACE2 transgenic mice, we observed a similar correlation between ORF3a-induced cytopathic changes and kidney injury. This correlation was further validated through reconstitution of the ORF3a effects via direct adenoviral injection into mouse kidneys. Through medicinal analysis, we identified a natural compound, glycyrrhizin (GL4419), which not only blocks viral replication in renal cells, but also mitigates ORF3a-induced renal cell death by inhibiting activation of a high mobility group box 1 (HMGB1) protein, leading to a reduction of KIM-1. Moreover, ORF3a interacts with HMGB1. Overproduction or downregulation of hmgb1 expression results in correlative changes in renal cellular KIM-1 response and respective cytokine production, implicating a crucial role of HMGB1 in ORF3a-inflicted kidney injuries. Our data suggest a direct functional link between ORF3a and kidney injury, highlighting ORF3a as a unique therapeutic target contributing to CAKI. IMPORTANCE The major challenge of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during the pandemic is COVID-19-related mortality, which has tragically claimed millions of lives. COVID-19-associated morbidity and mortality are often exacerbated by pre-existing medical conditions, such as chronic kidney diseases (CKDs), or the development of acute kidney injury (AKI) due to COVID-19, collectively known as COVID-19-associated kidney injuries (CAKIs). Patients who experience acute respiratory failure with CAKI have the poorest clinical outcomes, including increased mortality. Despite these alarming clinical findings, there is a critical gap in our understanding of the underlying causes of CAKI. Our study establishes a direct correlation between the expression of the SARS-CoV-2 viral ORF3a protein and kidney injury induced by ORF3a linking to CAKI. This functional relationship was initially observed in our clinical studies of COVID-19 patients with AKI and was further validated through animal and in vitro cellular studies, either by expressing ORF3a alone or in the context of viral infection. By elucidating this functional relationship and its underlying mechanistic pathways, our research deepens the understanding of COVID-19-associated kidney diseases and presents potential therapeutic avenues to address the healthcare challenges faced by individuals with underlying conditions.
Collapse
Affiliation(s)
- Chenyu Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ruth Cruz-Cosme
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Jiantao Zhang
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Orest Tsymbalyuk
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cigdem Tosun
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Dongxiao Liu
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - Kaspar Keledjian
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - John C. Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cinthia B. Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mohamed Nasr
- Division of AIDS, NIAID, NIH, Drug Development and Clinical Sciences Branch, Bethesda, Maryland, USA
| | - Yanjin Zhang
- Department of Veterinary Medicine, University of Maryland, College Park, Maryland, USA
| | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC, USA
| | - J. Marc Simard
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
| | - Richard Y. Zhao
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Research and Development Service, VA Maryland Health Care System, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA
- Institute of Global Health, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Chen H, Song J, Zeng L, Zha J, Zhu J, Chen A, Liu Y, Dong Z, Chen G. Dietary sodium modulates mTORC1-dependent trained immunity in macrophages to accelerate CKD development. Biochem Pharmacol 2024; 229:116505. [PMID: 39181336 DOI: 10.1016/j.bcp.2024.116505] [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: 05/24/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
Chronic Kidney Disease (CKD) is a significant global health issue linked to dietary habits, especially high salt intake. However, the precise mechanisms driving this progression remain incompletely understood. This study reveals that a high-salt diet intensifies macrophage trained immunity, leading to a marked pro-inflammatory response upon repeated pathogenic exposures, as evidenced by increased renal damage and fibrosis. Under high-salt conditions, there was an induction of CD45+F4/80+ macrophage infiltration into the renal tissue, accompanied by heightened production of inflammatory cytokines. Distinct responses were observed between circulating and resident renal macrophages to a high-salt diet, with a notable upsurge in the migration of pro-inflammatory macrophages, driven by CCL2-CCR2 signaling and aberrant mTORC1 pathway activation. Treatment with rapamycin-liposome effectively reduced this inflammatory cascade by mitigating mTORC1 signaling. Transplantation of monocytes from CKD mice with a high-salt diet significantly exacerbates renal inflammatory damage in the host mice, showing increased migratory tendency and inflammatory activity. The cell co-culture experiment further confirmed that macrophages derived from CKD mice, particularly those under conditions of high salt exposure, significantly induced apoptosis and inflammatory responses in renal tubular cells. Taken together, recurrent exposure to LPS elicits the activation of trained immunity, consequently augmenting inflammatory response of monocytes/macrophages in the involved kidneys. The high-salt diet exacerbates this phenomenon, attributable at least in part to the overactivation of the mTORC1 pathway. This research emphasizes the importance of dietary modulation and targeted immunological interventions in slowing CKD progression, providing new insights into mTORC1-mediated pathophysiological mechanisms and potential management strategies for CKD.
Collapse
Affiliation(s)
- Huihui Chen
- Department of Ophthalmology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Song
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Li Zeng
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jie Zha
- Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Jiefu Zhu
- Department of Transplantation, Renmin Hospital of Wuhan University, Wuhan, China
| | - Anqun Chen
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Yu Liu
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, GA, USA
| | - Guochun Chen
- Department of Nephrology, the Second Xiangya Hospital of Central South University, Changsha, China; Hunan Key Laboratory of Kidney Disease and Blood Purification, the Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
14
|
Seibt T, Wahida A, Hoeft K, Kemmner S, Linkermann A, Mishima E, Conrad M. The biology of ferroptosis in kidney disease. Nephrol Dial Transplant 2024; 39:1754-1761. [PMID: 38684468 DOI: 10.1093/ndt/gfae097] [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: 01/26/2024] [Indexed: 05/02/2024] Open
Abstract
Ferroptosis is a regulated cell death modality triggered by iron-dependent lipid peroxidation. Ferroptosis plays a causal role in the pathophysiology of various diseases, making it a promising therapeutic target. Unlike all other cell death modalities dependent on distinct signaling cues, ferroptosis occurs when cellular antioxidative defense mechanisms fail to suppress the oxidative destruction of cellular membranes, eventually leading to cell membrane rupture. Physiologically, only two such surveillance systems are known to efficiently prevent the lipid peroxidation chain reaction by reducing (phospho)lipid hydroperoxides to their corresponding alcohols or by reducing radicals in phospholipid bilayers, thus maintaining the integrity of lipid membranes. Mechanistically, these two systems are linked to the reducing capacity of glutathione peroxidase 4 (GPX4) by consuming glutathione (GSH) on one hand and ferroptosis suppressor protein 1 (FSP1, formerly AIFM2) on the other. Notably, the importance of ferroptosis suppression in physiological contexts has been linked to a particular vulnerability of renal tissue. In fact, early work has shown that mice genetically lacking Gpx4 rapidly succumb to acute renal failure with pathohistological features of acute tubular necrosis. Promising research attempting to implicate ferroptosis in various renal disease entities, particularly those with proximal tubular involvement, has generated a wealth of knowledge with widespread potential for clinical translation. Here, we provide a brief overview of the involvement of ferroptosis in nephrology. Our goal is to introduce this expanding field for clinically versed nephrologists in the hope of spurring future efforts to prevent ferroptosis in the pathophysiological processes of the kidney.
Collapse
Affiliation(s)
- Tobias Seibt
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
- Transplant Center, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Adam Wahida
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| | - Konrad Hoeft
- Division of Nephrology and Clinical Immunology, RWTH Aachen University, Aachen, Germany
| | - Stephan Kemmner
- Transplant Center, University Hospital Munich, Ludwig-Maximilians-University (LMU), Munich, Germany
| | - Andreas Linkermann
- Division of Nephrology, Clinic of Internal Medicine 3, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany
- Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
- Division of Nephrology, Rheumatology and Endocrinology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany
| |
Collapse
|
15
|
Yang Z, Cancio TS, Willis RP, Young MD, Kneifel DM, Salinas J, Meyer AD. An early HMGB1 rise 12 hours before creatinine predicts acute kidney injury and multiple organ failure in a smoke inhalation and burn swine model. Front Immunol 2024; 15:1447597. [PMID: 39534595 PMCID: PMC11554498 DOI: 10.3389/fimmu.2024.1447597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/26/2024] [Indexed: 11/16/2024] Open
Abstract
Background Acute kidney injury (AKI) and multiple organ failure (MOF) are leading causes of mortality in trauma injuries. Early diagnosis of AKI and MOF is vital to improve outcomes, but current diagnostic criteria rely on laboratory markers that are delayed or unreliable. In this study, we investigated whether damage associated molecular patterns such as high-mobility group box 1 (HMGB1), syndecan-1 (SDC-1) and C3a correlate with the development of trauma-induced AKI and MOF. Methods Thirty-nine swine underwent smoke inhalation and severe burns, then received critical care for 72 hours or until death. AKI was defined by the KDIGO (Kidney Disease: Improving Global Outcomes) criteria, which labels AKI when a 1.5-fold increase in blood creatinine levels from baseline or a urine output < 0.5 mL/kg/h for 6 hours or more occurs. MOF was defined by the presence of both AKI and acute respiratory distress syndrome (PaO2/FiO2<300 for 4 hours). Results Eight of 39 pigs developed AKI and seven of those developed MOF. Pathological analysis revealed that polytrauma induces significantly higher kidney injury scores compared to sham controls. The average time from injury to KDIGO AKI was 24 hours (interquartile range: 22.50-32.25). Twelve hours after injury, HMGB1 levels were significantly increased in animals that went on to develop AKI compared to those that did not (73.07 ± 18.66 ng/mL vs. 31.64 ± 4.15 ng/mL, p<0.01), as well as in animals that developed MOF compared to those that did not (81.52±19.68 ng/mL vs. 31.19 ± 3.972 ng/mL, p<0.05). SDC-1 and C3a levels were not significantly different at any time point between groups. ROC analysis revealed that HMGB1 levels at 12 hours post-injury were predictive of both AKI and MOF development (AKI: AUROC=0.81, cut-off value=36.41 ng/mL; MOF: AUROC=0.89, cut-off value=36.41 ng/mL). Spearman's correlation revealed that HMGB1 levels at 12 hours correlated with multiple parameters of AKI, including blood urea nitrogen, blood creatinine, and blood myoglobin. Conclusion Twelve-hour post-injury HMGB1 levels predict AKI and MOF in a smoke inhalation and burn swine model. Further research is needed to validate this result in other polytrauma models and in critical combat causalities.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Tomas S. Cancio
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Robert P. Willis
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Matthew D. Young
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Dustin M. Kneifel
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Jose Salinas
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Andrew D. Meyer
- Organ Support and Automation Technologies, United States Army Institute of Surgical Research, Fort Sam Houston, TX, United States
- Long School of Medicine, University of Texas Health Science Center, San Antonio, TX, United States
| |
Collapse
|
16
|
Han J, Ye L, Wang Y. Pyroptosis: An Accomplice in the Induction of Multisystem Complications Triggered by Obstructive Sleep Apnea. Biomolecules 2024; 14:1349. [PMID: 39595526 PMCID: PMC11592050 DOI: 10.3390/biom14111349] [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: 07/25/2024] [Revised: 10/05/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
Obstructive sleep apnea (OSA) is a common respiratory disorder, primarily characterized by two pathological features: chronic intermittent hypoxia (CIH) and sleep deprivation (SD). OSA has been identified as a risk factor for numerous diseases, and the inflammatory response related to programmed cell necrosis is believed to play a significant role in the occurrence and progression of multisystem damage induced by OSA, with increasing attention being paid to pyroptosis. Recent studies have indicated that OSA can elevate oxidative stress levels in the body, activating the process of pyroptosis within different tissues, ultimately accelerating organ dysfunction. However, the molecular mechanisms of pyroptosis in the multisystem damage induced by OSA remain unclear. Therefore, this review focuses on four major systems that have received concentrated attention in existing research in order to explore the role of pyroptosis in promoting renal diseases, cardiovascular diseases, neurocognitive diseases, and skin diseases in OSA patients. Furthermore, we provide a comprehensive overview of methods for inhibiting pyroptosis at different molecular levels, with the goal of identifying viable targets and therapeutic strategies for addressing OSA-related complications.
Collapse
Affiliation(s)
- Jingwen Han
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China; (J.H.); (L.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Lisong Ye
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China; (J.H.); (L.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
| | - Yan Wang
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200001, China
- Department of Preventive Dentistry, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai 200001, China
| |
Collapse
|
17
|
Zhu J, Gong Z, Wang X, Zhang K, Ma Y, Zou H, Song R, Zhao H, Liu Z, Dong W. mTORC1 and mTORC2 Co-Protect against Cadmium-Induced Renal Tubular Epithelial Cell Apoptosis and Acute Kidney Injury by Regulating Protein Kinase B. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19667-19679. [PMID: 39219293 PMCID: PMC11404484 DOI: 10.1021/acs.jafc.4c05702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
The potential threat of cadmium (Cd)-induced acute kidney injury (AKI) is increasing. In this study, our primary goal was to investigate the individual roles played by mTOR complexes, specifically mTORC1 and mTORC2, in Cd-induced apoptosis in mouse kidney cells. We constructed a mouse model with specific deletion of Raptor/Rictor renal cells. Inhibitors and activators of mTORC1 or mTORC2 were also applied. The effects of protein kinase B (AKT) activation and autophagy were studied. Both mTORC1 and mTORC2 were found to mediate the antiapoptotic mechanism of renal cells by regulating the AKT activity. Inhibition of mTORC1 or mTORC2 exacerbated Cd-induced kidney cell apoptosis, suggesting that both proteins exert antiapoptotic effects under Cd exposure. We further found that the AKT activation plays a key role in mTORC1/TORC2-mediated antiapoptosis, protecting Cd-exposed kidney cells from apoptosis. We also found that mTOR activators inhibited excessive autophagy, alleviated apoptosis, and promoted cell survival. These findings provide new insights into the regulatory mechanisms of mTOR in renal diseases and provide a theoretical basis for the development of novel therapeutic strategies to treat renal injury.
Collapse
Affiliation(s)
- Jiaqiao Zhu
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Zhonggui Gong
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Agricultural
High-tech Industrial Demonstration Area of the Yellow River Delta
of Shandong Province, Dongying, Shandong 257000, China
- National
Technological Innovation Center for Comprehensive Utilization of Saline-Alkali
Land, Dongying, Shandong 257000, China
| | - Xueru Wang
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Kanglei Zhang
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Yonggang Ma
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Hui Zou
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Ruilong Song
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Hongyan Zhao
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Zongping Liu
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Institute
of Agricultural Science and Technology Development (Joint International
Research Laboratory of Agriculture and Agri-Product Safety of the
Ministry of Education of China), Yangzhou
University, Yangzhou, Jiangsu 225009, China
- Jiangsu
Co-Innovation Center for Prevention and Control of Important Animal
Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225009, China
| | - Wenxuan Dong
- College
of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu 225009, China
- Laboratory
of Animal Nutrition Metabolic and Poisoning Diseases, College of Veterinary
Medicine, Qingdao Agricultural University, Qingdao, Shandong 266109, China
| |
Collapse
|
18
|
Li ZL, Li XY, Zhou Y, Wang B, Lv LL, Liu BC. Renal tubular epithelial cells response to injury in acute kidney injury. EBioMedicine 2024; 107:105294. [PMID: 39178744 PMCID: PMC11388183 DOI: 10.1016/j.ebiom.2024.105294] [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: 07/19/2024] [Accepted: 08/06/2024] [Indexed: 08/26/2024] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid and significant decrease in renal function that can arise from various etiologies, and is associated with high morbidity and mortality. The renal tubular epithelial cells (TECs) represent the central cell type affected by AKI, and their notable regenerative capacity is critical for the recovery of renal function in afflicted patients. The adaptive repair process initiated by surviving TECs following mild AKI facilitates full renal recovery. Conversely, when injury is severe or persistent, it allows the TECs to undergo pathological responses, abnormal adaptive repair and phenotypic transformation, which will lead to the development of renal fibrosis. Given the implications of TECs fate after injury in renal outcomes, a deeper understanding of these mechanisms is necessary to identify promising therapeutic targets and biomarkers of the repair process in the human kidney.
Collapse
Affiliation(s)
- Zuo-Lin Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Xin-Yan Li
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Yan Zhou
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Bin Wang
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospital, Southeast University School of Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
19
|
Mo C, Huang Q, Li L, Long Y, Shi Y, Lu Z, Wu N, Li Q, Zeng H, Li G, Qiu L, Gui C, Ji Q. High-mobility group box 1 and its related receptors: potential therapeutic targets for contrast-induced acute kidney injury. Int Urol Nephrol 2024; 56:2291-2299. [PMID: 38438703 DOI: 10.1007/s11255-024-03981-2] [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: 12/04/2023] [Accepted: 02/13/2024] [Indexed: 03/06/2024]
Abstract
Percutaneous coronary intervention (PCI) is a crucial diagnostic and therapeutic approach for coronary heart disease. Contrast agents' exposure during PCI is associated with a risk of contrast-induced acute kidney injury (CI-AKI). CI-AKI is characterized by a sudden decline in renal function occurring as a result of exposure to intravascular contrast agents, which is associated with an increased risk of poor prognosis. The pathophysiological mechanisms underlying CI-AKI involve renal medullary hypoxia, direct cytotoxic effects, endoplasmic reticulum stress, inflammation, oxidative stress, and apoptosis. To date, there is no effective therapy for CI-AKI. High-mobility group box 1 (HMGB1), as a damage-associated molecular pattern molecule, is released extracellularly by damaged cells or activated immune cells and binds to related receptors, including toll-like receptors and receptor for advanced glycation end product. In renal injury, HMGB1 is expressed in renal tubular epithelial cells, macrophages, endothelial cells, and glomerular cells, involved in the pathogenesis of various kidney diseases by activating its receptors. Therefore, this review provides a theoretical basis for HMGB1 as a therapeutic intervention target for CI-AKI.
Collapse
Affiliation(s)
- Changhua Mo
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qili Huang
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Lixia Li
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yusheng Long
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Ying Shi
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Zhengde Lu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Ning Wu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qingkuan Li
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Huayuan Zeng
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Guihua Li
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Lingyue Qiu
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University and Guangxi Key Laboratory Base of Precision Medicine in Cardiocerebrovascular Diseases Control and Prevention and Guangxi Clinical Research Center for Cardiocerebrovascular Diseases, Nanning, China.
| | - Qingwei Ji
- Department of Cardiology, The People's Hospital of Guangxi Zhuang Autonomous Region and Research Center of Cardiovascular Disease, Guangxi Academy of Medical Sciences, Nanning, China.
| |
Collapse
|
20
|
Berezin AE, Berezina TA, Hoppe UC, Lichtenauer M, Berezin AA. An overview of circulating and urinary biomarkers capable of predicting the transition of acute kidney injury to chronic kidney disease. Expert Rev Mol Diagn 2024; 24:627-647. [PMID: 39007888 DOI: 10.1080/14737159.2024.2379355] [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: 04/15/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
INTRODUCTION Acute kidney injury (AKI) defined by a substantial decrease in kidney function within hours to days and is often irreversible with higher risk to chronic kidney disease (CKD) transition. AREAS COVERED The authors discuss the diagnostic and predictive utilities of serum and urinary biomarkers on AKI and on the risk of AKI-to-CKD progression. The authors focus on the relevant literature covering evidence of circulating and urinary biomarkers' capability to predict the transition of AKI to CKD. EXPERT OPINION Based on the different modalities of serum and urinary biomarkers, multiple biomarker panel seems to be potentially useful to distinguish between various types of AKI, to detect the severity and the risk of AKI progression, to predict the clinical outcome and evaluate response to the therapy. Serum/urinary neutrophil gelatinase-associated lipocalin (NGAL), serum/urinary uromodulin, serum extracellular high mobility group box-1 (HMGB-1), serum cystatin C and urinary liver-type fatty acid-binding protein (L-FABP) were the most effective in the prediction of AKI-to-CKD transition regardless of etiology and the presence of critical state in patients. The current clinical evidence on the risk assessments of AKI progression is mainly based on the utility of combination of functional, injury and stress biomarkers, mainly NGAL, L-FABP, HMGB-1 and cystatin C.
Collapse
Affiliation(s)
- Alexander E Berezin
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Tetiana A Berezina
- Department of Internal Medicine & Nephrology, VitaCenter, Zaporozhye, Ukraine
| | - Uta C Hoppe
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | - Michael Lichtenauer
- Department of Internal Medicine II, Division of Cardiology, Paracelsus Medical University, Salzburg, Austria
| | | |
Collapse
|
21
|
Chen L, Hu J, Lu J, Gong X. Bibliometric and visual analysis of immunisation associated with acute kidney injury from 2003 to 2023. Front Pharmacol 2024; 15:1388527. [PMID: 39011500 PMCID: PMC11246997 DOI: 10.3389/fphar.2024.1388527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/06/2024] [Indexed: 07/17/2024] Open
Abstract
Objective This study aims to conduct a detailed bibliometric and visual analysis of acute kidney injury (AKI) and immune-related research conducted over the past two decades, focusing on identifying emerging trends and key areas of interest. Methods The Web of Science Core Collection (WoSCC) was utilised for the meticulous examination of various parameters including publication volume, authorship, geographic distribution, institutional contributions, journal sources, prevalent keywords and citation frequencies. Data were intricately visualised and interpreted using VOSviewer, CiteSpace and Excel 365 software. Results Analysis of the WoSCC database revealed 3,537 articles on AKI and immunisation, originating from 94 countries and regions, involving 3,552 institutions and authored by 18,243 individuals. Notably, the top five countries contributing to this field were the United States, China, Germany, Italy and the United Kingdom, with the United States leading with 35.76% of total publications. Among the 3,552 contributing institutions, those in the United States were predominant, with Harvard University leading with 134 papers and 3,906 citations. Key journals driving productivity included Frontiers in Immunology, Kidney International, Journal of the American Society of Nephrology and International Journal of Molecular Sciences, with Kidney International being the most cited, followed by Journal of the American Society of Nephrology and New England Journal of Medicine. Prominent authors in the field included Ronco Claudio, Okusa Mark D and Anders, Hans-Joachim. Co-citation clustering and timeline analysis highlighted recent research foci such as COVID-19, immune checkpoint inhibitors, regulated necrosis, cirrhosis and AKI. Keyword analysis identified "inflammation," "ischaemia-reperfusion injury," "sepsis," "covid-19," and "oxidative stress" as prevalent terms. Conclusion This study provides the first bibliometric analysis of AKI and immune research, offering a comprehensive overview of research hotspots and evolving trends within the field.
Collapse
Affiliation(s)
- Ling Chen
- Department of Nephrology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Hu
- Department of Nephrology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianrao Lu
- Department of Nephrology, Seventh People’s Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xuezhong Gong
- Department of Nephrology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
22
|
Tutunea-Fatan E, Arumugarajah S, Suri RS, Edgar CR, Hon I, Dikeakos JD, Gunaratnam L. Sensing Dying Cells in Health and Disease: The Importance of Kidney Injury Molecule-1. J Am Soc Nephrol 2024; 35:795-808. [PMID: 38353655 PMCID: PMC11164124 DOI: 10.1681/asn.0000000000000334] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Kidney injury molecule-1 (KIM-1), also known as T-cell Ig and mucin domain-1 (TIM-1), is a widely recognized biomarker for AKI, but its biological function is less appreciated. KIM-1/TIM-1 belongs to the T-cell Ig and mucin domain family of conserved transmembrane proteins, which bear the characteristic six-cysteine Ig-like variable domain. The latter enables binding of KIM-1/TIM-1 to its natural ligand, phosphatidylserine, expressed on the surface of apoptotic cells and necrotic cells. KIM-1/TIM-1 is expressed in a variety of tissues and plays fundamental roles in regulating sterile inflammation and adaptive immune responses. In the kidney, KIM-1 is upregulated on injured renal proximal tubule cells, which transforms them into phagocytes for clearance of dying cells and helps to dampen sterile inflammation. TIM-1, expressed in T cells, B cells, and natural killer T cells, is essential for cell activation and immune regulatory functions in the host. Functional polymorphisms in the gene for KIM-1/TIM-1, HAVCR1 , have been associated with susceptibility to immunoinflammatory conditions and hepatitis A virus-induced liver failure, which is thought to be due to a differential ability of KIM-1/TIM-1 variants to bind phosphatidylserine. This review will summarize the role of KIM-1/TIM-1 in health and disease and its potential clinical applications as a biomarker and therapeutic target in humans.
Collapse
Affiliation(s)
- Elena Tutunea-Fatan
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
| | - Shabitha Arumugarajah
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rita S. Suri
- Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Division of Nephrology, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Cassandra R. Edgar
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Ingrid Hon
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Jimmy D. Dikeakos
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London Health Sciences Centre, London, Ontario, Canada
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
23
|
Li T, Yang K, Tong Y, Guo S, Gao W, Zou X. Targeted Drug Therapy for Senescent Cells Alleviates Unilateral Ureteral Obstruction-Induced Renal Injury in Rats. Pharmaceutics 2024; 16:695. [PMID: 38931822 PMCID: PMC11206309 DOI: 10.3390/pharmaceutics16060695] [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: 04/18/2024] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024] Open
Abstract
Hydronephrosis resulting from unilateral ureteral obstruction (UUO) is a common cause of renal injury, often progressing to late-stage renal fibrosis or even potential renal failure. Renal injury and repair processes are accompanied by changes in cellular senescence phenotypes. However, the mechanism is poorly understood. The purpose of this study is to clarify the changes in senescence phenotype at different time points in renal disease caused by UUO and to further investigate whether eliminating senescent cells using the anti-senescence drug ABT263 could attenuate UUO-induced renal disease. Specifically, renal tissues were collected from established UUO rat models on days 1, 2, 7, and 14. The extent of renal tissue injury and fibrosis in rats was assessed using histological examination, serum creatinine, and blood urea nitrogen levels. The apoptotic and proliferative capacities of renal tissues and phenotypic changes in cellular senescence were evaluated. After the intervention of the anti-senescence drug ABT263, the cellular senescence as well as tissue damage changes were re-assessed. We found that before the drug intervention, the UUO rats showed significantly declined renal function, accompanied by renal tubular injury, increased inflammatory response, and oxidative stress, alongside aggravated cellular senescence. Meanwhile, after the treatment with ABT263, the rats had a significantly lower number of senescent cells, attenuated renal tubular injury and apoptosis, enhanced proliferation, reduced oxidative stress and inflammation, improved renal function, and markedly inhibited fibrosis. This suggests that the use of the anti-senescence drug ABT263 to eliminate senescent cells can effectively attenuate UUO-induced renal injury. This highlights the critical role of cellular senescence in the transformation of acute injury into chronic fibrosis.
Collapse
Affiliation(s)
| | | | | | | | - Wei Gao
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China; (T.L.); (K.Y.); (Y.T.); (S.G.)
| | - Xiangyu Zou
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang 261053, China; (T.L.); (K.Y.); (Y.T.); (S.G.)
| |
Collapse
|
24
|
Douvris A, Viñas J, Gutsol A, Zimpelmann J, Burger D, Burns K. miR-486-5p protects against rat ischemic kidney injury and prevents the transition to chronic kidney disease and vascular dysfunction. Clin Sci (Lond) 2024; 138:599-614. [PMID: 38739452 PMCID: PMC11130553 DOI: 10.1042/cs20231752] [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: 12/27/2023] [Revised: 03/12/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
AIM Acute kidney injury (AKI) increases the risk for progressive chronic kidney disease (CKD). MicroRNA (miR)-486-5p protects against kidney ischemia-reperfusion (IR) injury in mice, although its long-term effects on the vasculature and development of CKD are unknown. We studied whether miR-486-5p would prevent the AKI to CKD transition in rat, and affect vascular function. METHODS Adult male rats were subjected to bilateral kidney IR followed by i.v. injection of liposomal-packaged miR-486-5p (0.5 mg/kg). Kidney function and histologic injury were assessed after 24 h and 10 weeks. Kidney endothelial protein levels were measured by immunoblot and immunofluorescence, and mesenteric artery reactivity was determined by wire myography. RESULTS In rats with IR, miR-486-5p blocked kidney endothelial cell increases in intercellular adhesion molecule-1 (ICAM-1), reduced neutrophil infiltration and histologic injury, and normalized plasma creatinine (P<0.001). However, miR-486-5p attenuated IR-induced kidney endothelial nitric oxide synthase (eNOS) expression (P<0.05). At 10 weeks, kidneys from rats with IR alone had decreased peritubular capillary density and increased interstitial collagen deposition (P<0.0001), and mesenteric arteries showed impaired endothelium-dependent vasorelaxation (P<0.001). These changes were inhibited by miR-486-5p. Delayed miR-486-5p administration (96 h, 3 weeks after IR) had no impact on kidney fibrosis, capillary density, or endothelial function. CONCLUSION In rats, administration of miR-486-5p early after kidney IR prevents injury, and protects against CKD development and systemic endothelial dysfunction. These protective effects are associated with inhibition of endothelial ICAM-1 and occur despite reduction in eNOS. miR-486-5p holds promise for the prevention of ischemic AKI and its complications.
Collapse
Affiliation(s)
- Adrianna Douvris
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Jose L. Viñas
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Alexey Gutsol
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Joseph Zimpelmann
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
| | - Dylan Burger
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Kevin D. Burns
- Division of Nephrology, Department of Medicine and Kidney Research Centre, Ottawa Hospital Research Institute, University of Ottawa and the Ottawa Hospital, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| |
Collapse
|
25
|
Liu T, Zhao H, Wang Y, Qu P, Wang Y, Wu X, Zhao T, Yang L, Mao H, Peng L, Zhan Y, Li P. Serum high mobility group box 1 as a potential biomarker for the progression of kidney disease in patients with type 2 diabetes. Front Immunol 2024; 15:1334109. [PMID: 38481996 PMCID: PMC10932975 DOI: 10.3389/fimmu.2024.1334109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/15/2024] [Indexed: 04/10/2024] Open
Abstract
Background As a damage-associated molecular pattern protein, high mobility group box 1 (HMGB1) is associated with kidney and systemic inflammation. The predictive and therapeutic value of HMGB1 as a biomarker has been confirmed in various diseases. However, its value in diabetic kidney disease (DKD) remains unclear. Therefore, this study aimed to investigate the correlation between serum and urine HMGB1 levels and DKD progression. Methods We recruited 196 patients with type 2 diabetes mellitus (T2DM), including 109 with DKD and 87 T2DM patients without DKD. Additionally, 60 healthy participants without T2DM were also recruited as controls. Serum and urine samples were collected for HMGB1 analysis. Simultaneously, tumor necrosis factor receptor superfamily member 1A (TNFR-1) in serum and kidney injury molecule (KIM-1) in urine samples were evaluated for comparison. Results Serum and urine HMGB1 levels were significantly higher in patients with DKD than in patients with T2DM and healthy controls. Additionally, serum HMGB1 levels significantly and positively correlated with serum TNFR-1 (R 2 = 0.567, p<0.001) and urine KIM-1 levels (R 2 = 0.440, p<0.001), and urine HMGB1 has a similar correlation. In the population with T2DM, the risk of DKD progression increased with an increase in serum HMGB1 levels. Multivariate logistic regression analysis showed that elevated serum HMGB1 level was an independent risk factor for renal function progression in patients with DKD, and regression analysis did not change in the model corrected for multiple variables. The restricted cubic spline depicted a nonlinear relationship between serum HMGB1 and renal function progression in patients with DKD (p-nonlinear=0.007, p<0.001), and this positive effect remained consistent across subgroups. Conclusion Serum HMGB1 was significantly correlated with DKD and disease severity. When the HMGB1 level was ≥27 ng/ml, the risk of renal progression increased sharply, indicating that serum HMGB1 can be used as a potential biomarker for the diagnosis of DKD progression.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hailing Zhao
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Ying Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Peng Qu
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yanmei Wang
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Xiai Wu
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Tingting Zhao
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| |
Collapse
|
26
|
Ya-Fen Z, Jing C, Yue-Fei Z, Chang-Ping D. Reduction in NGAL at 48 h predicts the progression to CKD in patients with septic associated AKI: a single-center clinical study. Int Urol Nephrol 2024; 56:607-613. [PMID: 37382770 DOI: 10.1007/s11255-023-03689-9] [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: 05/08/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
BACKGROUND In this study, our objective was to investigate the predictive value of serum and urine fluctuations of neutrophil gelatinase-associated lipid transporters (NGAL) in relation to the progression of chronic kidney disease (CKD) among patients with septic associated AKI (SA-AKI). METHODS A total of 425 SA-AKI patients were enrolled in this study and divided into the recovery group (n = 320) and the AKI-to-CKD group (n = 105) based on 3-month follow-up data. The serum and urine NGAL levels on the day of AKI diagnosis (T0) and 48 h after anti-AKI treatment (T1) were recorded and calculated. RESULTS The levels of NGAL in serum and urine were found to be higher in the AKI-to-CKD group compared to the recovery group at T1 point (P < 0.05). The reductions of NGAL at 48 h in serum and urine were lower in the AKI-to-CKD group than those observed in the recovery group (P < 0.05). In comparison to T0, a significant decrease was noted for both serum and urine NGAL levels on T1 among patients who recovered from AKI (P < 0.05), whereas no such trend was observed among those with AKI-to-CKD transition (P > 0.05). After adjusting age, sex, and BMI through partial correlation analysis, the reduction of serum NGAL was found to be most strongly associated with the transition from AKI to CKD. ROC analysis showed an AUC of 0.832 for serum NGAL reduction, with a cut-off value of - 111.24 ng/ml and sensitivity and rates of 76.2% and 81.2%, respectively. Logistic regression analysis indicated that a reduction of serum NGAL ≥ - 111.24 ng/ml was the early warning indicator for the progression of CKD in SA-AKI patients. CONCLUSION The reduction of serum NGAL following 48 h of anti-AKI therapy represents a distinct hazard factor for the advancement of CKD in patients with SA-AKI, independent of other variables.
Collapse
Affiliation(s)
- Zhang Ya-Fen
- Department of Medical Laboratory, Yangzhou University Affiliated Hospital, Yangzhou, Jiangsu, China
| | - Chen Jing
- Department of Medical Laboratory, Yangzhou University Affiliated Hospital, Yangzhou, Jiangsu, China
| | - Zhang Yue-Fei
- Department of Emergence, Yangzhou University Affiliated Hospital, Yangzhou, Jiangsu, China
| | - Ding Chang-Ping
- Department of Medical Laboratory, Yangzhou University Affiliated Hospital, Yangzhou, Jiangsu, China.
| |
Collapse
|
27
|
Mohamed R, Sullivan JC. Sustained activation of 12/15 lipoxygenase (12/15 LOX) contributes to impaired renal recovery post ischemic injury in male SHR compared to females. Mol Med 2023; 29:163. [PMID: 38049738 PMCID: PMC10696802 DOI: 10.1186/s10020-023-00762-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 11/19/2023] [Indexed: 12/06/2023] Open
Abstract
BACKGROUND Acute kidney injury (AKI) due to ischemia-reperfusion (IR) is a serious and frequent complication in clinical settings, and mortality rates remain high. There are well established sex differences in renal IR, with males exhibiting greater injury following an ischemic insult compared to females. We recently reported that males have impaired renal recovery from ischemic injury vs. females. However, the mechanisms mediating sex differences in renal recovery from IR injury remain poorly understood. Elevated 12/15 lipoxygenase (LOX) activity has been reported to contribute to the progression of numerous kidney diseases. The goal of the current study was to test the hypothesis that enhanced activation of 12/15 LOX contributes to impaired recovery post-IR in males vs. females. METHODS 13-week-old male and female spontaneously hypertensive rats (SHR) were randomized to sham or 30-minute warm bilateral IR surgery. Additional male and female SHR were randomized to treatment with vehicle or the specific 12/15 LOX inhibitor ML355 1 h prior to sham/IR surgery, and every other day following up to 7-days post-IR. Blood was collected from all rats 1-and 7-days post-IR. Kidneys were harvested 7-days post-IR and processed for biochemical, histological, and Western blot analysis. 12/15 LOX metabolites 12 and 15 HETE were measured in kidney samples by liquid chromatography-mass spectrometry (LC/MS). RESULTS Male SHR exhibited delayed recovery of renal function post-IR vs. male sham and female IR rats. Delayed recovery in males was associated with activation of renal 12/15 LOX, increased renal 12-HETE, enhanced endoplasmic reticulum (ER) stress, lipid peroxidation, renal cell death and inflammation compared to females 7-days post-IR. Treatment of male SHR with ML355 lowered levels of 12-HETE and resulted in reduced renal lipid peroxidation, ER stress, tubular cell death and inflammation 7-days post-IR with enhanced recovery of renal function compared to vehicle-treated IR male rats. ML355 treatment did not alter IR-induced increases in plasma creatinine in females, however, tubular injury and cell death were attenuated in ML355 treated females compared to vehicle-treated rats 7 days post-IR. CONCLUSION Our data demonstrate that sustained activation 12/15 LOX contributes to impaired renal recovery post ischemic injury in male and female SHR, although males are more susceptible on this mechanism than females.
Collapse
Affiliation(s)
- Riyaz Mohamed
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, 30912, United States.
| | - Jennifer C Sullivan
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia, 30912, United States
| |
Collapse
|
28
|
Wang L, Zha H, Huang J, Shi L. Flavin containing monooxygenase 2 regulates renal tubular cell fibrosis and paracrine secretion via SMURF2 in AKI‑CKD transformation. Int J Mol Med 2023; 52:110. [PMID: 37800598 PMCID: PMC10558214 DOI: 10.3892/ijmm.2023.5313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023] Open
Abstract
In the follow‑up of hospitalized patients with acute kidney injury (AKI), it has been observed that 15‑30% of these patients progress to develop chronic kidney disease (CKD). Impaired adaptive repair of the kidneys following AKI is a fundamental pathophysiological mechanism underlying renal fibrosis and the progression to CKD. Deficient repair of proximal tubular epithelial cells is a key factor in the progression from AKI to CKD. However, the molecular mechanisms involved in the regulation of fibrotic factor paracrine secretion by injured tubular cells remain incompletely understood. Transcriptome analysis and an ischemia‑reperfusion injury (IRI) model were used to identify the contribution of flavin‑containing monooxygenase 2 (FMO2) in AKI‑CKD. Lentivirus‑mediated overexpression of FMO2 was performed in mice. Functional experiments were conducted using TGF‑β‑induced tubular cell fibrogenesis and paracrine pro‑fibrotic factor secretion. Expression of FMO2 attenuated kidney injury induced by renal IRI, renal fibrosis, and immune cell infiltration into the kidneys. Overexpression of FMO2 not only effectively blocked TGF secretion in tubular cell fibrogenesis but also inhibited aberrant paracrine activation of pro‑fibrotic factors present in fibroblasts. FMO2 negatively regulated TGF‑β‑mediated SMAD2/3 activation by promoting the expression of SMAD ubiquitination regulatory factor 2 (SMURF2) and its nuclear translocation. During the transition from AKI to CKD, FMO2 modulated tubular cell fibrogenesis and paracrine secretion through SMURF2, thereby affecting the outcome of the disease.
Collapse
Affiliation(s)
- Longfei Wang
- Children's Hospital Affiliated to Zhengzhou University, Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, Henan 450018, P.R. China
| | - Hongchu Zha
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Kidney Disease Research Institute of China Three Gorges University, Yichang, Hubei 443000, P.R. China
| | - Jing Huang
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Lang Shi
- Department of Nephrology, The First Clinical Medical College of Three Gorges University, Center People's Hospital of Yichang, Kidney Disease Research Institute of China Three Gorges University, Yichang, Hubei 443000, P.R. China
- Department of Nephrology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
29
|
Li Z, Qiao O, Wang Y, Li N, Gong Y. Potential therapeutic targets for trauma management. Trends Pharmacol Sci 2023; 44:S0165-6147(23)00234-1. [PMID: 39492319 DOI: 10.1016/j.tips.2023.10.001] [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: 08/20/2023] [Revised: 10/06/2023] [Accepted: 10/06/2023] [Indexed: 11/05/2024]
Abstract
Despite advances in medical treatments for severe trauma, it remains a critical condition associated with high mortality. During trauma, the release of endogenous damage-associated molecular patterns (DAMPs) can induce immune dysfunction, leading to sepsis or multiple organ dysfunction syndrome (MODS). Vaccines based on specific pathogen antigens and pathogen-associated molecular patterns (PAMPs) contribute largely to the prevention of communicable diseases through the induction of adaptive immune responses. Vaccines developed based on autologous molecules may also promote recovery from non-communicable diseases (NCDs) by eliciting appropriate immune responses, as recent clinical trials indicate. Developing new vaccines targeting DAMPs may be an effective pre-protective measure for trauma management. We describe the role of DAMPs in post-traumatic immune dysfunction and discuss the potential of harnessing them for trauma vaccine development as well as the risks and challenges.
Collapse
Affiliation(s)
- Zizheng Li
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Ou Qiao
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China
| | - Yuru Wang
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China; Thinking Biomed (Beijing) Co., Ltd, Beijing Economic and Technological Development Zone, Beijing, 100176, China
| | - Ning Li
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China.
| | - Yanhua Gong
- Institute of Disaster and Emergency Medicine, Medical School, Tianjin University, No. 92 Weijin Road, Nankai District, Tianjin, 300072, China; Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin, 300072, China.
| |
Collapse
|
30
|
Liu T, Li Q, Jin Q, Yang L, Mao H, Qu P, Guo J, Zhang B, Ma F, Wang Y, Peng L, Li P, Zhan Y. Targeting HMGB1: A Potential Therapeutic Strategy for Chronic Kidney Disease. Int J Biol Sci 2023; 19:5020-5035. [PMID: 37781525 PMCID: PMC10539693 DOI: 10.7150/ijbs.87964] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023] Open
Abstract
High-mobility group protein box 1 (HMGB1) is a member of a highly conserved high-mobility group protein present in all cell types. HMGB1 plays multiple roles both inside and outside the cell, depending on its subcellular localization, context, and post-translational modifications. HMGB1 is also associated with the progression of various diseases. Particularly, HMGB1 plays a critical role in CKD progression and prognosis. HMGB1 participates in multiple key events in CKD progression by activating downstream signals, including renal inflammation, the onset of persistent fibrosis, renal aging, AKI-to-CKD transition, and important cardiovascular complications. More importantly, HMGB1 plays a distinct role in the chronic pathophysiology of kidney disease, which differs from that in acute lesions. This review describes the regulatory role of HMGB1 in renal homeostasis and summarizes how HMGB1 affects CKD progression and prognosis. Finally, some promising therapeutic strategies for the targeted inhibition of HMGB1 in improving CKD are summarized. Although the application of HMGB1 as a therapeutic target in CKD faces some challenges, a more in-depth understanding of the intracellular and extracellular regulatory mechanisms of HMGB1 that underly the occurrence and progression of CKD might render HMGB1 an attractive therapeutic target for CKD.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Jin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Peng Qu
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Jing Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bo Zhang
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuyang Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liang Peng
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Ping Li
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
31
|
Ren Y, Zhu D, Han X, Zhang Q, Chen B, Zhou P, Wei Z, Zhang Z, Cao Y, Zou H. HMGB1: a double-edged sword and therapeutic target in the female reproductive system. Front Immunol 2023; 14:1238785. [PMID: 37691930 PMCID: PMC10484633 DOI: 10.3389/fimmu.2023.1238785] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/03/2023] [Indexed: 09/12/2023] Open
Abstract
HMGB1 that belongs to the High Mobility Group-box superfamily, is a nonhistone chromatin associated transcription factor. It is present in the nucleus of eukaryotes and can be actively secreted or passively released by kinds of cells. HMGB1 is important for maintaining DNA structure by binding to DNA and histones, protecting it from damage. It also regulates the interaction between histones and DNA, affecting chromatin packaging, and can influence gene expression by promoting nucleosome sliding. And as a DAMP, HMGB1 binding to RAGE and TLRs activates NF-κB, which triggers the expression of downstream genes like IL-18, IL-1β, and TNF-α. HMGB1 is known to be involved in numerous physiological and pathological processes. Recent studies have demonstrated the significance of HMGB1 as DAMPs in the female reproductive system. These findings have shed light on the potential role of HMGB1 in the pathogenesis of diseases in female reproductive system and the possibilities of HMGB1-targeted therapies for treating them. Such therapies can help reduce inflammation and metabolic dysfunction and alleviate the symptoms of reproductive system diseases. Overall, the identification of HMGB1 as a key player in disease of the female reproductive system represents a significant breakthrough in our understanding of these conditions and presents exciting opportunities for the development of novel therapies.
Collapse
Affiliation(s)
- Yu Ren
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| | - Damin Zhu
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Xingxing Han
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Qiqi Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Beili Chen
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Ping Zhou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Anhui Key Laboratory of Reproductive Health and Genetics, Anhui Medical University, Hefei, Anhui, China
- Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| | - Huijuan Zou
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- National Health Commission (NHC) Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei, China
- Key Laboratory of Population Health Across Life Cycle, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|