1
|
Pasare MA, Prepeliuc CS, Grigoriu MG, Miftode IL, Miftode EG. Biomarkers as Beacons: Illuminating Sepsis-Associated Hepato-Renal Injury. Int J Mol Sci 2025; 26:4825. [PMID: 40429966 DOI: 10.3390/ijms26104825] [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: 03/31/2025] [Revised: 05/13/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Sepsis, defined as a dysregulated host response to infection, is one of the leading causes of mortality worldwide. It unleashes in the organism a cascade of molecules, cytokines, and proteins which leads to an inflammatory storm. If this response to infection is uncontrolled, any organ is susceptible to damage. Acute kidney injury (AKI) is one of the most frequent organ dysfunctions in septic patients, and while it can be reversible, its presence leads to a higher burden of morbidity and mortality. While serum creatinine is essential in evaluating kidney function, the pathophysiology of AKI is not completely elucidated, and a plethora of novel biomarkers have been studied in the hope of an early diagnosis and fast treatment. While the liver is not as affected by sepsis, it plays an important role as a guardian by providing acute-phase proteins, activating neutrophils, and controlling iron balance. Acute liver failure (ALF) could impair the organism's capacity to contain and eliminate pathogens. Some molecules have been associated with either AKI or ALF, although biomarkers specific for organ dysfunction are difficult to validate. The aim of this review is to understand the role of several molecules in the pathophysiology of sepsis and their clinical ability for diagnosing or predicting sepsis-induced hepato-renal dysfunction.
Collapse
Affiliation(s)
- Maria-Antoanela Pasare
- Doctoral School, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- "Sf Parascheva" Clinical Hospital of Infectious Diseases, 700116 Iasi, Romania
| | - Cristian Sorin Prepeliuc
- Doctoral School, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Emergency Hospital "Mavromati", 710221 Botosani, Romania
| | | | - Ionela-Larisa Miftode
- "Sf Parascheva" Clinical Hospital of Infectious Diseases, 700116 Iasi, Romania
- Department of Infectious Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Egidia Gabriela Miftode
- "Sf Parascheva" Clinical Hospital of Infectious Diseases, 700116 Iasi, Romania
- Department of Infectious Diseases, Faculty of Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
2
|
Thakur N, Shukla S, Kumar M, Chethan G, Singh A, Radhika, Singh AK, Mandal KD, Saminathan M, Choudhary P, De U, Sarma K. Alteration in oxidative/nitrosative disparity and nephroprotective effect of hydroethanolic extract of Ocimum tenuiflorum L. in gentamicin-induced acute kidney injury. Ann Med Surg (Lond) 2025; 87:2674-2688. [PMID: 40337432 PMCID: PMC12055117 DOI: 10.1097/ms9.0000000000003230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/20/2025] [Indexed: 05/09/2025] Open
Abstract
Background Antibacterial, antioxidant, and antilipidemic properties of Ocimum tenuiflorum are well known from previous studies. This study was designed to study the phytochemical constituents, antioxidant efficacy, in vivo nephroprotective activity, and immunomodulatory potential of Ocimum tenuiflorum hydroethanolic extract in gentamicin-induced acute kidney injury (AKI) rat model. Methods Ocimum tenuiflorum extract was given for 8 days to gentamicin-induced toxicity (100 mg/kg) in rats. Nephroprotective and immunomodulatory efficacy of O. tenuiflorum extract was evaluated based on urine and serum biochemistry, blood and tissue oxidative stress indices, cytokine levels, kidney injury biomarkers, and histopathology. Results Gentamicin toxicity resulted in a reduction in catalase, glutathione reductase, superoxide dismutase, and interleukin-10 levels in blood and tissue homogenates, while an increase in serum creatinine, blood urea nitrogen, lipid peroxide, tumor necrosis factor-alpha, cystatin C, kidney injury molecule-1, and gamma-glutamyl transpeptidase levels. Treatment with O. tenuiflorum ameliorated oxidative stress, cytokine imbalance, and kidney injury; however, the results were almost similar to standard drug. Furthermore, histopathological analysis of kidney, liver, and heart tissues confirmed the organoprotective efficacy of O. tenuiflorum extract. Conclusion The present findings demonstrate the curative efficacy of O. tenuiflorum in gentamicin-induced AKI, probably mediated through phenolic and flavonoid phytoconstituents, antioxidant properties, and down-regulation of inflammatory cytokines. Therefore, future studies may be established to evaluate its efficacy and safety for clinical trials.
Collapse
Affiliation(s)
- Neeraj Thakur
- Department of Veterinary Medicine, College of Veterinary and Animal Sciences, GBPUA&T, Pantnagar, Uttarakhand, India
- Department of Veterinary Medicine, College of Veterinary Sciences and Animal Husbandry, CAU, Selesih, Aizawl, Mizoram, India
| | - S.K. Shukla
- Department of Veterinary Medicine, College of Veterinary and Animal Sciences, GBPUA&T, Pantnagar, Uttarakhand, India
| | - Mahesh Kumar
- Department of Veterinary Medicine, College of Veterinary and Animal Sciences, GBPUA&T, Pantnagar, Uttarakhand, India
| | - G.E. Chethan
- Department of Veterinary Medicine, College of Veterinary Sciences and Animal Husbandry, CAU, Selesih, Aizawl, Mizoram, India
| | - Alok Singh
- Department of Veterinary Medicine, College of Veterinary and Animal Sciences, GBPUA&T, Pantnagar, Uttarakhand, India
- Department of Teaching Veterinary Clinical Complex, College of Veterinary Sciences and Animal Husbandry, ANDUA&T, Kumarganj, Ayodhya, Uttar Pradesh, India
| | - Radhika
- Department of Veterinary Medicine, College of Veterinary and Animal Sciences, GBPUA&T, Pantnagar, Uttarakhand, India
| | - Anand Kumar Singh
- Department of Veterinary Medicine, College of Veterinary and Animal Sciences, GBPUA&T, Pantnagar, Uttarakhand, India
- Livestock Farm Complex (Veterinary Medicine), College of Veterinary and Animal Sciences, Nauner, Datia, Madhya Pradesh, India
| | - Kruti Debnath Mandal
- Veterinary Clinical Complex, Faculty of Veterinary and Animal Sciences, RGSC-Banaras Hindu University, Barkachha, Mirzapur, Uttar Pradesh, India
| | - M. Saminathan
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Priyanka Choudhary
- Department of Veterinary Microbiology, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Rampura Phul, Bathinda, Punjab, India
| | - U.K. De
- Division of Medicine, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Kalyan Sarma
- Department of Veterinary Medicine, College of Veterinary Sciences and Animal Husbandry, CAU, Selesih, Aizawl, Mizoram, India
| |
Collapse
|
3
|
Zhang C, Xiang Z, Yang P, Zhang L, Deng J, Liao X. Advances in Nano-Immunomodulatory Systems for the Treatment of Acute Kidney Injury. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409190. [PMID: 40145715 PMCID: PMC12061249 DOI: 10.1002/advs.202409190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/26/2025] [Indexed: 03/28/2025]
Abstract
Acute kidney injury (AKI) occurs when there is an imbalance in the immune microenvironment, leading to ongoing and excessive inflammation. Numerous immunomodulatory therapies have been suggested for the treatment of AKI, the current immunomodulatory treatment delivery systems are suboptimal and lack efficiency. Given the lack of effective treatment, AKI can result in multi-organ dysfunction and even death, imposing a significant healthcare burden on both the family and society. This underscores the necessity for innovative treatment delivery systems, such as nanomaterials, to better control pathological inflammation, and ultimately enhance AKI treatment outcomes. Despite the modification of numerous immunomodulatory nanomaterials to target the AKI immune microenvironment with promising therapeutic results, the literature concerning their intersection is scarce. In this article, the pathophysiological processes of AKI are outlined, focusing on the immune microenvironment, discuss significant advances in the comprehension of AKI recovery, and describe the multifunctionality and suitability of nanomaterial-based immunomodulatory treatments in managing AKI. The main obstacles and potential opportunities in the swiftly advancing research field are also clarified.
Collapse
Affiliation(s)
- Chenli Zhang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
- Department of nephrologySecond People's Hospital of YibinYibin644000China
| | - Zeli Xiang
- Department of nephrologySecond People's Hospital of YibinYibin644000China
| | - Pengfei Yang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Ling Zhang
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| | - Jun Deng
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
- Institute of Burn Research, Southwest HospitalState Key Lab of Trauma and Chemical PoisoningArmy Medical University (Third Military Medical University)Chongqing400038China
| | - Xiaohui Liao
- Department of NephrologyThe Second Affiliated HospitalChongqing Medical UniversityChongqing400016China
| |
Collapse
|
4
|
Wang H, Liao J, Wang W, Zhang J. A crucial role of miR-155 in the pathomechanism of acute kidney injury. Front Pharmacol 2025; 16:1570000. [PMID: 40308762 PMCID: PMC12040948 DOI: 10.3389/fphar.2025.1570000] [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: 02/02/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Acute kidney injury (AKI) is one of the nonnegligible causes of mortality worldwide. It is important to understand the underlying molecular mechanism of AKI to effective therapeutic targets. miR-155 has been found to play a pivotal role in the development of AKI, while a comprehensive review on this topic is currently still lacking. Based on this review, we found that miR-155and is strongly correlated with the pathophysiological development of AKI by modulating cell apoptosis, inflammation, and proliferation. Mechanistically, miR-155 exerts a promoting function in multiple types of AKI by regulating multiple proteins or signaling pathways, such as SOCS-1, ERRFI1, SOCS-1, TRF1, CDK12, and TCF4/Wnt/β-catenin pathway. The inhibition of miR-155 has a renoprotective effect in drug- or substance-induced AKI. Therefore, drugs or biological compounds targeted by miR-155 and its pathways may recover the process of AKI by altering apoptosis, inflammation, and pyroptosis. A miRNA nanocarrier system that has already been developed could offer a novel approach to treat AKI, providing a direction for future research. Further large-scale studies are necessary to elucidate the clinical significance of miR-155 as a potential therapeutic target for multiple types of AKI.
Collapse
Affiliation(s)
- Hui Wang
- Department of Urology, The First People’s Hospital of Linhai, Linhai, Zhejiang, China
| | - Jian Liao
- Department of Nephrology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, China
| | - Wei Wang
- Department of Urology, Tiantai People’s Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People’s Hospital), Hangzhou Medical College, Taizhou, Zhejiang, China
| | - Jianhua Zhang
- Department of Urology, Tiantai People’s Hospital of Zhejiang Province (Tiantai Branch of Zhejiang Provincial People’s Hospital), Hangzhou Medical College, Taizhou, Zhejiang, China
| |
Collapse
|
5
|
Huang X, Lu J, An Y, Xu M, Chen X, Liu C, Zhou X, Shan H, Qian Y, Zhang M. Electrospun PLGA/PCL Nanofiber Film Loaded with LPA Promotes Full-Layer Wound Healing by Regulating the Keratinocyte Pyroptosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:20756-20767. [PMID: 40152284 DOI: 10.1021/acsami.4c22495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Electrospun nanofibers have a number of qualities that make them a suitable choice for skin wound healing. Lysophosphatidic acid (LPA) stimulates the keratinocytes and fibroblasts to proliferate, differentiate, and migrate and enhances skin wound healing. Here, we developed the electrospun scaffolds contained in polycaprolactone (PCL) and polylactic-co-glycolic acid (PLGA). The scaffolds loaded with LPA nanoparticles retained a porous nanofiber structure and showed better physicochemical properties and biocompatibility. The scaffold continuously releases LPA to quickly initiate cell signaling and maintain long-term anti-inflammatory activity. In this study, we found that PP scaffold with LPA reduces the disordered collagen deposition and the thickness of the newborn epidermis, improves skin healing, and reduces scar formation. Explaining the mechanism of LPA mineralized tissue regeneration in skin wound healing, LPA inhibited the pyroptosis of keratinocyte, a cell death process that induces inflammation and scar formation by inhibiting the expression of TNF-α and β-catenin proteins. Thus, the electrospun PP scaffold with LPA can be potentially developed as a therapeutic avenue to target skin wound healing.
Collapse
Affiliation(s)
- Xinqi Huang
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Jianghuiwen Lu
- Department of Medical Aesthetic, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, Jiangsu, China
| | - Yumei An
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Mingyuan Xu
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xueshi Chen
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Chao Liu
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| | - Xuefeng Zhou
- School of Biological Science and Medical Engineering, Southeast University, Nanjing 210000, Jiangsu, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215000, Jiangsu, China
| | - Yunzhu Qian
- Department of Stomatology, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou 215123, Jiangsu, China
| | - Mingyang Zhang
- Department of Forensic Sciences, The Affiliated Guangji Hospital, School of Basic Medicine, Suzhou Medical College of Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
6
|
Bhattarai SM, Jhawer A, Frampton G, Troyanovskaya E, DeMorrow S, McMillin M. Characterization of hepatic pathology during azoxymethane-induced acute liver failure. World J Gastroenterol 2025; 31:103952. [PMID: 40182596 PMCID: PMC11962848 DOI: 10.3748/wjg.v31.i12.103952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/26/2025] [Accepted: 03/03/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Acute liver failure (ALF) is a loss of liver function due to a severe hepatic insult. Studies utilizing the azoxymethane (AOM) mouse model of ALF, which also generates hepatic encephalopathy, have primarily focused on development of neurological deficits. However, the molecular processes that generate liver damage have not been fully characterized. Therefore, a more comprehensive characterization of the hepatic consequences of AOM toxicity is needed to better understand this disease model. AIM To identify molecular pathology contributing to hepatic injury during the progression of AOM-induced ALF. METHODS C57BL/6 mice were injected with AOM to produce ALF and hepatic encephalopathy. Tissue was collected at defined stages of neurological decline up to coma. Liver injury, CYP2E1 expression, oxidative stress, inflammation, apoptosis, necroptosis, and hepatocellular senescence were assessed. RESULTS Increased hepatic necrosis and exacerbated liver injury were observed after AOM injection as mice progressed towards coma. CYP2E1 expression decreased in AOM-treated mice as liver injury progressed. Malondialdehyde, myeloperoxidase and other measures of oxidative stress were significantly increased during AOM-induced ALF. Hepatic CCL2 and tumor necrosis factor α expression increased as AOM-induced liver injury progressed. Mixed lineage kinase domain-like protein phosphorylation was increased early during the progression of AOM-induced liver injury. Measures of apoptosis and cellular senescence all increased as the time course of AOM progressed. CONCLUSION These data support that necrosis, oxidative stress, inflammation, apoptosis, and senescence were elevated in AOM-treated mice, with inflammation being the earliest significant change.
Collapse
Affiliation(s)
- Shadikchhya Maya Bhattarai
- Department of Internal Medicine, University of Texas at Austin Dell Medical School, Austin, TX 78701, United States
| | - Ashwin Jhawer
- Department of Internal Medicine, University of Texas at Austin Dell Medical School, Austin, TX 78701, United States
| | - Gabriel Frampton
- Department of Internal Medicine, University of Texas at Austin Dell Medical School, Austin, TX 78701, United States
| | - Eleonora Troyanovskaya
- Department of Research, Central Texas Veterans Health Care System, Austin, TX 78701, United States
| | - Sharon DeMorrow
- Department of Internal Medicine, University of Texas at Austin Dell Medical School, Austin, TX 78701, United States
- Department of Research, Central Texas Veterans Health Care System, Austin, TX 78701, United States
- Division of Pharmacology and Toxicology, University of Texas at Austin College of Pharmacy, Austin, TX 78701, United States
| | - Matthew McMillin
- Department of Internal Medicine, University of Texas at Austin Dell Medical School, Austin, TX 78701, United States
- Department of Research, Central Texas Veterans Health Care System, Austin, TX 78701, United States
- Huffington Department of Education, Baylor College of Medicine, Temple, TX 76548, United States
- Department of Medicine, Baylor College of Medicine, Temple, TX 76548, United States
| |
Collapse
|
7
|
Zhang H, Cui JG, Chen MS, Wang JX, Sun XH, Zhao Y, Li JL. TNF/TNFR1 Signaling Mediates DEHP-Induced Hepatocyte Pyroptosis via the GSDMD-mtROS Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7432-7444. [PMID: 39999303 DOI: 10.1021/acs.jafc.4c11022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Di(2-ethylhexyl) phthalate (DEHP), which is widely used in agricultural plastics, accumulates in humans and animals through the food chain over time, resulting in liver toxicity. Recent studies have reported that pyroptosis and mitochondrial damage are closely related to a variety of liver diseases, but the specific mechanism is still unclear. To address this issue, in vitro and in vivo hepatotoxicity models were established. The results demonstrated that exposure to DEHP caused a buildup of MEHP in livers, altered liver metabolite composition, and caused pyroptosis-like changes in hepatocytes. After DEHP treatment, REDOX homeostasis was unbalanced, and mitochondrial reactive oxygen species (mtROS) were overproduced. MEHP exposure activates pyroptosis mediated by TNF/TNFR1 signaling and upregulates the perforating protein GSDMD-N to destroy the mitochondrial membrane of hepatocytes. Above all, this study elucidates the potential involvement of TNF/TNFR1 signaling-mediated pyroptosis in mitochondrial damage and confirms that the regulation of pyroptosis is helpful in maintaining normal mitochondrial function.
Collapse
Affiliation(s)
- Hao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Jia-Gen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Ming-Shan Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Jia-Xin Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Xiao-Han Sun
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Yi Zhao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P.R. China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
| | - Jin-Long Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P.R. China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, Northeast Agricultural University, Harbin 150030, P.R. China
| |
Collapse
|
8
|
Wang JJ, Zheng Y, Li YL, Xiao Y, Ren YY, Tian YQ. Emerging role of mesenchymal stem cell-derived exosomes in the repair of acute kidney injury. World J Stem Cells 2025; 17:103360. [PMID: 40160687 PMCID: PMC11947899 DOI: 10.4252/wjsc.v17.i3.103360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/26/2024] [Accepted: 02/13/2025] [Indexed: 03/21/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid deterioration in kidney function and has a significant impact on patient health and survival. Mesenchymal stem cells (MSCs) have the potential to enhance renal function by suppressing the expression of cell cycle inhibitors and reducing the expression of senescence markers and microRNAs via paracrine and endocrine mechanisms. MSC-derived exosomes can alleviate AKI symptoms by regulating DNA damage, apoptosis, and other related signaling pathways through the delivery of proteins, microRNAs, long-chain noncoding RNAs, and circular RNAs. This technique is both safe and effective. MSC-derived exosomes may have great application prospects in the treatment of AKI. Understanding the underlying mechanisms will foster the development of new and promising therapeutic strategies against AKI. This review focused on recent advancements in the role of MSCs in AKI repair as well as the mechanisms underlying the role of MSCs and their secreted exosomes. It is anticipated that novel and profound insights into the functionality of MSCs and their derived exosomes will emerge.
Collapse
Affiliation(s)
- Juan-Juan Wang
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yu Zheng
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yan-Lin Li
- Clinical Laboratory, The First People's Hospital of Yancheng, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School, Yancheng 224000, Jiangsu Province, China
| | - Yin Xiao
- Department of Medical Imaging, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu Province, China
| | - Yang-Yang Ren
- Clinical Laboratory, Xinyi People's Hospital, Xuzhou 221000, Jiangsu Province, China
| | - Yi-Qing Tian
- Clinical Laboratory, Xuzhou Central Hospital, Xuzhou 221000, Jiangsu Province, China.
| |
Collapse
|
9
|
Huang CL, Qu HS, Li AL, Ying CQ, Shao H, Tang YZ, Chen HZ, Tung TH, Zhu JS. Design of a Highly Active Peptide Inhibitor of Farnesyltransferase and Its Protective Effect Against Acute Liver Failure. Drug Des Devel Ther 2025; 19:1909-1926. [PMID: 40098903 PMCID: PMC11912918 DOI: 10.2147/dddt.s505541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose Acute liver failure (ALF) is a fatal syndrome associated with massive hepatocyte death. Previous studies have found that Farnesyltransferase (FTase) inhibitors improve disease progression in mouse models of endotoxemia, sepsis, and autoimmune hepatitis. PANoptosis is a novel type of programmed cell death (PCD), including pyroptosis, apoptosis, and necrosis, that plays an important role in ALF. This study was designed and investigated whether the FTase inhibitor PD083176 (d2,d3,d5) could attenuate ALF progression by modulating PANoptosis. Methods Combining the technical tools of computational biology, structural biology and pharmacology, we designed and obtained three high-affinity human FTase inhibitors of PD083176(d2,d3,d5). Then, these FTase inhibitors were investigated by animal experiments by administering PD083176(d2,d3,d5) (10 mg/kg) before modeling with LPS (100 μg/kg)/D-GalN (300 mg/kg) or TAA (800 mg/kg). Results We found that ALF induced by LPS/D-GaIN or TAA were associated with increased farnesylated protein in the liver. PD083176(d2,d3,d5) not only inhibited hepatic farnesylated proteins but also significantly attenuated liver injury and mortality in ALF mice. Importantly, PD083176(d2,d3,d5) treatment effectively inhibited hepatocyte apoptosis (Bax, Bcl-xL and TUNEL cell counts), pyroptosis (Caspase-1 and GSDMD), and necrotic apoptosis (RIPK1 and RIPK3). Conclusion Collectively, these findings demonstrate that PD081376(d2,d3,d5) could alleviate LPS/D-GaIN or TAA-induced ALF by regulating apoptosis, pyroptosis, and necrotizing apoptosis, which might provide a new therapeutic strategy and scalability challenge for ALF.
Collapse
Affiliation(s)
- Chun-Lian Huang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Hang-Shuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - A-Li Li
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Chen-Qian Ying
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Hui Shao
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Yong-Zhi Tang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Hua-Zhong Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Tao-Hsin Tung
- Evidence-Based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Jian-Sheng Zhu
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| |
Collapse
|
10
|
Zhang S, Lu M, Shang W, Du H, Wang C, Wen Z, Duan T, Xu W, Liu J, Du J, Chen D. Network pharmacology, molecular docking, and experimental verification reveal the mechanism of Yi-Shen-Hua-Shi granules treating acute kidney injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 343:119320. [PMID: 39755185 DOI: 10.1016/j.jep.2025.119320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/06/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yi-Shen-Hua-Shi granules (YSHSG) have been shown to improve kidney function in various renal disorders, which are characterized by the sudden decline and impairment of kidney function. AIM OF THE STUDY To investigate the precise mechanisms and targets of YSHSG in combating sepsis-induced AKI. MATERIALS AND METHODS Through network pharmacology, the active ingredients, main target proteins, and related signaling pathways of YSHSG in the treatment of sepsis-induced AKI were predicted. The AKI model was induced by sepsis using the cecal ligation and puncture (CLP) technique. Prior to the operation, YSHSG was administered intragastrically once daily for 1 week. Blood and kidney tissues were collected 48 h post-CLP to verify the network pharmacology analysis. RESULTS The core target proteins of YSHSG in the treatment of sepsis-induced AKI include AKT1, JUN, IL6, PTGS2, NFKBIA, MAPK3, Caspase-3 and MMP9, which were further confirmed by molecular docking. Pathway analyses such as Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) show that YSHSG plays a role in protecting the kidneys from sepsis-induced AKI through the PI3K/AKT, TNF, and IL17 signaling pathways. These findings were validated using qPCR and western blotting. In vivo experiments demonstrated that YSHSG inhibits the activation of TNF and IL17 signaling pathways while protecting against deactivation of the PI3K/AKT signaling pathway in sepsis-induced AKI. YSHSG also exhibits an effect on attenuating inflammation response and pyroptosis processes associated with the PI3K/AKT, TNF, and IL17 signaling pathways. CONCLUSION YSHSG mitigated sepsis-induced AKI by influencing the PI3K/AKT, TNF, and IL17 signaling pathways associated with inflammation and pyroptosis.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Minmin Lu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Weifeng Shang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Hangxiang Du
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Changnan Wang
- School of Life Sciences, Shanghai University, No.99 Shangda Road, Shanghai, 200444, China
| | - Zhenliang Wen
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Tingting Duan
- Institute of Consun Co. for Chinese Medicine in Kidney Diseases, Guangdong Consun Pharmaceutical Group, Dongpeng Road 71, Guangzhou, China
| | - Wei Xu
- Department of Critical Care Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, No.639 Zhizaoju Road, Shanghai, 200011, China.
| | - Jiao Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China.
| | - Jiankui Du
- Department of Physiology, Navy Medical University, No.800 Xiangyin Road, Shanghai, 200433, China.
| | - Dechang Chen
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin 2nd Road, Shanghai, 200025, China.
| |
Collapse
|
11
|
Liu C, Liu W, Lu H, Fan Y, Wang A. Effects of Baicalin on Gout Based on Network Pharmacology, Molecular Docking, and in vitro Experiments. J Inflamm Res 2025; 18:1543-1556. [PMID: 39925939 PMCID: PMC11806711 DOI: 10.2147/jir.s480911] [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: 06/01/2024] [Accepted: 12/31/2024] [Indexed: 02/11/2025] Open
Abstract
Purpose Baicalin is a flavonoid of Scutellaria baicalensis Georgi. It possesses antipyretic, analgesic, and anti-inflammatory effects. It has great potential to treat gout. A network pharmacology approach, molecular docking and experimental validation were applied to investigate the pharmacological mechanisms of baicalin in treating gout. Methods The potential targets of baicalin were retrieved from the TCMSP, PharmMapper, STITCH, and Swiss Target Prediction databases. The gout-related targets were retrieved from the DrugBank, TTD, and Genecards databases. Then, the potential targets and signaling pathways were acquired via protein-protein interaction (PPI), as well as the Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Subsequently, the key targets were selected to dock with baicalin based on molecular docking. Finally, in vitro experiments were conducted to further validate the predictions. Results A total of 318 potential targets of baicalin and 752 gout-related targets were screened. TNF, VEGFA, MMP9, PTGS2, and TLR4 might be the hub therapeutic target genes. The TLR4/NF-κB signaling pathway might be the foremost pathway in baicalin against gout. Moreover, molecular docking showed that baicalin combined well with TNF, VEGFA, MMP9, COX-2, and TLR4, respectively. The results of cell experiments suggested that baicalin could reduce the levels of inflammatory cytokines by inhibiting the TLR4/NF-κB signaling pathway in MSU-stimulated THP-1 cells and regulate the expression of these hub targets. Conclusion These results revealed that baicalin possesses "multitarget, multipathway, multilevel" regulatory effects. From a therapeutic standpoint, baicalin may be a promising anti-inflammatory agent for alleviating gout.
Collapse
Affiliation(s)
- Chunliu Liu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Department of Respiratory Medicine, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, People’s Republic of China
| | - Wei Liu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Hang Lu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| | - Yihua Fan
- Department of Rheumatism and Immunity, Hospital of Chengdu University of Traditional Chinese Medicine, Sichuan Chengdu, People’s Republic of China
| | - Aihua Wang
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
| |
Collapse
|
12
|
Pei X, Ma S, Hong L, Zuo Z, Xu G, Chen C, Shen Y, Liu D, Li C, Li D. Molecular insights of T-2 toxin exposure-induced neurotoxicity and the neuroprotective effect of dimethyl fumarate. Food Chem Toxicol 2025; 196:115166. [PMID: 39617286 DOI: 10.1016/j.fct.2024.115166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
T-2 toxin, a potent environmental pollutant, has been proved to stimulate neuroinflammation, while the connection between T-2 toxin and pyroptosis remain elusive. Dimethyl fumarate (DMF), recently identified as a neuroprotectant and pyroptosis inhibitor, has potential therapeutic applications that are underexplored. Based on present study in vitro and vivo, we demonstrated that T-2 toxin induced the activation of NLRP3-Caspase-1 inflammasome in hippocampal neurons. In addition to proinflammatory mediator overexpression, gasdermin D (GSDMD)-dependently pyroptosis in the mouse hippocampal neuron cell line (HT22) treated by T-2 toxin was determined in our study. Moreover, the palliative effect of knockdown sequence of high mobility group B1 protein (HMGB1) provided more details for T-2 toxin-initiated pyroptosis. Importantly, we confirmed that DMF, as a novel inhibitor of GSDMD, could alleviate pyroptosis induced by T-2 toxin in an GSDMD targeting manner. In summary, our studies exposed the evidence that T-2 toxin could induce NLRP3 inflammasome activation and hippocampal neuronal pyroptosis. More notably, DMF was turn out to be a critical executioner for attenuating GSDMD-mediated pyroptosis. Our data found a new function of DMF and suggested a novel therapy strategy against mycotoxin-triggered neuronal inflammation, which leads to varieties of neurological diseases.
Collapse
Affiliation(s)
- Xingyao Pei
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Shuhui Ma
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Liang Hong
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Zonghui Zuo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Gang Xu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Chun Chen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Yao Shen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Dingkuo Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China
| | - Cun Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Daowen Li
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China.
| |
Collapse
|
13
|
Wei S, Han C, Mo S, Huang H, Luo X. Advancements in programmed cell death research in antitumor therapy: a comprehensive overview. Apoptosis 2025; 30:401-421. [PMID: 39487314 DOI: 10.1007/s10495-024-02038-0] [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] [Accepted: 10/24/2024] [Indexed: 11/04/2024]
Abstract
Cell death is a normal physiological process within cells that involves multiple pathways, such as normal DNA damage, cell cycle arrest, and programmed cell death (PCD). Cell death has been a hot spot of research in tumor-related fields, especially programmed cell death, which is a key form of cell death and is classified into different types according to the mechanism of occurrence, such as apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and disulfidptosis. Given the important role of PCD in maintaining tissue homeostasis and inhibiting tumorigenesis and development, more and more basic and clinical studies are devoted to revealing its potential application in anti-tumor strategies. The purpose of this review is to systematically review the regulatory mechanisms of PCD and to summarize the latest research progress of anti-tumor treatment strategies based on PCD.
Collapse
Affiliation(s)
- Shuxin Wei
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Shutian Mo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Hailian Huang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China
| | - Xiaoling Luo
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, 530021, China.
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning, 530021, China.
| |
Collapse
|
14
|
Datta S, Rahman MA, Koka S, Boini KM. High mobility group box 1 (HMGB1) mediates nicotine-induced podocyte injury. Front Pharmacol 2025; 15:1540639. [PMID: 39840112 PMCID: PMC11747285 DOI: 10.3389/fphar.2024.1540639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Cigarette smoking is a well-established risk factor for renal dysfunction. Smoking associated with renal damage bears distinct physiological correlations in conditions such as diabetic nephropathy and obesity-induced glomerulopathy. However, the cellular and molecular basis of such an association remains poorly understood. High mobility group box 1(HMGB1) is a highly conserved non-histone chromatin associated protein that largely contributes to the pathogenesis of chronic inflammatory and autoimmune diseases such as sepsis, atherosclerosis, and chronic kidney diseases. Hence, the present study tested whether HMGB1 contributes to nicotine-induced podocyte injury. Methods and Results Biochemical analysis showed that nicotine treatment significantly increased the HMGB1 expression and release compared to vehicle treated podocytes. However, prior treatment with glycyrrhizin (Gly), a HMGB1 binder, abolished the nicotine-induced HMGB1 expression and release in podocytes. Furthermore, immunofluorescent analysis showed that nicotine treatment significantly decreased the expression of podocyte functional proteins- podocin and nephrin as compared to control cells. However, prior treatment with Gly attenuated the nicotine-induced nephrin and podocin reduction. In addition, nicotine treatment significantly increased desmin expression and cell permeability compared to vehicle treated podocytes. However, prior treatment with Gly attenuated the nicotine-induced desmin expression and cell permeability. Mechanistic elucidation revealed that nicotine treatment augmented the expression of toll like receptor 4 (TLR4) and pre-treatment with Gly abolished nicotine induced TLR4 upregulation. Pharmacological inhibition of TLR4 with Resatorvid, a TLR4 specific inhibitor, also attenuated nicotine induced podocyte damage. Conclusion HMGB1 is one of the important mediators of nicotine-induced podocyte injury through TLR4 activation.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Mohammad Atiqur Rahman
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX, United States
| | - Krishna M. Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| |
Collapse
|
15
|
Tian X, Yuan L, Zeng Y. Butyrate attenuates SA-AKI by inhibiting pyroptosis via the STING-GSDMD axis. Biochem Biophys Res Commun 2025; 743:151143. [PMID: 39693943 DOI: 10.1016/j.bbrc.2024.151143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/24/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
Sepsis-associated acute kidney injury (SA-AKI) is a common and serious complication with high morbidity and mortality. The pathophysiology of SA-AKI is complex. The underlying mechanisms of SA-AKI remain unclear, and effective therapeutic strategies are limited. Butyrate is a type of short-chain fatty acid (SCFA) derived from the gut microbiota that plays a key role in kidney disease. However, the effect of butyrate on SA-AKI and its underlying mechanisms remain unclear. In this study, LPS was used to establish an SA-AKI model in C57BL/6 mice. Our results indicated that butyrate levels were substantially reduced in SA-AKI model mice. Notably, butyrate intervention attenuated kidney injury and inflammation in SA-AKI model mice. Moreover, the levels of NLRP3, STING, and GSDMD (a marker of pyroptosis) were significantly decreased by butyrate intervention. An in vitro model induced by LPS was established using HK-2 cells. Butyrate mitigated pyroptosis and reduced NLRP3, STING, and GSDMD protein expression. Furthermore, STING overexpression abrogated the downregulation of several proteins (NLRP3 and caspase 1) invovled in NLRP3 inflammsome-mediated pyroptosis and weakened the protective effect of butyrate. Hence, butyrate may attenuate SA-AKI by inhibiting pyroptosis via the STING-GSDMD axis, which provides a potential therapeutic strategy for preventing SA-AKI progression.
Collapse
Affiliation(s)
- Xiaofang Tian
- Department of Nephrology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 563000, Zunyi, Guizhou, China; Scientific Research Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 563000, Zunyi, Guizhou, China
| | - Liying Yuan
- Department of Nephrology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 563000, Zunyi, Guizhou, China; Scientific Research Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 563000, Zunyi, Guizhou, China
| | - Yizhou Zeng
- Department of Urology, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 563000, Zunyi, China; Scientific Research Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), 563000, Zunyi, Guizhou, China.
| |
Collapse
|
16
|
Wang L, Dong Z, Zhang Y, Peng L. Emerging Roles of High-mobility Group Box-1 in Liver Disease. J Clin Transl Hepatol 2024; 12:1043-1056. [PMID: 39649031 PMCID: PMC11622203 DOI: 10.14218/jcth.2024.00317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 12/10/2024] Open
Abstract
High-mobility group box-1 (HMGB1) is an architectural chromosomal protein with various roles depending on its cellular localization. Extracellular HMGB1 functions as a prototypical damage-associated molecular pattern that triggers inflammation and adaptive immune responses, mediated by specific cell surface receptors, including receptors for advanced glycation end products and toll-like receptors. Post-translational modifications of HMGB1 significantly impact various cellular processes that contribute to the pathogenesis of liver diseases. Recent studies have highlighted the close relationship between HMGB1 and the pathogenesis of acute liver injuries, including acetaminophen-induced liver injury, hepatic ischemia-reperfusion injury, and acute liver failure. In chronic liver diseases, HMGB1 plays a role in nonalcoholic fatty liver disease, alcohol-associated liver disease, liver fibrosis, and hepatocellular carcinoma. Targeting HMGB1 as a therapeutic approach, either by inhibiting its release or blocking its extracellular function, is a promising strategy for treating liver diseases. This review aimed to summarize the available evidence on HMGB1's role in liver disease, focusing on its multifaceted signaling pathways, impact on disease progression, and the translation of these findings into clinical interventions.
Collapse
Affiliation(s)
- Lu Wang
- Department of Diagnostics, Second School of Clinical Medicine, Binzhou Medical University, Yantai, Shandong, China
| | - Zhiwei Dong
- Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yeqiong Zhang
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liang Peng
- Department of Infectious Diseases, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
He X, Tian Y, Dong J, Yuan Y, Zhang S, Jing H. RNA-Seq Reveals the Mechanism of Pyroptosis Induced by Oxygen-Enriched IR780 Nanobubbles-Mediated Sono-Photodynamic Therapy. Int J Nanomedicine 2024; 19:13029-13045. [PMID: 39654803 PMCID: PMC11625641 DOI: 10.2147/ijn.s487412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Background Sono-photodynamic therapy (SPDT), the combination of sonodynamic therapy (SDT) and photodynamic therapy (PDT), is a promising tumor treatment method. However, the hypoxic tumor microenvironment greatly compromises the efficacy of SPDT. Pyroptosis, a new type of programmed cell death, is mainly induced by some chemotherapeutic drugs in the current research, and rarely by SPDT. RNA sequencing (RNA-seq) is a high-throughput sequencing technique that comprehensively profiles the transcriptome, revealing the full spectrum of RNA molecules in a cell. Here, we constructed IR780@O2 nanobubbles (NBs) with photoacoustic dual response and hypoxia improvement properties to fight triple negative breast cancer (TNBC), and demonstrated that SPDT could kill TNBC cells through pyroptosis pathway. RNA-seq further revealed potential mechanisms and related differentially expressed genes. Methods Thin-film hydration and mechanical vibration method were utilized to synthesize IR780@O2 NBs. Subsequently, we characterized IR780@O2 NBs and examined the cytotoxicity as well as ROS production ability. A series of experiments were conducted to verify that SPDT killed TNBC cells through pyroptosis. Results IR780@O2 NBs were successfully prepared and had certain stability. Compared with SDT alone, SPDT increased therapeutic effect by 1.67 times by generating more ROS, and the introduction of NBs and O2 NBs (2.23 times and 2.93 times compared with SDT alone) could further promote this process. Other experiments proved that TNBC cells died by pyroptosis pathway. Moreover, the in-depth mechanism revealed that colony stimulating factor (CSF) and C-X-C motif chemokine ligand (CXCL) could be potential targets for the occurrence of pyroptosis in TNBC cells. Conclusion The IR780@O2 NBs prepared in this study increased the degree of TNBC cell pyroptosis through SPDT effect and alleviation of hypoxia, and cellular senescence might be a biological process closely related to pyroptosis in TNBC.
Collapse
Affiliation(s)
- Xiang He
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Jialin Dong
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Shijie Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, People’s Republic of China
| |
Collapse
|
18
|
Li W, Liu X, Li H, Zeng J, Chen Y, Xu B. Metabolomic and transcriptomic insights into the mechanisms of renal ischemia-reperfusion injury progression. Sci Rep 2024; 14:30101. [PMID: 39627404 PMCID: PMC11615214 DOI: 10.1038/s41598-024-81600-x] [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: 07/12/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
Renal ischemia-reperfusion injury (IRI) is an important cause of acute kidney injury (AKI). However, the pathophysiological changes and mechanisms during IRI-AKI progression remain unclear. This study aims toinvestigate the potential mechanisms in the progression of IRI-AKI by integrating metabolomics and transcriptomics data, providing a reference for the subsequent identification of biomarkers and therapeutic targets. IRI-AKI rat models with 30 min of ischemia and 24-72 h of reperfusion surgery simulating the progression of AKI were established. Compared to the control group underwent sham surgery (NC group), most of the differentially expressed metabolites (DEMs) in IRI-AKI 24 h and IRI-AKI 72 h decreased, mainly including amino acids, organic acids, and carnitines. Additionally, we found that DEMs were mainly enriched in amino acid-related pathways, among which valine, leucine, and isoleucine biosynthesis were dramatically altered in all comparisons. Transcriptomics revealed that differentially expressed genes (DEGs) were primarily involved in amino acid, lipid, and fatty acid metabolism. By integrating metabolomics and transcriptomics, we found valine, leucine, and isoleucine biosynthesis play key roles in IRI-AKI development. Our findings concluded that valine, leucine, and isoleucine pathways are hubs that potentially connect transcriptomes to metabolomes, providing new insights regarding the pathogenesis of IRI-AKI and its potential biomarkers and therapeutic strategies.
Collapse
Affiliation(s)
- Wanyi Li
- Department of Clinical Laboratory, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, Sichuan, China
| | - Xiaoqing Liu
- Department of Clinical Laboratory, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, Sichuan, China
| | - Honglin Li
- Department of Clinical Laboratory, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, Sichuan, China
| | - Jiawei Zeng
- Department of Clinical Laboratory, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, Sichuan, China.
| | - Yan Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, 610000, Sichuan, China.
| | - Bei Xu
- Department of Clinical Laboratory, School of Medicine, Mianyang Central Hospital, University of Electronic Science and Technology of China, Mianyang, 621000, Sichuan, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, 621000, Sichuan, China.
| |
Collapse
|
19
|
Zhang H, Zhang J, Pan H, Yang K, Hu C. Astragaloside IV promotes the pyroptosis of airway smooth muscle cells in childhood asthma by suppressing HMGB1/RAGE axis to inactivate NF-κb pathway. Autoimmunity 2024; 57:2387100. [PMID: 39097915 DOI: 10.1080/08916934.2024.2387100] [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: 04/09/2024] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/06/2024]
Abstract
Childhood asthma, a common chronic childhood disease, leads to high mortality and morbidity in the world. Airway smooth muscle cells (ASMCs) is a group of multifunctional cells that has been found to be correlated with the pathogenesis of asthma. Astragaloside IV (AS-IV) is a compound extracted from Astragalus membranaceus, which has the anti-asthmatic effect. However, the role of molecular mechanisms regulated by AS-IV in the biological processes of ASMCs in asthma remains unclear. Our current study aims to investigate the downstream molecular mechanism of AS-IV in modulating the aberrant proliferation and pyroptosis of ASMCs in asthma. At first, we determined that the viability of ASMCs could be efficiently suppressed by AS-IV treatment (200 μM). Moreover, AS-IV promoted the pyroptosis and suppressed PDGF-BB-induced aberrant proliferation. Through mechanism investigation, we confirmed that AS-IV could suppress high mobility group box 1 (HMGB1) expression and prevent it from entering the cytoplasm. Subsequently, AS-IV blocked the interaction between HMGB1 and advanced glycosylation end product-specific receptor (RAGE) to inactivate NF-κB pathway. Finally, in vivo experiments demonstrated that AS-IV treatment can alleviate the lung inflammation in asthma mice. Collectively, AS-IV alleviates asthma and suppresses the pyroptosis of AMSCs through blocking HMGB1/RAGE axis to inactivate NF-κB pathway.
Collapse
Affiliation(s)
- Huahong Zhang
- Department of Pediatrics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Zhang
- Department of Pediatrics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Hangli Pan
- Department of Pediatrics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Ke Yang
- Department of Pediatrics, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Chongwei Hu
- Department of Pediatrics, First People's Hospital of Chun'an County, Hangzhou, Zhejiang Province, China
| |
Collapse
|
20
|
Gholampour F, Masjedi F, Janfeshan S, Karimi Z. Remote limb ischemic pre-conditioning prevents renal Ischemia/reperfusion injury in rats by modulating oxidative stress and TNF-α/NF-κB/TGF-/βapelin signaling pathway. Mol Biol Rep 2024; 52:4. [PMID: 39570475 DOI: 10.1007/s11033-024-10109-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Remote limb ischemic pre-conditioning (RIPreC) can invoke potent renal protection. The involvement of oxidative stress and inflammatory pathways in renal ischemia/reperfusion injury (I/RI) was also confirmed. This study was designed to investigate the RIPreC effects on IRI-induced kidney dysfunction in rats through NFĸB/TNF-α/TGF-ꞵ/apelin signaling pathway. METHODS Renal I/RI was induced by occluding the kidney arteries for 45 min, then reperfusion for 24 h. Four similar cycles of left femoral artery ischemia (2 min)/reperfusion (3 min) before the onset of kidney ischemia were performed to create RIPreC. Animals were randomly divided into three groups: sham, I/R, and RIPreC + I/R. Following the reperfusion phase, urine and blood samples were taken, and the kidney was removed for functional, molecular, and histological examination. RESULTS When compared to sham rats, renal IRI resulted in decreased creatinine clearance and increased sodium fractional excretion, lower antioxidant enzyme activities, higher malondialdehyde content and higher nuclear factor-kappa B (NF-κB), tumor necrosis factor-alpha (TNF-α), transforming growth factor-betta (TGF-β), and Apelin expression levels, and histologically damaged kidney tissue. All of the alterations, as mentioned earlier, were alleviated using the RIPreC treatment. CONCLUSION Thus, RIPreC can protect against renal dysfunction after renal I/RI via modulation of the TNF-α/NF-κB/TGF-ꞵ/Apelin signaling pathway and strengthening the antioxidant defense system.
Collapse
Affiliation(s)
| | - Fatemeh Masjedi
- Nephro-Urology Research Center, Shiraz University of Medical Sciences, Research Tower, Khalili Avenue, Shiraz, 7193635899, Iran
| | - Sahar Janfeshan
- Nephro-Urology Research Center, Shiraz University of Medical Sciences, Research Tower, Khalili Avenue, Shiraz, 7193635899, Iran
| | - Zeinab Karimi
- Nephro-Urology Research Center, Shiraz University of Medical Sciences, Research Tower, Khalili Avenue, Shiraz, 7193635899, Iran.
| |
Collapse
|
21
|
Choi A, Woo JS, Park YS, Kim JH, Chung YE, Lee S, Beom JH, You JS. TARGETED TEMPERATURE MANAGEMENT AT 36°C IMPROVES SURVIVAL AND PROTECTS TISSUES BY MITIGATING THE DELETERIOUS INFLAMMATORY RESPONSE FOLLOWING HEMORRHAGIC SHOCK. Shock 2024; 62:716-727. [PMID: 39186053 DOI: 10.1097/shk.0000000000002453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
ABSTRACT Hemorrhagic shock (HS) is a life-threatening condition with high mortality rates despite current treatments. This study investigated whether targeted temperature management (TTM) could improve outcomes by modulating inflammation and protecting organs following HS. Using a rat model of HS, TTM was applied at 33°C and 36°C after fluid resuscitation. Surprisingly, TTM at 33°C increased mortality, while TTM at 36°C significantly improved survival rates. It also reduced histological damage in lung and kidney tissues, lowered serum lactate levels, and protected against apoptosis and excessive reactive oxygen species production. TTM at 36°C inhibited the release of high mobility group box 1 protein (HMGB1), a key mediator of inflammation, and decreased proinflammatory cytokine levels in the kidneys and lungs. Moreover, it influenced macrophage behavior, suppressing the harmful M1 phenotype while promoting the beneficial M2 polarization. Cytokine array analysis confirmed reduced levels of proinflammatory cytokines with TTM at 36°C. These results collectively highlight the potential of TTM at 36°C as a therapeutic approach to improve outcomes in HS. By addressing multiple aspects of injury and inflammation, including modulation of macrophage responses and cytokine profiles, TTM at 36°C offers promising implications for critical care management after HS, potentially reducing mortality and improving patient recovery.
Collapse
Affiliation(s)
- Arom Choi
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji Sun Woo
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoo Seok Park
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hee Kim
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yong Eun Chung
- Department of Radiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sojung Lee
- Class of 2025, Biology B.S., Emory University, Atlanta, Georgia
| | - Jin Ho Beom
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Je Sung You
- Department of Emergency Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
22
|
Bao T, Liao T, Cai X, Lu B, Dai G, Pei S, Zhang Y, Li Y, Xu B. METTL3 mediated ferroptosis in chondrocytes and promoted pain in KOA via HMGB1 m6A modification. Cell Biol Int 2024; 48:1755-1765. [PMID: 39129231 DOI: 10.1002/cbin.12229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/23/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Methyltransferase-like 3 (METTL3) plays a role in the development of knee osteoarthritis (KOA). However, the mechanism underlying the role of METTL3 in KOA is unclear. This work investigated the effects of MELLT3 on ferroptosis and pain relief in in vitro and in vivo KOA models. Chondrocytes were treated with 10 ng/mL interleukin-1β (IL-1β) or 5 μM Erastin (ferroptosis inducer). IL-1β or Erastin treatment inhibited cell viability and glutathione levels; increased Fe2+, lipid reactive oxygen species and malondialdehyde production; and decreased glutathione peroxidase 4, ferritin light chain and solute carrier family 7 member 11 levels. The overexpression of METTL3 facilitated the N6-methyladenosine methylation of high mobility group box 1 (HMGB1). HMGB1 overexpression reversed the effect of sh-METTL3 on IL-1β-treated chondrocytes. A KOA rat model was established by the injection of monosodium iodoacetate into the joints and successful model establishment was confirmed by haematoxylin and eosin staining and Safranin O/Fast Green staining. METTL3 depletion alleviated cartilage damage, the inflammatory response, ferroptosis and knee pain in KOA model rats, and these effects were reversed by the addition of HMGB1. In conclusion, METTL3 depletion inhibited ferroptosis and the inflammatory response, and ameliorated cartilage damage and knee pain during KOA progression by regulating HMGB1.
Collapse
Affiliation(s)
- Tianchi Bao
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Taiyang Liao
- Department of Orthopedics and Traumatology, Affiliated Hospital of Nanjing University of Chinese Medicine/Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Xuefeng Cai
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Binjie Lu
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Gaole Dai
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Shuai Pei
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yunqing Zhang
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Yuwei Li
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Bo Xu
- Department of Orthopedics and Traumatology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| |
Collapse
|
23
|
Huang L, Wu Y, Sai W, Wang Y, Feng G, Lu Y, Chen F, Huang X, Zhao H, Gu Z, Yang B. HBSP inhibits tubular cell pyroptosis and apoptosis, promotes macrophage M2 polarization, and protects LPS-induced acute kidney injury. J Cell Mol Med 2024; 28:e70202. [PMID: 39584501 PMCID: PMC11586777 DOI: 10.1111/jcmm.70202] [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: 07/28/2024] [Revised: 10/11/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Sepsis-associated acute kidney injury (AKI) has high morbidity and mortality, but without cause-specific treatment. Erythropoietin derived Helix B surface peptide (HBSP) alleviates AKI, whereas its underlying mechanisms remain to be further explored. Here, the effects of HBSP on pyroptosis, apoptosis, macrophage polarization and repair were investigated in lipopolysaccharide (LPS)-induced AKI mouse model and cultured kidney epithelial cells. Systemic inflammation, compromised renal function and histology were demonstrated in LPS-treated mice, with upregulated pyroptotic and apoptotic key proteins in the kidneys including GSDMD-N, cleaved IL-1β, IL-18 and caspase-3. These proteins were localized in tubular areas and colocalized with aquaporin-1 (AQP1), with increased F4/80+ M1 macrophages. However, HBSP mitigated pyroptosis, apoptosis and inflammation, and promoted macrophage M2 polarization. In addition, HMGB1 and erythropoietin receptor (EPOR) were increased by LPS and decreased by HBSP, both of which were positively correlated with pyroptotic and apoptotic proteins. Moreover, HBSP reduced TNF-α and IL-6 mRNA levels, as well as pyroptosis and apoptosis in LPS-stimulated TCMK-1 cells. In conclusion, HBSP inhibited tubular pyroptosis and apoptosis, EPOR expression, promoted macrophage M2 polarization, and protected against LPS-induced AKI. These findings provide new mechanistic insights into the renoprotection of HBSP, and facilitate its potential for clinical applications and therapeutic strategies in sepsis-associated AKI.
Collapse
Affiliation(s)
- Lili Huang
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Yuanyuan Wu
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of Pathology, Medical SchoolNantong UniversityNantongChina
| | - Wenli Sai
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Clinical Medical Research CenterAffiliated Hospital of Nantong UniversityNantongChina
| | - Yanan Wang
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of NephrologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Guijuan Feng
- Department of StomatologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Yuqing Lu
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of NephrologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Fei Chen
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of NephrologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Xinzhong Huang
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of NephrologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Hongsheng Zhao
- Department of Critical Care MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Zhifeng Gu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Medical School of Nantong UniversityNantong UniversityNantongChina
| | - Bin Yang
- Nantong‐Leicester Joint Institute of Kidney ScienceAffiliated Hospital of Nantong UniversityNantongChina
- Department of NephrologyAffiliated Hospital of Nantong UniversityNantongChina
- Department of Cardiovascular Sciences, College of Life Sciences, University Hospitals of Leicester NHS TrustUniversity of LeicesterLeicesterUK
| |
Collapse
|
24
|
Gao K, Liu Y, Sun C, Wang Y, Bao H, Liu G, Ou J, Sun P. TNF-ɑ induces mitochondrial dysfunction to drive NLRP3/Caspase-1/GSDMD-mediated pyroptosis in MCF-7 cells. Sci Rep 2024; 14:25880. [PMID: 39468189 PMCID: PMC11519391 DOI: 10.1038/s41598-024-76997-4] [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: 03/31/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Pyroptosis is a gasdermin-mediated pro-inflammatory form of programmed cell death (PCD). Tumor necrosis factor-ɑ (TNF-ɑ) is an inflammatory cytokine, and some studies have shown that TNF-ɑ can cause pyroptosis of cells and exert anti-tumor effects. However, whether TNF-ɑ exerts anti-tumor effects on breast cancer cells by inducing pyroptosis has not been reported. In this study, to explore the impact of TNF-ɑ on pyroptosis in breast cancer cells, we treated MCF-7 cells with TNF-ɑ and found that TNF-ɑ induced cell death. Moreover, we observed that the dead cells were swollen with obvious balloon-like bubbles, which was a typical sign of pyroptosis. Further studies have found that the anti-tumor effect of TNF-ɑ on breast cancer cells in vitro was achieved through the canonical pyroptosis pathway. In addition, TNF-ɑ-induced pyroptosis in MCF-7 cells was associated with mitochondrial dysfunction, in which mitochondrial membrane potential was decreased and mitochondrial ROS production was increased. After inhibiting ROS production, the activation effect of TNF-ɑ on NLRP3/Caspase-1/GSDMD pathway was weakened, and the inhibitory effect of TNF-ɑ on the growth of MCF-7 cells in vitro was also decreased, further confirming the involvement of ROS in TNF-ɑ-induced pyroptosis. Overall, our study revealed a new mechanism by which TNF-ɑ exerts an anti-tumor effect by inducing pyroptosis in MCF-7 cells through the ROS/NLRP3/Caspase-1/GSDMD pathway, which may provide new therapeutic ideas for the treatment of breast cancer.
Collapse
Affiliation(s)
- Kexin Gao
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Yancui Liu
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Cheng Sun
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Ying Wang
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Hongrong Bao
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Guoyang Liu
- Department of Nuclear Medicine, Hongqi Hospital affiliated to Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China
| | - Jinrui Ou
- Department of Nuclear Magnetic, the Second People's Hospital of Mudanjiang City, Mudanjiang City, 157000, Heilongjiang, China
| | - Ping Sun
- Department of Anatomy, Mudanjiang Medical University, Mudanjiang City, 157000, Heilongjiang, China.
| |
Collapse
|
25
|
Sun M, Sun Q, Li T, Ren X, Xu Q, Sun Z, Duan J. Silica nanoparticles induce liver lipid metabolism disorder via ACSL4-mediated ferroptosis. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 359:124590. [PMID: 39043312 DOI: 10.1016/j.envpol.2024.124590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 07/25/2024]
Abstract
The disease burden of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide. Emerging evidence has revealed that silica nanoparticles (SiNPs) could disorder the liver lipid metabolism and cause hepatotoxicity, but the underlying mechanism remains unknown. The purpose of this study is to elucidate the molecular mechanism of hepatic lipid metabolism disorder caused by SiNPs, and to reveal the role of ferroptosis in SiNPs-induced hepatotoxicity. To explore the phenotypic changes in liver, the wild-type C57BL/6J mice were exposed to different doses of SiNPs (5, 10, 20 mg/kg·bw) with or without melatonin (20 mg/kg·bw). SiNPs accelerated hepatic oxidative stress and promoted pathological injury and lipid accumulation, resulting in NAFLD development. Melatonin significantly inhibited the oxidative damage caused by SiNPs. Then, the hepatocytes were treated with SiNPs, the ferroptosis inducer and inhibitor, respectively. In vitro, SiNPs (25 μg/mL) generated mitochondrial and intracellular Fe2+ accumulation and lipid peroxidation repair ability impairment, decreased the activity of GPX4 through ACSL4/p38 MAPK signaling pathway, resulting in ferroptosis of hepatocytes. Notably, Erastin (the ferroptosis activator, 5 μM) increased the sensitivity of hepatocytes to ferroptosis. Ferrostatin-1 (Fer-1, the ferroptosis inhibitor, 5 μM) restored GPX4 activity and protected against deterioration of lipid hydroperoxides (LOOHs) to salvage SiNPs-induced cytotoxicity. Finally, the liver tissue conditional ACSL4 knockout (cKO) mice and ACSL4-KO hepatocytes were adopted to further identify the role of the ACSL4-mediated ferroptosis on SiNPs-induced NAFLD development. The results displayed ACSL4 knockout could down-regulate the lipid peroxidation and ferroptosis, ultimately rescuing the progression of NAFLD. In summary, our data indicated that ACSL4/p38 MAPK/GPX4-mediated ferroptosis was a novel and critical mechanism of SiNPs-induced NAFLD.
Collapse
Affiliation(s)
- Mengqi Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Qinglin Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Tianyu Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Xiaoke Ren
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Qing Xu
- Core Facilities for Electrophysiology, Core Facilities Center, Capital Medical University, Beijing, 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, PR China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, PR China.
| |
Collapse
|
26
|
Hong EP, Han SW, Kim BJ, Youn DH, Rhim JK, Jeon JP, Park JJ. Target Gene-Based Association Study of High Mobility Group Box Protein 1 in Intracranial Aneurysms in Koreans. Brain Sci 2024; 14:969. [PMID: 39451983 PMCID: PMC11505682 DOI: 10.3390/brainsci14100969] [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: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
Objective: We investigated the effect of high mobility group box 1 (HMGB1) on intracranial aneurysms (IAs) by analyzing single-nucleotide polymorphisms (SNPs) based on genome-wide association study (GWAS) data. HMGB1 mRNA and protein expression levels in plasma were also analyzed. Methods: This study was a comprehensive analysis of a GWAS dataset, including 250 patients with IAs and 294 controls. The HMGB1 gene region was targeted within SNP rs3742305 ± 10 kbp. Multivariate logistic regression analysis determined its association with IAs after adjusting for relevant clinical factors. HMGB1 mRNA expression was analyzed in the plasma of 24 patients selected from the GWAS dataset. The HMGB1 protein was analyzed by Western blotting. Results: A total of seven polymorphisms, including rs1360485, rs185382445, rs2039338, rs1045411, rs3742305, rs2249825, and rs189034241, were observed. Two SNPs, including rs1045411 (UTR-3) and rs3742305 (intron), showed strong linkage disequilibrium (r2 = 0.99). However, none of the seven SNPs associated with IAs had an adjusted p-value of < 0.0016 on multiple comparison analysis. HMGB1 mRNA levels (2-ΔCt) did not differ significantly between patients with IAs and the control subjects [1.07 (1.00-1.15) in patients with IAs vs. 1.05 (0.94-1.12) in controls; p = 0.67)]. Also, no significant difference in the degree of plasma HMGB1 protein expression was seen between the two groups (p = 0.82). Conclusions: The number of SNPs associated with HMGB1 and the degree of HMGB1 mRNA and protein expression were not significantly different between patients diagnosed with IAs and the controls.
Collapse
Affiliation(s)
- Eun Pyo Hong
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24254, Republic of Korea; (E.P.H.); (S.W.H.); (B.J.K.); (D.H.Y.)
| | - Sung Woo Han
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24254, Republic of Korea; (E.P.H.); (S.W.H.); (B.J.K.); (D.H.Y.)
| | - Bong Jun Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24254, Republic of Korea; (E.P.H.); (S.W.H.); (B.J.K.); (D.H.Y.)
| | - Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24254, Republic of Korea; (E.P.H.); (S.W.H.); (B.J.K.); (D.H.Y.)
| | - Jong Kook Rhim
- Department of Neurosurgery, Jeju National University College of Medicine, Jeju 63241, Republic of Korea;
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea;
| | - Jeong Jin Park
- Department of Neurology, Konkuk University Medical Center, Seoul 05030, Republic of Korea
| |
Collapse
|
27
|
Katoch S, Patial V. Sirtuin 1 in regulating the p53/glutathione peroxidase 4/gasdermin D axis in acute liver failure. World J Gastroenterol 2024; 30:3850-3855. [PMID: 39350786 PMCID: PMC11438651 DOI: 10.3748/wjg.v30.i34.3850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/10/2024] Open
Abstract
In this editorial, we comment on the article by Zhou et al. The study reveals the connection between ferroptosis and pyroptosis and the effect of silent information regulator sirtuin 1 (SIRT1) activation in acute liver failure (ALF). ALF is characterized by a sudden and severe liver injury resulting in significant hepatocyte damage, often posing a high risk of mortality. The predominant form of hepatic cell death in ALF involves apoptosis, ferroptosis, autophagy, pyroptosis, and necroptosis. Glutathione peroxidase 4 (GPX4) inhibition sensitizes the cell to ferroptosis and triggers cell death, while Gasdermin D (GSDMD) is a mediator of pyroptosis. The study showed that ferroptosis and pyroptosis in ALF are regulated by blocking the p53/GPX4/GSDMD pathway, bridging the gap between the two processes. The inhibition of p53 elevates the levels of GPX4, reducing the levels of inflammatory and liver injury markers, ferroptotic events, and GSDMD-N protein levels. Reduced p53 expression and increased GPX4 on deletion of GSDMD indicated ferroptosis and pyroptosis interaction. SIRT1 is a NAD-dependent deacetylase, and its activation attenuates liver injury and inflammation, accompanied by reduced ferroptosis and pyroptosis-related proteins in ALF. SIRT1 activation also inhibits the p53/GPX4/GSDMD axis by inducing p53 acetylation, attenuating LPS/D-GalN-induced ALF.
Collapse
Affiliation(s)
- Swati Katoch
- Division of Dietetics and Nutrition Technology, Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Vikram Patial
- Division of Dietetics and Nutrition Technology, Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
28
|
Dai X, Liu Y, Wu Y, Wang S, Guo Q, Feng X, Zhao F, Li Y, Lan L, Li X. DYZY01 alleviates pulmonary hypertension via inhibiting endothelial cell pyroptosis and rescuing endothelial dysfunction. Eur J Pharmacol 2024; 978:176785. [PMID: 38942262 DOI: 10.1016/j.ejphar.2024.176785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024]
Abstract
Pulmonary hypertension (PH) is a malignant pulmonary vascular disease with a poor prognosis. Although the development of targeted drugs for this disease has made some breakthroughs in recent decades, PH remains incurable. Therefore, innovative clinical treatment methods and drugs for PH are still urgently needed. DYZY01 is a new drug whose main ingredient is high-purity cannabidiol, a non-psychoactive constituent of cannabinoids that was demonstrated to have anti-inflammatory and anti-pyroptosis properties. Several recent studies have found cannabidiol could improve experimental PH, whereas the mechanistic effect of it warrants further investigation. Thus, this study aimed to investigate whether DYZY01 can treat PH by inhibiting inflammation and pyroptosis and to reveal its underlying mechanism. We established hypoxia and monocrotaline (MCT)-induced PH rat models in vivo and treated them with either DYZY01 (10,50 mg/kg/d) or Riociguat (10 mg/kg/d) by oral administration. The mean pulmonary arterial pressure (mPAP), right ventricular hypertrophy index (RVHI), and extent of vascular remodeling were measured. Meanwhile, the effect of DYZY01 on human pulmonary arterial endothelial cells (HPAECs) was assessed in vitro. The results indicated that DYZY01 significantly reduced mPAP and RVHI in PH rats and reversed the extent of pulmonary vascular remodeling. This improvement may have been achieved by reducing endothelial cell pyroptosis via inhibiting the NF-κB/NLRP3/Caspase-1 pathway. Furthermore, DYZY01 could improve endothelial vascular function, possibly by regulating the secretion of vasodilator factors and inhibiting the proliferation and migration of pulmonary endothelial cells.
Collapse
MESH Headings
- Animals
- Pyroptosis/drug effects
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/physiopathology
- Hypertension, Pulmonary/pathology
- Rats
- Male
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Humans
- Rats, Sprague-Dawley
- NF-kappa B/metabolism
- Vascular Remodeling/drug effects
- Cannabidiol/pharmacology
- Cannabidiol/therapeutic use
- Disease Models, Animal
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Pulmonary Artery/drug effects
- Pulmonary Artery/pathology
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Xuejing Dai
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Yi Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Yusi Wu
- School of Medicine, Hunan Normal University, Changsha, 410013, Hunan, China
| | - Shubin Wang
- Deyi Pharmaceutical Company Ltd., 102600, Beijing, China
| | - Qing Guo
- Deyi Pharmaceutical Company Ltd., 102600, Beijing, China
| | - Xuexiang Feng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Feilong Zhao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, China
| | - Lan Lan
- Deyi Pharmaceutical Company Ltd., 102600, Beijing, China.
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
29
|
Li ZH, Wang Y, Yu XY. Exploring the role of pyroptosis and immune infiltration in sepsis based on bioinformatic analysis. Immunobiology 2024; 229:152826. [PMID: 38981197 DOI: 10.1016/j.imbio.2024.152826] [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: 01/16/2024] [Revised: 05/20/2024] [Accepted: 06/09/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE Sepsis is a disease that is typically treated in intensive care units with high mortality and morbidity. Pyroptosis is a newly identified type of programmed cell death and is characterized by inflammatory cytokine secretion. However, the role of pyroptosis in sepsis remains unclear. METHODS GSE28750 and GSE134347 datasets were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed pyroptosis genes (DEPGs) were identified between sepsis and healthy controls. Machine learning was used to further narrow the gene range. Receiver operating curves (ROC) were generated to estimate the diagnostic efficacy. Immune infiltration levels were estimated via single-sample gene set enrichment analysis (ssGSEA). A network database was used to predict the upstream transcription factors and miRNAs of DEPGs. Finally, the expression of the genes was validated by qRT-PCR between sepsis patients and healthy controls. RESULTS We found that the pyroptosis pathway was enriched and activated in sepsis. 8 DEPGs were identified. A heatmap showed that the genes, NLRC4, NAIP, IL-18, AIM2 and ELANE, were abundant in the sepsis samples, and the genes, NLRP1, CHMP7 and TP53, were abundant in the healthy control samples. The ssGSEA results showed that the abundances of activated dendritic cells, MDSC, macrophage, plasmacytoid dendritic cells, regulatory T-cells, and Th17-cells were significantly higher, while the activated B-cell, activated CD8 T-cell, CD56 dim tural killer cell, immature B-cell, monocyte, and T follicular helper cell abundances were lower in sepsis samples compared to healthy controls. The qRT-PCR results showed that the expression levels of NAIP, IL-18, TP53, CHMP7, NLRC4, ELANE and NLRP1 were consistant with the bioinformatic analyses, while the expression level of AIM2 has no significant difference. CONCLUSION Our study identified seven potential pyroptosis-related genes, NAIP, IL-18, TP53, CHMP7, NLRC4, ELANE and NLRP1. This study revealed that pyroptosis may promote sepsis development by activating the immune response.
Collapse
Affiliation(s)
- Zhi-Hua Li
- Department of critical medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Yi Wang
- Department of critical medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Xiang-You Yu
- Department of critical medicine, the First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China.
| |
Collapse
|
30
|
Neira G, Becerril S, Valentí V, Moncada R, Catalán V, Gómez-Ambrosi J, Colina I, Silva C, Escalada J, Frühbeck G, Rodríguez A. FNDC4 reduces hepatocyte inflammatory cell death via AMPKα in metabolic dysfunction-associated steatotic liver disease. Clin Nutr 2024; 43:2221-2233. [PMID: 39173437 DOI: 10.1016/j.clnu.2024.08.007] [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: 03/20/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND The molecular mediators responsible for the progression of metabolic dysfunction-associated steatotic liver disease (MASLD) to steatohepatitis (MASH) have not yet been completely disentangled. We sought to analyze whether FNDC4, an hepatokine and adipokine with anti-inflammatory properties, is involved in TNF-α-induced inflammatory cell death in patients with MASLD. METHODS Plasma FNDC4 (n = 168) and hepatic FNDC4 and inflammatory cell death (n = 65) were measured in samples from patients with severe obesity with available liver biopsy-proven MASLD diagnosis. The effect of FNDC4 on TNF-α-induced pyroptosis, apoptosis and necroptosis (PANoptosis) and mitochondrial dysfunction was studied in vitro using human HepG2 hepatocytes. RESULTS Compared with individuals with normal liver, patients with type 2 diabetes and MASLD exhibited decreased hepatic FNDC4 mRNA and protein levels, which were related to liver inflammation. An overexpression of TNF-α, its receptor TNF-R1 and factors involved in inflammatory cell death was also found in the liver of these patients. FNDC4-knockdown in HepG2 hepatocytes increased apoptotic cell death, while FNDC4 treatment blunted NLRP3 inflammasome-induced pyroptosis, apoptosis and necroptosis in TNF-α-stimulated hepatocytes. Moreover, FNDC4 improved TNF-α-induced hepatocyte mitochondrial dysfunction by enhancing mitochondrial DNA (mtDNA) copy number and OXPHOS complex subunits I, II, III and V protein expression. Mechanistically, AMP-activated protein kinase α (AMPKα) was required for the FNDC4-mediated inhibition of cell death and increase in mtDNA content. CONCLUSIONS FNDC4 acts as a hepatocyte survival factor favouring mitochondrial homeostasis and decreasing inflammatory cell death via AMPKα. Collectively, our study identifies FNDC4 as an attractive target to prevent hepatocellular damage in patients with MASLD.
Collapse
Affiliation(s)
- Gabriela Neira
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain; Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain; Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Inmaculada Colina
- Department of Internal Medicine, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Escalada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain; Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Spain; Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain.
| |
Collapse
|
31
|
Shen X, Zhang X, Li K, Huang G, Li X, Hou Y, Ge X. Combined bacterial translocation and cholestasis aggravates liver injury by activation pyroptosis in obstructive jaundice. Heliyon 2024; 10:e35793. [PMID: 39220957 PMCID: PMC11363856 DOI: 10.1016/j.heliyon.2024.e35793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
This study explores the mechanism by which obstructive jaundice (OJ) induces liver damage through pyroptosis. We induced OJ in rats via bile duct ligation and assessed liver damage using serum biochemical markers and histological analysis of liver tissue. Pyroptosis was investigated through immunofluorescence, ELISA, Western blot, and quantitative RT-PCR techniques. Additionally, we examined intestinal function and fecal microbiota alterations in the rats using 16S rDNA sequencing. In vitro experiments involved co-culturing Kupffer cells and hepatocytes, which were then exposed to bile and lipopolysaccharide (LPS). Our findings indicated that OJ modified the gut microbiota, increasing LPS levels, which, in conjunction with bile, initiated a cycle of inflammation, fibrosis, and cell death in the liver. Mechanistically, OJ elevated necrotic markers such as ATP, which in turn activated pyroptotic pathways. Increased levels of pyroptosis-related molecules, including NLRP3, caspase-1, gasdermin D, and IL-18, were confirmed. In our co-cultured cell model, bile exposure resulted in cell death and ATP release, leading to the activation of the NLRP3 inflammasome and its downstream effectors, caspase-1 and IL-18. The combination of bile and LPS significantly intensified pyroptotic responses. This study is the first to demonstrate that LPS and bile synergistically exacerbate liver injury by promoting necrosis and pyroptosis, unveiling a novel mechanism of OJ-associated hepatic damage and suggesting avenues for potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Xin Shen
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Zhang
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471002, Henan, China
| | - Kaiyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Guangming Huang
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Xinyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Yunlong Hou
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, 050000, Hebei, China
| | - Xin Ge
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| |
Collapse
|
32
|
Lu D, Tu L, Hu Y, Cai X. Prognostic value of systemic inflammatory response index for acute kidney injury and the prognosis of pediatric patients in critical care units. PLoS One 2024; 19:e0306884. [PMID: 39208322 PMCID: PMC11361669 DOI: 10.1371/journal.pone.0306884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/25/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND We proposed a link between the first systemic inflammatory response index (SIRI) and acute kidney injury (AKI), as well as the prognosis of pediatric patients in intensive care units (PICU). METHODS This study comprised 5114 children from the pediatric-specific intensive care (PIC) database. SIRI was estimated as a neutrophil monocyte lymphocyte ratio. All patients were arbitrarily allocated to the training set (n = 3593) and the validation cohort (n = 1521) and divided into two groups depending on their SIRI levels. The diagnostic value of SIRI for pediatric ICU patients was subsequently determined using LASSO regression models. RESULTS After controlling for additional confounding variables in the training set, the higher SIRI value (≥ 0.59) had a greater risk of AKI (adjusted odds ratio, OR, 3.95, 95% confidence interval, 95%CI, 2.91-5.36, P<0.001) and in-hospital mortality (hazard ratio, HR, 5.01, 95%CI 2.09-12.03, P<0.001). Similar findings were discovered in the validation set. Furthermore, the suggested nomogram derived from SIRI and other clinical metrics showed outstanding calibration capability as well as therapeutic usefulness in both groups. CONCLUSIONS SIRI is a reliable and useful factor for AKI and fatality in pediatric ICU patients, and the proposed nomogram based on SIRI yields an appropriate prediction value for critically sick pediatric patients.
Collapse
Affiliation(s)
- Danchi Lu
- Department of Emergency, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Lijuan Tu
- Department of Emergency, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaofang Cai
- Department of Emergency, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
33
|
Pan K, Lu Y, Cao D, Peng J, Zhang Y, Li X. Long Non-coding RNA SNHG1 Suppresses the Osteogenic Differentiation of Bone Marrow Mesenchymal Stem Cells by Binding with HMGB1. Biochem Genet 2024; 62:2869-2883. [PMID: 38038773 DOI: 10.1007/s10528-023-10564-w] [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: 05/26/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023]
Abstract
Osteoporosis (OP) has a significant detrimental impact on the health of the elder. Long-term clinical effectiveness of current drugs used for OP treatment is limited. Therefore, it is very important to explore novel treatment targets for OP. The expression of SNHG1, HMGB1, OCN and OPN in gene level was measured using RT-qPCR, and the protein expression was determined by Western blotting assay. The concentration of IL-1β and IL-18 in supernatant of the bone marrow mesenchymal stem cells (BMSCs) was measured by ELISA. The interaction between SNHG1 and HMGB1 was confirmed by RNA pull down. Besides, alizarin red staining was performed to evaluate the differentiation of BMSCs into osteoblast. SNHG1 and HMGB1 were found to be upregulated in the serum of OP patients. During the osteogenic differentiation of BMSCs, the expression of osteoblastogenesis markers (OCN and OPN) and the activity of ALP were upregulated, while the expression levels of SNHG1 and HMGB1 were decreased in a time-dependent manner. In addition, the interaction between SNHG1 and HMGB1, expression of pyroptosis-associated factors (caspase-1 p20 and GSDMD-N), and secretion of IL-1β and IL-18 were also decreased during osteogenic differentiation. Interestingly, increasing SNHG1 promoted HMGB1 expression, activated pyroptosis, but inhibited osteogenic differentiation. Silencing HMGB1 or inhibiting caspase-1 partially rescued the inhibitory effect of SNHG1 on osteogenic differentiation. Our findings indicate that SNHG1 suppresses the osteogenic differentiation of BMSCs by activating pyroptosis through interaction with HMGB1 and promotion of HMGB1 expression. Our work provides further evidence supporting SNHG1 acts as a potential target for OP treatment, and reveals for the first time that SNHG1 regulates osteogenic differentiation by affecting pyroptosis.
Collapse
Affiliation(s)
- Kaihua Pan
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Yuanyuan Lu
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Daning Cao
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Jiang Peng
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Yunqing Zhang
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China
| | - Xiaoming Li
- Department of Orthopaedics, The First Hospital of Changsha, No. 311, Yingpan Road, Kaifu District, Changsha, 410005, Hunan, People's Republic of China.
| |
Collapse
|
34
|
Chen L, Wang R, Lv X, Kan M, Zhang H, Qiu W, Chen S, Zhao J, Wen X, Meng X, Wang H, Zang H. Hepatic-derived BMP9 is involved in hepatic fibrosis-induced kidney injury through inhibition of renal VEGFA. Biochem Pharmacol 2024; 226:116371. [PMID: 38885771 DOI: 10.1016/j.bcp.2024.116371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 05/01/2024] [Accepted: 06/14/2024] [Indexed: 06/20/2024]
Abstract
Clinical observations suggest that acute kidney injury (AKI) occurs in approximately 20-50% of hospitalized cirrhotic patients, suggesting a link between the liver and kidney. Bone morphogenetic protein 9 (BMP9) is a protein produced primarily by the liver and can act on other tissues at circulating systemic levels. Previous studies have demonstrated that controlling abnormally elevated BMP9 in acute liver injury attenuates liver injury; however, reports on whether BMP9 plays a role in liver injury-induced AKI are lacking. By testing we found that liver injury in mice after bile duct ligation (BDL) was accompanied by a significant upregulation of the kidney injury marker kidney injury molecule (KIM-1). Interestingly, all these impairments were alleviated in the kidneys of hepatic BMP9 knockout (BMP9-KO) mice. Peritubular capillary injury is a key process leading to the progression of AKI, and previous studies have demonstrated that vascular endothelial growth factor A (VEGFA) plays a key role in maintaining the renal microvascular system. In animal experiments, we found that high levels of circulating BMP9 had an inhibitory effect on VEGFA expression, while renal tubular epithelial cell injury was effectively attenuated by VEGFA supplementation in the hypoxia-enriched-oxygen (H/R) constructs of the AKI cell model in both humans and mice. Overall, we found that elevated BMP9 in hepatic fibrosis can affect renal homeostasis by regulating VEGFA expression. Therefore, we believe that targeting BMP9 therapy may be a potential means to address the problem of clinical liver fibrosis combined with AKI.
Collapse
Affiliation(s)
- Le Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ruonan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaodong Lv
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Kan
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongtao Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wei Qiu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shao Chen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jiuling Zhao
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xin Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongmei Zang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innova-tive Drugs, School of Pharmacy, Anhui Medical University, The Key Laboratory of Anti-inflammatory of Immune Medicines, Hefei, China; Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
35
|
Wang Y, Liu X, Li K, Wang X, Zhang X, Qian D, Meng X, Yu L, Yan X, He Z. Self-Sulfhydrated, Nitro-Fixed Albumin Nanoparticles as a Potent Therapeutic Agent for the Treatment of Acute Liver Injury. ACS NANO 2024. [PMID: 39041805 DOI: 10.1021/acsnano.4c07297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Exogenous polysulfhydryls (R-SH) supplementation and nitric oxide (NO) gas molecules delivery provide essential antioxidant buffering pool components and anti-inflammatory species in cellular defense against injury, respectively. Herein, the intermolecular disulfide bonds in bovine serum albumin (BSA) molecules were reductively cleaved under native and mild conditions to expose multiple sulfhydryl groups (BSA-SH), then sulfhydryl-nitrosylated (R-SNO), and nanoprecipitated to form injectable self-sulfhydrated, nitro-fixed albumin nanoparticles (BSA-SNO NPs), allowing albumin to act as a NO donor reservoir and multiple sulfhydryl group transporter while also preventing unfavorable oxidation and self-cross-linking of polysulfhydryl groups. In two mouse models of ischemia/reperfusion-induced and endotoxin-induced acute liver injury (ALI), a single low dosage of BSA-SNO NPs (S-nitrosothiols: 4 μmol·kg-1) effectively attenuated oxidative stress and systemic inflammation cascades in the upstream pathophysiology of disease progression, thus rescuing dying hepatocytes, regulating host defense, repairing microcirculation, and restoring liver function. By mechanistically upregulating the antioxidative signaling pathway (Nrf-2/HO-1/NOQ1) and inhibiting the inflammatory cytokine storm (NF-κB/p-IκBα/TNF-α/IL-β), BSA-SNO NPs blocked the initiation of the mitochondrial apoptotic signaling pathway (Cyto C/Bcl-2 family/caspase-3) and downregulated the cell pyroptosis pathway (NLRP3/ASC/IL-1β), resulting in an increased survival rate from 26.7 to 73.3%. This self-sulfhydrated, nitro-fixed functionalized BSA nanoformulation proposes a potential drug-free treatment strategy for ALI.
Collapse
Affiliation(s)
- Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xiaohu Liu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Keyang Li
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xinyuan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xue Zhang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Deyao Qian
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xinlei Meng
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Xuefeng Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education/Sanya Oceanographic Institution, Ocean University of China, Qingdao/Sanya 266100/572024, China
- Sanya Oceanographic Institution, Sanya 572024, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
36
|
Zhang Q, Guo J, Shi C, Zhang D, Wang Y, Wang L, Gong Z. The SIRT2-AMPK axis regulates autophagy induced by acute liver failure. Sci Rep 2024; 14:16278. [PMID: 39009648 PMCID: PMC11251177 DOI: 10.1038/s41598-024-67102-w] [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: 01/23/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024] Open
Abstract
This study explores the role of SIRT2 in regulating autophagy and its interaction with AMPK in the context of acute liver failure (ALF). This study investigated the effects of SIRT2 and AMPK on autophagy in ALF mice and TAA-induced AML12 cells. The results revealed that the liver tissue in ALF model group had a lot of inflammatory cell infiltration and hepatocytes necrosis, which were reduced by SIRT2 inhibitor AGK2. In comparison to normal group, the level of SIRT2, P62, MDA, TOS in TAA group were significantly increased, which were decreased in AGK2 treatment. Compared with normal group, the expression of P-PRKAA1, Becilin1 and LC3B-II was decreased in TAA group. However, AGK2 enhanced the expression of P-PRKAA1, Becilin1 and LC3B-II in model group. Overexpression of SIRT2 in AML12 cell resulted in decreased P-PRKAA1, Becilin1 and LC3B-II level, enhanced the level of SIRT2, P62, MDA, TOS. Overexpression of PRKAA1 in AML12 cell resulted in decreased SIRT2, TOS and MDA level and triggered more autophagy. In conclusion, the data suggested the link between AMPK and SIRT2, and reveals the important role of AMPK and SIRT2 in autophagy on acute liver failure.
Collapse
Affiliation(s)
- Qingqi Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jin Guo
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chunxia Shi
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Danmei Zhang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yukun Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Luwen Wang
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zuojiong Gong
- Department of Infectious Diseases, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
37
|
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
|
38
|
Sun Y, Li F, Liu Y, Qiao D, Yao X, Liu GS, Li D, Xiao C, Wang T, Chi W. Targeting inflammasomes and pyroptosis in retinal diseases-molecular mechanisms and future perspectives. Prog Retin Eye Res 2024; 101:101263. [PMID: 38657834 DOI: 10.1016/j.preteyeres.2024.101263] [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: 07/30/2023] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/26/2024]
Abstract
Retinal diseases encompass various conditions associated with sight-threatening immune responses and are leading causes of blindness worldwide. These diseases include age-related macular degeneration, diabetic retinopathy, glaucoma and uveitis. Emerging evidence underscores the vital role of the innate immune response in retinal diseases, beyond the previously emphasized T-cell-driven processes of the adaptive immune system. In particular, pyroptosis, a newly discovered programmed cell death process involving inflammasome formation, has been implicated in the loss of membrane integrity and the release of inflammatory cytokines. Several disease-relevant animal models have provided evidence that the formation of inflammasomes and the induction of pyroptosis in innate immune cells contribute to inflammation in various retinal diseases. In this review article, we summarize current knowledge about the innate immune system and pyroptosis in retinal diseases. We also provide insights into translational targeting approaches, including novel drugs countering pyroptosis, to improve the diagnosis and treatment of retinal diseases.
Collapse
Affiliation(s)
- Yimeng Sun
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Fan Li
- Eye Center, Zhongshan City People's Hospital, Zhongshan, 528403, China
| | - Yunfei Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Dijie Qiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Xinyu Yao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Guei-Sheung Liu
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dequan Li
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chuanle Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China
| | - Tao Wang
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, Guangming District, Shenzhen, 518132, China; School of Basic Medical Sciences, Capital Medical University, 10 Xitoutiao You'anMen Street, Beijing, 100069, China
| | - Wei Chi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, 510060, China.
| |
Collapse
|
39
|
Zhang J, Che T, Wang L, Sun W, Zhao J, Chen J, Liu Y, Pu Q, Zhang Y, Li J, Li Z, Zhu Z, Fu Q, Wang X, Yuan J. Proteomics coupled transcriptomics reveals Slc34a1 and Slc34a3 downregulation as potential features of nephrotoxin-induced acute kidney injury. J Proteomics 2024; 302:105203. [PMID: 38782357 DOI: 10.1016/j.jprot.2024.105203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 05/11/2024] [Accepted: 05/20/2024] [Indexed: 05/25/2024]
Abstract
Acute kidney injury (AKI) stands as a prevalent and economically burdensome condition worldwide, yet its complex molecular mechanisms remain incompletely understood. To address this gap, our study employs a multifaceted approach, combining mass spectrometry and RNA sequencing technologies, to elucidate the intricate molecular landscape underlying nephrotoxin-induced AKI in mice by cisplatin- and LPS-induced. By examining the protein and RNA expression profiles, we aimed to uncover novel insights into the pathogenesis of AKI and identify potential diagnostic and therapeutic targets. Our results demonstrate significant down-regulation of Slc34a1 and Slc34a3, shedding light on their crucial roles in AKI pathology and highlighting their promise as actionable targets for diagnosis and treatment. This comprehensive analysis not only enhances our understanding of AKI pathophysiology but also offers valuable avenues for the development of targeted interventions to mitigate its clinical impact. SIGNIFICANCE: Nephrotoxicity acute kidney injury (AKI) is a common clinical condition whose pathogenesis is the process by which some drugs, chemicals or other factors cause damage to the kidneys, resulting in impaired kidney function. Although it has been proved that different nephrotoxic substances can affect the kidney through different pathways, whether they have a commonality has not been registered. Here, we combined transcriptomics and proteomics to study the molecular mechanism of LPS and cisplatin-induced nephrotoxic acute kidney injury finding that the down-regulation of Slc34a1 and Slc34a3 may be a critical link in nephrotoxic acute kidney injury, which can be used as a marker for its early diagnosis.
Collapse
Affiliation(s)
- Junying Zhang
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Tiantian Che
- Chongqing Nanan District Center for Diseases Control and Prevention, Chongqing 401336, China
| | - Liting Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Wei Sun
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jing Zhao
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiajia Chen
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yang Liu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Qi Pu
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Yu Zhang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China
| | - Jiani Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhangfu Li
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China
| | - Zhaojing Zhu
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China.
| | - Qihuan Fu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China.
| | - Xiaoyang Wang
- Biomedical Analysis Center, Army Medical University, Chongqing 400038, China..
| | - Jiangbei Yuan
- Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China; Hepato-Pancreato-Biliary Surgery, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Guangdong province, Shenzhen 518036, China.; Center for General Practice Medicine, Department of Infectious Diseases, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou 310014, Zhejiang, China; Department of Infectious Diseases, Affiliated Banan Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
40
|
Liu Z, Yuan X, Huang Y, Gu Z, Xue L, Xue S, Wang J. The Role of Interferon-Induced Proteins with Tetratricopeptide Repeats 1 and 2 in Sepsis-Induced Acute Liver Injury. Infect Drug Resist 2024; 17:2337-2349. [PMID: 38882652 PMCID: PMC11180434 DOI: 10.2147/idr.s459838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024] Open
Abstract
Background Sepsis refers to a life-threatening organ dysfunction which can be resulted from the infection-induced dysregulated host response. A large number of inflammatory cytokines are released to act on the liver, making the liver one of the common target organs for the development of multiple organ dysfunction syndrome (MODS) in patients with sepsis. Sepsis-induced acute liver injury (SALI) can aggravate systemic disease. As a result, it is of great clinical significance to comprehend the molecular biological mechanism of SALI and to identify the markers for evaluating SALI. Interferon-induced proteins with tetratricopeptide repeats 1 and 2 (IFIT1, IFIT2) have been recognized as the anti-inflammatory factors that are widely expressed in various organs. The present study was aimed at clarifying the roles of IFIT1 and IFIT2 in the development of SALI. Methods A two-sample Mendelian randomization (MR) analysis was employed. Summary statistics datas were obtained from GWAS for inflammatory factors [tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6)], IFIT2, and sepsis as well as liver injury. Independent SNPs were selected as instrumental variables (IVs). Inverse variance weighted (IVW) in the MR analysis was adopted as the primary method for estimating the causal associations of inflammatory factors and IFIT2 with two diseases, and the associations of inflammatory factors with IFIT2. Additionally, weighted median method, MR-Egger and sensitivity analyses were applied in assessing the robustness of the results and ensure the result reliability. Subsequently, 119 healthy volunteers, 116 patients with sepsis and 116 SALI patients were recruited. The ELISA method was employed to quantify the expression levels of TNF-α, IL-1β, and IL-6. Additionally, qRT-PCR was conducted to measure the expression of IFIT1 and IFIT2. Furthermore, the correlations of IFIT1 and IFIT2 with inflammatory factors, alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were explored. Results As shown by the MR analysis, the genetically predisposed sepsis was significantly associated with the risk of IL-1β, with an odds ratio (OR) of 1.069 (95% confidence interval (CI), 1.015-1.127, p = 0.0119), and negatively associated with the risk of IL-6, with an OR of 0.880 (95% CI: 0.792-0.979, p= 0.0184). Meanwhile, there were positive causal effects of IL-6 (OR = 1.269, 95% CI: 1.032-1.561, p= 0.0238), IL-1β (OR = 1.106, 95% CI: 1.010-1.211, p = 0.0299) and IFIT2 (OR = 1.191, 95% CI: 1.045-1.359, p = 0.0090) on liver injury. Additionally, there was a positive causal effect of IFIT2 (OR = 1.164, 95% CI: 1.035-1.309, p= 0.0110) on IL-1β. Upon sensitivity analyses, there was weak evidence of such effects, indicating that the findings of this study were robust and reliable. Our results revealed the elevated levels of TNF-α, IL-1β, and IL-6 in the blood samples of sepsis and SALI patients (p < 0.0001). Conversely, IFIT1 and IFIT2 demonstrated the significantly decreased levels in peripheral blood mononuclear cells (PBMCs) of SALI patients (p < 0.0001). Furthermore, the expression levels of IFIT1 and IFIT2 were both negatively correlated with ALT activity (r = -0.3426, p = 0.0002; r = -0.3069, p = 0.0008) and AST activity (r = -0.2483, p = 0.0072; r = -0.3261, p = 0.0004), respectively. Moreover, the expression of IFIT1 and IFIT2 was both negatively related to the levels of TNF-α (r = -0.5027, p < 0.0001; r = -0.4218, p < 0.0001), IL-1β (r = -0.3349, p = 0.0002; r = -0.4070, p < 0.0001) and IL-6 (r = -0.2734, p = 0.0030; r = -0.3536, p < 0.0001), respectively. Conclusion IFIT1 and IFIT2 can serve as the diagnostic markers for sepsis-related liver injury, and IFIT1 and IFIT2 may participate in the pathological process of sepsis-related liver injury by regulating inflammation and liver function.
Collapse
Affiliation(s)
- Zhipeng Liu
- Information Department, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, 225300, People's Republic of China
| | - Xinyu Yuan
- Emergency Department, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, 225300, People's Republic of China
| | - Yan Huang
- Medical College, Yangzhou University, Yangzhou, 225009, People's Republic of China
| | - Zihan Gu
- Nanjing University of Finance & Economics, Nanjing, 210023, People's Republic of China
| | - Lu Xue
- Department of Critical Care Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, 225300, People's Republic of China
| | - Shanshan Xue
- Institute of Clinical Laboratory, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, 225300, People's Republic of China
| | - Jun Wang
- Emergency Department, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, Jiangsu Province, 225300, People's Republic of China
| |
Collapse
|
41
|
Gu Z, Gu W, Zhang G, Tang Y, Wang M, Guo Y, Zhou L. Effects of magnesium sulfate combined with labetalol on inflammatory stress and pregnancy outcome of patients with gestational hypertension. Exp Ther Med 2024; 27:266. [PMID: 38756909 PMCID: PMC11097299 DOI: 10.3892/etm.2024.12554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/21/2023] [Indexed: 05/18/2024] Open
Abstract
Gestational hypertension (GH) is a common disorder during pregnancy that can cause adverse pregnancy outcomes. In the present study, magnesium sulfate (MgSO4) combined with labetalol was used for clinical treatment. Randomized controlled trial was conducted in 100 patients with GH, documented in the Department of Obstetrics and Gynecology (Taicang TCM Hospital) grouped into the experimental (Expt) and control (Ctrl) groups (n=50 cases/group). The Ctrl group was treated with MgSO4, whereas the Expt group was treated with MgSO4 + labetalol. The systolic blood pressure (SBP) and diastolic blood pressure (DBP) in the Expt group were not significantly different from those in the Ctrl group (P>0.05). By contrast, the SBP and DBP were significantly lower after treatment than those before treatment in both groups (P<0.05). Whole blood viscosity, plasma viscosity and hematocrit were significantly lower in the Expt group compared with those in the Ctrl group after treatment (P<0.05). High mobility group box-1 protein, homocysteine and serum cystatin C levels in the Expt group were also markedly lower than those in the Ctrl group after treatment (P<0.05). In the Expt group, the rate of spontaneous vaginal delivery was much higher, whereas the rates of cesarean section and postpartum hemorrhage were markedly lower than those in the Ctrl group (P<0.05). The occurrence of fetal intrauterine distress, placental abruption, neonatal asphyxia, premature birth and neonatal death were also significantly lower in the Expt group than those in the Ctrl group (P<0.05). In conclusion, MgSO4 + labetalol could improve inflammatory stress and the hemodynamics of patients with GH, and may have a marked antihypertensive effect. Thus, it may improve pregnancy outcome and reduce perinatal complications.
Collapse
Affiliation(s)
- Zhijuan Gu
- Department of Obstetrics and Gynecology, Taicang Hospital of Traditional Chinese Medicine (TCM) Affiliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu 215400, P.R. China
| | - Wenqing Gu
- Department of Obstetrics and Gynecology, Taicang Hospital of Traditional Chinese Medicine (TCM) Affiliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu 215400, P.R. China
| | - Guiping Zhang
- Department of Obstetrics and Gynecology, Taicang Hospital of Traditional Chinese Medicine (TCM) Affiliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu 215400, P.R. China
| | - Ye Tang
- Department of Obstetrics and Gynecology, Taicang Hospital of Traditional Chinese Medicine (TCM) Affiliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu 215400, P.R. China
| | - Minfang Wang
- Department of Obstetrics and Gynecology, Taicang Hospital of Traditional Chinese Medicine (TCM) Affiliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu 215400, P.R. China
| | - Yan Guo
- Department of Obstetrics and Gynecology, Taicang Hospital of Traditional Chinese Medicine (TCM) Affiliated to Nanjing University of Chinese Medicine, Taicang, Jiangsu 215400, P.R. China
| | - Liping Zhou
- Department of Obstetrics and Gynecology, Suzhou Municipal Hospital, Suzhou, Jiangsu 215002, P.R. China
| |
Collapse
|
42
|
Zhang H, Shi H, Xie W, Meng M, Wang Y, Ma N, Chang G, Shen X. Subacute ruminal acidosis induces pyroptosis via the mitophagy-mediated NLRP3 inflammasome activation in the livers of dairy cows fed a high-grain diet. J Dairy Sci 2024; 107:4092-4107. [PMID: 38278294 DOI: 10.3168/jds.2023-23718] [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: 05/09/2023] [Accepted: 12/23/2023] [Indexed: 01/28/2024]
Abstract
High-grain (HG) feeding can trigger subacute ruminal acidosis (SARA) and subsequent liver tissue injury. This study investigated pyroptosis and NLRP3 inflammasome activation in SARA-induced liver injury, and the role of mitophagy during this process. Twelve mid-lactating Holstein cows equipped with rumen fistulas were randomly divided into 2 groups: a low-grain (LG) diet group (grain:forage = 4:6) and a HG diet group (grain:forage = 6:4). Each group had 6 cows. The experiment lasted for 3 wk. The ruminal fluid was collected through the rumen fistula on experimental d 20 and 21, and the pH immediately measured. At the end of the experiment, all animals were slaughtered, and peripheral blood and liver tissue were collected. The ruminal pH was lower in the HG group than that in the LG group at all time points. In addition, the ruminal pH in the HG group was lower than 5.6 at 3 consecutive time points after feeding (4, 6, and 8 h on d 20; 2, 4, and 6 h on d 21), indicating that HG feeding induced SARA. The content of lipopolysaccharide, IL-1β, and apoptosis-related cysteine protease 1 (caspase-1) and the activity of alanine aminotransferase and aspartate aminotransferase in the blood plasma of the HG group were all significantly increased. Hepatic caspase-1 activity was increased in the livers of the HG group. The increased expression levels of pyroptosis- and NLRP3 inflammasome-related genes IL1B, IL18, gasdermin D (GSDMD), apoptosis-associated speck-like protein containing a card (ASC), NLR family pyrin domain-containing 3 (NLRP3), and caspase-1 (CASP1) in liver tissue of the HG group were detected. Furthermore, western blot analysis showed that HG feeding led to increased expression of pyroptosis- and NLRP3 inflammasome-related proteins GSDMD N-terminal (GSDMD-NT), IL-1β, IL-18, cleaved-caspase-1, ASC, NLRP3, and cleaved-caspase-11 and upregulated expression of mitophagy-related proteins microtubule-associated protein 1 light chain 3 II (MAP1LC3-II), beclin 1 (BECN1), Parkin, and PTEN-induced kinase 1 (PINK1) in liver tissue. Collectively, our results revealed that SARA caused increased mitophagy and activated the NLRP3 inflammasome, causing pyroptosis and subsequent liver injury in dairy cows fed a HG diet.
Collapse
Affiliation(s)
- Hongzhu Zhang
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Huimin Shi
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Wan Xie
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Meijuan Meng
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Yan Wang
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Nana Ma
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Guangjun Chang
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China
| | - Xiangzhen Shen
- Ministry of Education Joint International Research Laboratory of Animal Health and Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, Jiangsu, PR China.
| |
Collapse
|
43
|
He Q, Qi Q, Ibeanu GC, Li PA. B355252 Suppresses LPS-Induced Neuroinflammation in the Mouse Brain. Brain Sci 2024; 14:467. [PMID: 38790446 PMCID: PMC11119117 DOI: 10.3390/brainsci14050467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 04/29/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
B355252 is a small molecular compound known for potentiating neural growth factor and protecting against neuronal cell death induced by glutamate in vitro and cerebral ischemia in vivo. However, its other biological functions remain unclear. This study aims to investigate whether B355252 suppresses neuroinflammatory responses and cell death in the brain. C57BL/6j mice were intraperitoneally injected with a single dosage of lipopolysaccharide (LPS, 1 mg/kg) to induce inflammation. B355252 (1 mg/kg) intervention was started two days prior to the LPS injection. The animal behavioral changes were assessed pre- and post-LPS injections. The animal brains were harvested at 4 and 24 h post-LPS injection, and histological, biochemical, and cytokine array outcomes were examined. Results showed that B355252 improved LPS-induced behavioral deterioration, mitigated brain tissue damage, and suppressed the activation of microglial and astrocytes. Furthermore, B355252 reduced the protein levels of key pyroptotic markers TLR4, NLRP3, and caspase-1 and inhibited the LPS-induced increases in IL-1β, IL-18, and cytokines. In conclusion, B355252 demonstrates a potent anti-neuroinflammatory effect in vivo, suggesting that its potential therapeutic value warrants further investigation.
Collapse
Affiliation(s)
- Qingping He
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| | - Qi Qi
- Human Vaccine Institute, Department of Surgery, Duke University Medical Center, Durham, NC 27707, USA;
| | - Gordon C. Ibeanu
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| | - P. Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute and Technology Enterprise (BRITE), College of Health and Sciences, North Carolina Central University, Durham, NC 27707, USA; (Q.H.); (G.C.I.)
| |
Collapse
|
44
|
Qin Y, Zhou W, Zhou X, Li H. Case report: Recombinant human type II tumour necrosis factor receptor-antibody fusion protein induced occult hepatitis B virus reactivation leading to liver failure. J Int Med Res 2024; 52:3000605241252580. [PMID: 38760056 PMCID: PMC11107333 DOI: 10.1177/03000605241252580] [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: 10/18/2023] [Accepted: 04/15/2024] [Indexed: 05/19/2024] Open
Abstract
Recombinant human type II tumour necrosis factor receptor-antibody fusion protein (rh TNFR:Fc) is an immunosuppressant approved for treating rheumatoid arthritis (RA). This case report describes a case of hepatitis B reactivation in a patient with drug-induced acute-on-chronic liver failure. A 58-year-old woman with a history of RA was treated with rh TNFR:Fc; and then subsequently received 25 mg rh TNFR:Fc, twice a week, as maintenance therapy. No anti-hepatitis B virus (HBV) preventive treatment was administered. Six months later, she was hospitalized with acute jaundice. HBV reactivation was observed, leading to acute-on-chronic liver failure. After active treatment, the patient's condition improved and she recovered well. Following careful diagnosis and treatment protocols are essential when treating RA with rh TNFR:Fc, especially in anti-hepatitis B core antigen antibody-positive patients, even when the HBV surface antigen and the HBV DNA are negative. In the case of HBV reactivation, liver function parameters, HBV surface antigen and HBV DNA should be closely monitored during treatment, and antiviral drugs should be used prophylactically when necessary, as fatal hepatitis B reactivation may occur in rare cases. A comprehensive evaluation and medication should be administered in a timely manner after evaluating the patient's physical condition and closely monitoring the patient.
Collapse
Affiliation(s)
- Yujie Qin
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Wenxiu Zhou
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xingnian Zhou
- Department of Infectious Diseases, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Hong Li
- Department of Infectious Diseases, Guizhou Provincial People’s Hospital, Guiyang, Guizhou Province, China
| |
Collapse
|
45
|
Chandra Shill M, El-Nashar HAS, Prova Mollick P, Nath Acharyya R, Afrin S, Hossain H, Halder S, Torequl Islam M, Bhuia MS, Reza HM, El-Shazly M, Mubarak MS. Longevity Spinach (Gynura procumbens) Ameliorated Oxidative Stress and Inflammatory Mediators in Cisplatin-Induced Organ Dysfunction in Rats: Comprehensive in vivo and in silico Studies. Chem Biodivers 2024; 21:e202301719. [PMID: 38361048 DOI: 10.1002/cbdv.202301719] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 02/17/2024]
Abstract
This study focused to assess the efficacy of Gynura procumbens (GP) leaf extract against cisplatin (CP)-induced hepatorenal complications in Wister albino rats. Additionally, it aims to detect polyphenolic compounds using high-performance liquid chromatography with diode-array detection (HPLC-DAD). The rats were treated intraperitoneally with CP (7.5 mg/kg) to mediate hepatorenal damage. They were then treated with GP extract (75 and 150 mg/kg, P.O.) for 7 consecutive days. Although GP extract significantly ameliorated CP-mediated hepatorenal biomarkers like alkaline phosphatase (ALP), alanine aminotransferase (ALT), aspartate aminotransferase (AST), creatinine, and blood urea nitrogen (BUN) levels in a dose-dependent manner, GP extract at 150 mg/kg dose normalized hepatorenal biomarkers ALP (45.11 U/L), ALT (34 U/L), AST (29 U/L), creatinine (10.3 mg/dl) and BUN (11.19 mg/dl) while comparing to control and disease group. Similarly, though it significantly reduced CP-induced oxidative stress inducers, including nitric oxide (NO) and advanced oxidative protein products (AOPP), higher dose (150 mg/kg) exhibited better activity in reducing NO (281.54 mmol/gm tissue in liver and 52.73 mmol/gm tissue in the kidney) and AOPP (770.95 mmol/mg protein in liver and 651.90 mmol/mg protein in the kidney). Besides, it showed better enhancement in the antioxidant enzymes superoxide dismutase, and glutathione levels at a higher dose (150 mg/kg). Histopathological studies showed that CP caused collagen accumulation in the liver and kidney tissues. GP extract drained the collagen mass and acted against hepatorenal damage. Ellagic acid, gallic acid, quercetin hydrate, kaempferol, and rutin hydrate were revealed in GP extract. In-silico modelling showed good docking scores of the polyphenolic compounds with molecular targets including CYP4502E1, NF-κB, caspase-3, and TNF-α. GP could be an effective therapeutic option for management of anticancer drugs' complications like CP-induced organ damage, although clinical studies are required to establish herbal formulation.
Collapse
Affiliation(s)
- Manik Chandra Shill
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Heba A S El-Nashar
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | | | | | - Silvia Afrin
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Hemayet Hossain
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka, 1205, Bangladesh
| | - Shimul Halder
- Department of Pharmaceutical Technology, Dhaka University, Dhaka, 1000, Bangladesh
| | - Muhammad Torequl Islam
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
- Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Bioluster Research Center, Gopalganj, 8100, Dhaka, Bangladesh
| | - Md Shimul Bhuia
- Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Bioluster Research Center, Gopalganj, 8100, Dhaka, Bangladesh
| | - Hasan Mahmud Reza
- Department of Pharmaceutical Sciences, North South University, Dhaka, 1229, Bangladesh
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt
| | | |
Collapse
|
46
|
Liu H, Liu H, Huang G, Yuan H, Zhang X. The roles of pyroptosis in genitourinary diseases. Int Urol Nephrol 2024; 56:1515-1523. [PMID: 38103146 PMCID: PMC11001749 DOI: 10.1007/s11255-023-03894-6] [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: 09/02/2023] [Accepted: 11/15/2023] [Indexed: 12/17/2023]
Abstract
Pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, is thought to be closely associated with the pathogenesis of diseases. Recently, the association between pyroptosis and urinary diseases has attracted considerable attention, and a comprehensive review focusing on this issue is not available. In this study, we reviewed the role of pyroptosis in the development and progression of benign urinary diseases and urinary malignancies. Based on this, pyroptosis has been implicated in the development of urinary diseases. In summary, this review sheds light on future research directions and provides novel ideas for using pyroptosis as a powerful tool to fight urinary diseases.
Collapse
Affiliation(s)
- Haopeng Liu
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Haoran Liu
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Guoshuai Huang
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China
| | - Hexing Yuan
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.
| | - Xuefeng Zhang
- Department of Urology, The First Affiliated Hospital of Soochow University, 188 Shizi Road, Suzhou, China.
| |
Collapse
|
47
|
Qu S, Fang J, Zhao S, Wang Y, Gao W, Li Z, Xu H, Zhang Y, Shi S, Cheng X, Liu Z, Jin L, Yao Y. Associations of dietary inflammatory index with low estimated glomerular filtration rate, albuminuria and chronic kidney disease in U.S adults: Results from the NHANES 2011-2018. Nutr Metab Cardiovasc Dis 2024; 34:1036-1045. [PMID: 38267324 DOI: 10.1016/j.numecd.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 11/09/2023] [Accepted: 11/12/2023] [Indexed: 01/26/2024]
Abstract
BACKGROUND AND AIMS Chronic Kidney Disease (CKD) is characterized by a high inflammation status with ever-increasing prevalence, and defined as low estimated glomerular filtration rate (eGFR) or albuminuria. Both low eGFR and albuminuria can have independent effects on the body. The dietary inflammatory index (DII) is a validated tool used to assess the inflammatory potential of the diet. We aim to explore not only the association between DII and CKD, but also the associations of DII with low eGFR and albuminuria, respectively. In addition, their associations in different subgroups remain to be explored. METHODS AND RESULTS 18,070 participants from the 2011-2018 NHANES with complete data of dietary intake and laboratory data were involved in our study. The data of 24-hour dietary recall interview was used to calculate DII, CKD could be reflected by laboratory data of creatinine and albumin. Then weighted multivariate logistic regression models and subgroup analyses were performed. The prevalence of low eGFR, albuminuria and CKD were 6.8%, 9.8% and 14.5%, respectively. A positive association between DII and low eGFR was observed (OR=1.12, 95%CI: 1.05-1.21), Q2, Q3 and Q4 are positively associated with a significant 39%, 65% and 71% increased risk of low eGFR compared with Q1 (P for trend<0.05). DII was also associated with CKD (OR=1.06, 95%CI: 1.01-1.11). CONCLUSION Significant positive associations of DII with CKD and low eGFR were observed. But we didn't find such association between DII and albuminuria.
Collapse
Affiliation(s)
- Shifang Qu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Jiaxin Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Saisai Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Yuxiang Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Wenhui Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Zhiyao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Han Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Yuan Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Shunyao Shi
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Xiaowei Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Zhigang Liu
- Osteopathic Center, The Second Hospital of Jilin University, No.4026, Yatai Street, Nanguan District, Changchun, Jilin, China.
| | - Lina Jin
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| | - Yan Yao
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
48
|
Xiong J, Zhao J. Pyroptosis: The Determinator of Cell Death and Fate in Acute Kidney Injury. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:118-131. [PMID: 38751798 PMCID: PMC11095617 DOI: 10.1159/000535894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/15/2023] [Indexed: 05/18/2024]
Abstract
Background Acute kidney injury (AKI) is kidney damage that leads to a rapid decline in function. AKI primarily occurs when the tubular epithelium is damaged, causing swelling, loss of brush margin, and eventual apoptosis. Research has shown that tubular epithelial cell damage in AKI is linked to cell cycle arrest, autophagy, and regulation of cell death. Summary Pyroptosis, a type of programmed cell death triggered by inflammation, is believed to play a role in the pathophysiology of AKI. Cumulative evidence has shown that pyroptosis is the main cause of tubular cell death in AKI. Thus, targeted intervention of pyroptosis may be a promising therapeutic approach for AKI. This review delves deep into the cutting-edge research surrounding pyroptosis in the context of AKI, shedding light on its intricate mechanisms and potential implications for clinical practice. Additionally, we explore the exciting realm of potential preclinical treatment options for AKI, aiming to pave the way for future therapeutic advancements. Key Messages Pyroptosis, a highly regulated form of cell death, plays a crucial role in determining the fate of cells during the development of AKI. This intricate process involves the activation of inflammasomes, which are multi-protein complexes that initiate pyroptotic cell death. By understanding the mechanisms underlying pyroptosis, researchers aim to gain insights into the pathogenesis of AKI and potentially identify new therapeutic targets for this condition.
Collapse
Affiliation(s)
- Jiachuan Xiong
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| | - Jinghong Zhao
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Kidney Center of PLA, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, PR China
| |
Collapse
|
49
|
Osna NA, Tikhanovich I, Ortega-Ribera M, Mueller S, Zheng C, Mueller J, Li S, Sakane S, Weber RCG, Kim HY, Lee W, Ganguly S, Kimura Y, Liu X, Dhar D, Diggle K, Brenner DA, Kisseleva T, Attal N, McKillop IH, Chokshi S, Mahato R, Rasineni K, Szabo G, Kharbanda KK. Alcohol-Associated Liver Disease Outcomes: Critical Mechanisms of Liver Injury Progression. Biomolecules 2024; 14:404. [PMID: 38672422 PMCID: PMC11048648 DOI: 10.3390/biom14040404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/28/2024] Open
Abstract
Alcohol-associated liver disease (ALD) is a substantial cause of morbidity and mortality worldwide and represents a spectrum of liver injury beginning with hepatic steatosis (fatty liver) progressing to inflammation and culminating in cirrhosis. Multiple factors contribute to ALD progression and disease severity. Here, we overview several crucial mechanisms related to ALD end-stage outcome development, such as epigenetic changes, cell death, hemolysis, hepatic stellate cells activation, and hepatic fatty acid binding protein 4. Additionally, in this review, we also present two clinically relevant models using human precision-cut liver slices and hepatic organoids to examine ALD pathogenesis and progression.
Collapse
Affiliation(s)
- Natalia A. Osna
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | - Irina Tikhanovich
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA;
| | - Martí Ortega-Ribera
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (M.O.-R.); (G.S.)
| | - Sebastian Mueller
- Center for Alcohol Research, University of Heidelberg, 69120 Heidelberg, Germany; (S.M.); (C.Z.); (J.M.); (S.L.)
- Viscera AG Bauchmedizin, 83011 Bern, Switzerland
| | - Chaowen Zheng
- Center for Alcohol Research, University of Heidelberg, 69120 Heidelberg, Germany; (S.M.); (C.Z.); (J.M.); (S.L.)
| | - Johannes Mueller
- Center for Alcohol Research, University of Heidelberg, 69120 Heidelberg, Germany; (S.M.); (C.Z.); (J.M.); (S.L.)
| | - Siyuan Li
- Center for Alcohol Research, University of Heidelberg, 69120 Heidelberg, Germany; (S.M.); (C.Z.); (J.M.); (S.L.)
| | - Sadatsugu Sakane
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Raquel Carvalho Gontijo Weber
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Hyun Young Kim
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Wonseok Lee
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Souradipta Ganguly
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Yusuke Kimura
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Xiao Liu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Debanjan Dhar
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
| | - Karin Diggle
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - David A. Brenner
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; (S.S.); (R.C.G.W.); (H.Y.K.); (W.L.); (S.G.); (Y.K.); (X.L.); (D.D.); (K.D.); (D.A.B.)
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California San Diego, La Jolla, CA 92093, USA;
| | - Neha Attal
- Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC 28203, USA; (N.A.); (I.H.M.)
| | - Iain H. McKillop
- Department of Surgery, Atrium Health Carolinas Medical Center, Charlotte, NC 28203, USA; (N.A.); (I.H.M.)
| | - Shilpa Chokshi
- The Roger Williams Institute of Hepatology, Foundation for Liver Research, London SE59NT, UK;
- School of Microbial Sciences, King’s College, London SE59NT, UK
| | - Ram Mahato
- Department of Pharmaceutical Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68106, USA;
| | - Karuna Rasineni
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68106, USA;
| | - Gyongyi Szabo
- Department of Medicine, Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; (M.O.-R.); (G.S.)
| | - Kusum K. Kharbanda
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68106, USA
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68106, USA;
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE 68105, USA
| |
Collapse
|
50
|
Zhou XN, Zhang Q, Peng H, Qin YJ, Liu YH, Wang L, Cheng ML, Luo XH, Li H. Silent information regulator sirtuin 1 ameliorates acute liver failure via the p53/glutathione peroxidase 4/gasdermin D axis. World J Gastroenterol 2024; 30:1588-1608. [PMID: 38617450 PMCID: PMC11008418 DOI: 10.3748/wjg.v30.i11.1588] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 12/20/2023] [Accepted: 02/18/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Acute liver failure (ALF) has a high mortality with widespread hepatocyte death involving ferroptosis and pyroptosis. The silent information regulator sirtuin 1 (SIRT1)-mediated deacetylation affects multiple biological processes, including cellular senescence, apoptosis, sugar and lipid metabolism, oxidative stress, and inflammation. AIM To investigate the association between ferroptosis and pyroptosis and the upstream regulatory mechanisms. METHODS This study included 30 patients with ALF and 30 healthy individuals who underwent serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) testing. C57BL/6 mice were also intraperitoneally pretreated with SIRT1, p53, or glutathione peroxidase 4 (GPX4) inducers and inhibitors and injected with lipopolysaccharide (LPS)/D-galactosamine (D-GalN) to induce ALF. Gasdermin D (GSDMD)-/- mice were used as an experimental group. Histological changes in liver tissue were monitored by hematoxylin and eosin staining. ALT, AST, glutathione, reactive oxygen species, and iron levels were measured using commercial kits. Ferroptosis- and pyroptosis-related protein and mRNA expression was detected by western blot and quantitative real-time polymerase chain reaction. SIRT1, p53, and GSDMD were assessed by immunofluorescence analysis. RESULTS Serum AST and ALT levels were elevated in patients with ALF. SIRT1, solute carrier family 7a member 11 (SLC7A11), and GPX4 protein expression was decreased and acetylated p5, p53, GSDMD, and acyl-CoA synthetase long-chain family member 4 (ACSL4) protein levels were elevated in human ALF liver tissue. In the p53 and ferroptosis inhibitor-treated and GSDMD-/- groups, serum interleukin (IL)-1β, tumour necrosis factor alpha, IL-6, IL-2 and C-C motif ligand 2 levels were decreased and hepatic impairment was mitigated. In mice with GSDMD knockout, p53 was reduced, GPX4 was increased, and ferroptotic events (depletion of SLC7A11, elevation of ACSL4, and iron accumulation) were detected. In vitro, knockdown of p53 and overexpression of GPX4 reduced AST and ALT levels, the cytostatic rate, and GSDMD expression, restoring SLC7A11 depletion. Moreover, SIRT1 agonist and overexpression of SIRT1 alleviated acute liver injury and decreased iron deposition compared with results in the model group, accompanied by reduced p53, GSDMD, and ACSL4, and increased SLC7A11 and GPX4. Inactivation of SIRT1 exacerbated ferroptotic and pyroptotic cell death and aggravated liver injury in LPS/D-GalN-induced in vitro and in vivo models. CONCLUSION SIRT1 activation attenuates LPS/D-GalN-induced ferroptosis and pyroptosis by inhibiting the p53/GPX4/GSDMD signaling pathway in ALF.
Collapse
Affiliation(s)
- Xing-Nian Zhou
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Quan Zhang
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Hong Peng
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang 550001, Guizhou Province, China
| | - Yu-Jie Qin
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Yu-Hong Liu
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Lu Wang
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Ming-Liang Cheng
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
| | - Xin-Hua Luo
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang 550001, Guizhou Province, China
| | - Hong Li
- Department of Infectious Diseases, The Affiliated Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang 550004, Guizhou Province, China
- Department of Infectious Diseases, Guizhou Provincial People's Hospital, Guiyang 550001, Guizhou Province, China
| |
Collapse
|