1
|
Chen R, Chai X, Zhang Y, Zhou T, Xia Y, Jiang X, Lv B, Zhang J, Zhou L, Tian X, Wang R, Mao L, Zhao F, Zhang H, Hu J, Qiu J, Zou Z, Chen C. Novel role of FTO in regulation of gut-brain communication via Desulfovibrio fairfieldensis-produced hydrogen sulfide under arsenic exposure. Gut Microbes 2025; 17:2438471. [PMID: 39852343 PMCID: PMC11776478 DOI: 10.1080/19490976.2024.2438471] [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: 12/21/2023] [Revised: 10/26/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025] Open
Abstract
Fat mass and obesity-associated protein (FTO) is the key demethylase that reverses the abnormally altered N6-methyladenosine (m6A) modification in eukaryotic cells under environmental pollutants exposure. Arsenic is an environmental metalloid and can cause severe symptoms in human mainly through drinking water. However, there is no specific treatment for its toxic effects due to the uncovered mechanisms. We previously revealed that exposure to arsenic increased the level of m6A via down-regulation of FTO, which might serve as a potential target for intervention against arsenic-related disorders. In this study, our results demonstrated that chronic exposure to arsenic significantly disrupted the intestinal barrier and microenvironment. Also, this administration resulted in the enhancement of m6A modification and the reduction of FTO expression in the intestine. By using both CRISPR/Cas9-based FTO knock-in strategy and adeno-associated virus (AAV)-mediated overexpression of FTO in the intestine, we established for the first time that up-regulation of FTO remarkably ameliorated arsenic-induced disruption of intestinal barriers and altered microenvironment of mice. We also firstly identified a dominant gut microbial species, Desulfovibrio fairfieldensis, which was sharply reduced in arsenic-exposed mice, was able to proceed arsenic-induced neurobehavioral impairments by declining the levels of its major metabolite hydrogen sulfide. Administration of Desulfovibrio fairfieldensis could significantly alleviate the neurotoxicity of arsenic. Intriguingly, the beneficial effects of FTO against arsenic neurotoxicity possibly occurred through a novel gut-brain communication via Desulfovibrio fairfieldensis and its produced hydrogen sulfide. Collectively, these findings will provide new ideas for understanding the mechanisms of arsenic-induced toxic effects from a gut-brain communication perspective, and will assist the development of explicit intervention strategy via regulation of a new potential target FTO for prevention and treatment against arsenic-related both intestinal and neurological disorders.
Collapse
Affiliation(s)
- Ruonan Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xiaoqin Chai
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yunxiao Zhang
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Tianxiu Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yinyin Xia
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xuejun Jiang
- Center of Experimental Teaching for Public Health, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Bo Lv
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Zhang
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lixiao Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Xin Tian
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ruonan Wang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lejiao Mao
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Feng Zhao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Hongyang Zhang
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Hu
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing, People’s Republic of China
| | - Jingfu Qiu
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Zhen Zou
- Molecular Biology Laboratory of Respiratory Disease, Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
- Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
2
|
Zhang C, Chen S, Wang Z, Zhang J, Yu W, Wang Y, Si W, Zhang Y, Zhang Y, Liang T. Exploring the mechanism of intestinal bacterial translocation after severe acute pancreatitis: the role of Toll-like receptor 5. Gut Microbes 2025; 17:2489768. [PMID: 40243695 PMCID: PMC11980482 DOI: 10.1080/19490976.2025.2489768] [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: 01/04/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Severe acute pancreatitis (SAP)-induced intestinal bacterial translocation and enterogenic infection are among the leading causes of mortality in patients. However, the mechanisms by which SAP disrupted the intestinal barrier and led to bacterial translocation remained unclear. Therefore, we employed multi-omics analysis including microbiome, metabolome, epigenome, transcriptome, and mass cytometry (CyTOF) to identify potential targets, followed by functional validation using transgenic mice. The integrated multi-omics analysis primarily indicated overgrowth of intestinal flagellated bacteria, upregulation of intestinal Toll-like receptor 5 (TLR5) and acute inflammatory response, and increased infiltration of intestinal high-expressing TLR5 lamina propria dendritic cells (TLR5hi LPDC) after SAP. Subsequently, intestinal flagellin-TLR5 signaling was activated after SAP. Intestinal barrier disruption, bacterial translocation, and helper T cells (Th) differentiation imbalance caused by SAP were alleviated in TLR5 knocked out (Tlr5-/-) or conditionally knocked out on LPDC (Tlr5ΔDC) mice. However, TLR5 conditional knockout on intestinal epithelial cells (Tlr5ΔIEC) failed to improve SAP-induced bacterial translocation. Moreover, depletion of LPDC and regulatory T cells (Treg) ameliorated bacterial translocation after SAP. Our findings identify TLR5 on LPDC as a potential novel target for preventing or treating intestinal bacterial translocation caused by SAP.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Shiyin Chen
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Zhien Wang
- Department of Rehabilitation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Jian Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Wenqiao Yu
- Department of Surgical Intensive Care Unit, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yanshuai Wang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Weiwei Si
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| | - Yuwei Zhang
- College of Science, Mathematics and Technology, Wenzhou-Kean University, Wenzhou, Zhejiang Province, China
| | - Yun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- Department of Nutrition, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, Zhejiang Province, China
- Innovation Center for the Study of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, Zhejiang Province, China
| |
Collapse
|
3
|
Du P, Zhang T, Wu Y, Yu Z, Liu C. Monitoring variations in mitochondrial hydrogen sulfide using two-photon cyclometalated iridium(III) complex probe: A new strategy for ischemia-reperfusion drug discovery and efficacy evaluation. Talanta 2025; 292:128021. [PMID: 40157194 DOI: 10.1016/j.talanta.2025.128021] [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: 02/05/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is one of the main causes of liver insufficiency and failure after liver surgery. However, the effectiveness of current methods of treating HIRI is generally limited. Previous studies have shown that hydrogen sulfide (H2S) has a beneficial effect on HIRI, and an appropriate concentration of H2S can significantly reduce HIRI by protecting the mitochondria. Therefore, establishing an accurate imaging platform for monitoring variations in mitochondrial H2S is an effective strategy for anti-HIRI drug discovery and efficacy evaluation. To this end, a cyclometalated iridium(III) complex-based probe, Cym-Ir-EDB, was developed for detecting mitochondrial H2S in HIRI. Cym-Ir-EDB possesses good sensitivity, high selectivity, negligible cytotoxicity, and excellent mitochondrial-targeting ability, rendering it a promising imaging tool for analyzing variations in mitochondrial H2S in HIRI cells. Using Cym-Ir-EDB as a probe, anti-HIRI drugs were screened from isothiocyanates by monitoring variations in mitochondrial H2S in HIRI cells, for the first time. Moreover, the dynamics of mitochondrial H2S in HIRI cells were visualized and the response of HIRI to treatment with the screened erucin was monitored. The findings indicate that Cym-Ir-EDB can serve as a useful imaging platform for the precise imaging of mitochondrial H2S in HIRI, thereby contributing to anti-HIRI drug discovery and efficacy evaluation.
Collapse
Affiliation(s)
- Peng Du
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266071, China
| | - Tianao Zhang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266071, China
| | - Ying Wu
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China; Education and Training Department, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| | - Zongping Yu
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Chaolong Liu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
4
|
Alanazi M, Al-Kuraishy HM, Albuhadily AK, Al-Gareeb AI, Abdelaziz AM, Alexiou A, Papadakis M, Batiha GES. The protective effect of amylin in type 2 diabetes: Yes or no. Eur J Pharmacol 2025; 996:177593. [PMID: 40187597 DOI: 10.1016/j.ejphar.2025.177593] [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: 12/23/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
Amylin, which is also called a human islet amyloid polypeptide, is a peptide hormone made up of 37 amino acids that is released from pancreatic β cells. It helps keep blood sugar levels stable by controlling the release of insulin and glucagon. Various studies have indicated its involvement in the pathogenesis of type 2 diabetes (T2D) through the induction of apoptosis in pancreatic cells. Conversely, other studies found that amylin plays a critical role in the pathogenesis of T2D by affecting the release of insulin and glucagon. Therefore, amylin has protective and detrimental effects on the pathogenesis of T2D. Consequently, this review aims to discuss the beneficial and detrimental roles of amylin in T2D.
Collapse
Affiliation(s)
- Mansour Alanazi
- Department of Internal Medicine, College of Medicine, Northern Border University, Arar, Saudi Arabia.
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, 14132, Baghdad, Iraq.
| | - Ali K Albuhadily
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, 14132, Baghdad, Iraq.
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, 14132, Baghdad, Iraq; Jabir ibn Hayyan Medical University, Al-Ameer Qu, PO. Box13 Kufa, Najaf, Iraq.
| | - Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University-Arish Branch, Arish, 45511, Egypt.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India; Department of Research & Development, Funogen, Athens, 11741, Greece.
| | - Marios Papadakis
- University Hospital Witten-Herdecke, University of Witten, Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt.
| |
Collapse
|
5
|
Yenice G, Ozkanlar S, Bolat I, Yildirim S. The Potential Ability of Betulinic Acid to Prevent Experimentally Induced Acute Pancreatitis in Rats. Basic Clin Pharmacol Toxicol 2025; 136:e70052. [PMID: 40344370 DOI: 10.1111/bcpt.70052] [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: 06/10/2024] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Acute pancreatitis (AP) is a serious pancreatic inflammatory disease that results in pancreatic enzyme activation and autodegradation. Betulinic acid (BA), a pentacyclic triterpene of natural origin that was isolated from several plants, has anti-inflammatory, immunomodulatory and antioxidant effects that can help with AP. With this study, we aimed to investigate the potential positive effects of BA on l-arginine-induced AP. A total of 24 male rats were divided into four groups (control, BA, AP and BA + AP). Animals in the BA group were given BA 50 mg/kg/day for 7 days. AP was induced by administering two doses of 250-mg/100-g l-arginine to animals in the AP group. The animals in the BA + AP group were administered 50-mg/kg/day BA (gavage) for 7 days and two doses of 250-mg/100-g l-arginine on the seventh day. BA pretreatment inhibited the increased lipase activity caused by AP and showed protective activity against oxidative damage to pancreatic tissue. It decreased the severity of inflammation by suppressing the release of pro-inflammatory cytokines while increasing the level of the anti-inflammatory cytokine IL-10. It showed a protective effect on pancreatic tissue by inhibiting tumour necrosis factor (TNF-α) and Bax expression. The findings of the study show that BA exhibits multifaceted protective activity in experimental AP induced with l-arginine.
Collapse
Affiliation(s)
- Guler Yenice
- Faculty of Veterinary Medicine, Department of Animal Nutrition and Nutritional Disorders, Ataturk University, Erzurum, Turkey
| | - Seckin Ozkanlar
- Faculty of Veterinary Medicine, Department of Biochemistry, Ataturk University, Erzurum, Turkey
| | - Ismail Bolat
- Faculty of Veterinary Medicine, Department of Pathology, Ataturk University, Erzurum, Turkey
| | - Serkan Yildirim
- Faculty of Veterinary Medicine, Department of Pathology, Ataturk University, Erzurum, Turkey
| |
Collapse
|
6
|
Ralston MR, McCreath G, Lees ZJ, Salt IP, Sim MA, Watson MJ, Freeman DJ. Beyond body mass index: exploring the role of visceral adipose tissue in intensive care unit outcomes. BJA OPEN 2025; 14:100391. [PMID: 40223920 PMCID: PMC11986990 DOI: 10.1016/j.bjao.2025.100391] [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: 06/20/2024] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Obesity is a worldwide health crisis and poses significant challenges in critical care. Many studies suggest an 'obesity paradox', in which obesity, defined by body mass index (BMI), is associated with better outcomes. However, the inability of BMI to discriminate between fat and muscle or between visceral adipose tissue and subcutaneous adipose tissue, limits its prediction of metabolic ill health. We suggest that the 'obesity paradox' may be more reflective of the limitations of BMI than the protective effect of obesity. We explore the biological processes leading to visceral fat accumulation, and the evidence linking it to outcomes in critical illness. In the 'spillover' hypothesis of adipose tissue expansion, caloric excess and impaired expansion of storage capacity in the subcutaneous adipose tissue lead to accumulation of visceral adipose tissue. This is associated with a chronic inflammatory state, which is integral to the link between visceral adiposity, type 2 diabetes mellitus, and ischaemic heart disease. We review the current evidence on visceral adiposity and critical illness outcomes. In COVID-19, increased visceral adipose tissue, irrespective of BMI, is associated with more severe disease. This is mirrored in acute pancreatitis, suggesting visceral adiposity is linked to poorer outcomes in some hyperinflammatory conditions. We suggest that visceral adiposity's chronic inflammatory state may potentiate acute inflammation in conditions such as COVID-19 and acute pancreatitis. Further work is required to investigate other critical illnesses, especially sepsis and acute respiratory distress syndrome, in which current evidence is scarce. This may give further insights into pathophysiology and inform tailored treatment and nutrition strategies based on body fat distribution.
Collapse
Affiliation(s)
- Maximilian R. Ralston
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
- Academic Unit of Anaesthesia, Critical Care & Perioperative Medicine, University of Glasgow, Glasgow, UK
| | - Gordan McCreath
- Academic Unit of Anaesthesia, Critical Care & Perioperative Medicine, University of Glasgow, Glasgow, UK
| | - Zoe J. Lees
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| | - Ian P. Salt
- School of Molecular Biosciences, University of Glasgow, Glasgow, UK
| | - Malcolm A.B. Sim
- Academic Unit of Anaesthesia, Critical Care & Perioperative Medicine, University of Glasgow, Glasgow, UK
- Department of Critical Care, Queen Elizabeth University Hospital, Glasgow, UK
| | - Malcolm J. Watson
- School of Medicine, Dentistry & Nursing, University of Glasgow, Glasgow, UK
- Department of Anaesthesia, Queen Elizabeth University Hospital, Glasgow, UK
| | - Dilys J. Freeman
- School of Cardiovascular & Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
7
|
Liu P, Hu P, Jin M, Sun W, Wu J, Tang Y, Shi D, Xie T, Tong Y, Huang L, Zhang D, Zheng H, Xu X, He H. Compound probiotics alleviate hyperuricemia-induced renal injury via restoring gut microbiota and metabolism. BMC Microbiol 2025; 25:280. [PMID: 40335932 PMCID: PMC12060558 DOI: 10.1186/s12866-025-04012-5] [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: 02/01/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
To investigate the role and mechanisms of gut microbiota in hyperuricemia-induced renal injury, we established renal failure models using unilateral nephrectomized mice. After four weeks of adenine and potassium oxalate-supplemented diet, probiotic intervention was administered. Renal pathological and functional changes were assessed through H&E staining and plasma biochemical analysis. Gut microbiota composition and metabolite profiles were evaluated using 16 S rRNA gene sequencing and non-targeted metabolomics of fecal samples.Our findings demonstrate that the compound probioticS effectively attenuated hyperuricemia-associated renal dysfunction and interstitial fibrosis. The intervention reduced oxidative stress, mitophagy, and apoptosis in renal tubules. Probiotic treatment enhanced gut microbiota diversity, notably increasing the abundance of Prevotella_9, Dorea, and unclassified Bacteroidota, while decreasing unclassified Desulfovibrio. KEGG enrichment analysis revealed that probiotic intervention upregulated arginine and proline metabolism, as well as tyrosine metabolism in feces. Furthermore, it enhanced the metabolism of arginine, proline, valine, leucine, and isoleucine in plasma.Notably, sulfocholic acid and urocanic acid showed negative correlations with oxidative stress markers, autophagy, and apoptosis indicators. Similarly, plasma L-proline levels were inversely correlated with these pathological parameters.These results suggest that the compound probiotics may mitigate hyperuricemia-induced kidney damage through restoration of gut microbiota homeostasis and preservation of plasma and fecal metabolites. The protective mechanisms likely involve attenuation of hyperuricemia-associated oxidative stress, mitochondrial dysregulation, and phagocytosis-induced apoptosis.Our study provides compelling evidence that probiotic supplementation represents a promising therapeutic strategy for hyperuricemia-induced renal injury, potentially through modulation of gut microbiota and associated metabolic pathways.
Collapse
Affiliation(s)
- Ping Liu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Ping Hu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Meiping Jin
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Weiqian Sun
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Jiajun Wu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Yuyan Tang
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Danye Shi
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Ting Xie
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Yijing Tong
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Lusheng Huang
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Dongliang Zhang
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China
| | - Hui Zheng
- Division of Endocrinology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| | - Xudong Xu
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| | - Haidong He
- Division of Nephrology, Minhang Hospital, Fudan University, No. 170 Xinsong Road, Shanghai, 201199, China.
| |
Collapse
|
8
|
Mohsin M, Zaki A, Tabassum G, Khan S, Ali S, Ahmad T, Syed MA. Urolithin-A supplementation alleviates sepsis-induced acute lung injury by reducing mitochondrial dysfunction and modulating macrophage polarization. Mitochondrion 2025; 84:102047. [PMID: 40328344 DOI: 10.1016/j.mito.2025.102047] [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: 12/23/2024] [Revised: 04/24/2025] [Accepted: 05/01/2025] [Indexed: 05/08/2025]
Abstract
Sepsis is a severe and life-threatening condition marked by excessive inflammation, mitochondrial dysfunction, and epithelial barrier disruption, often leading to Acute Lung Injury (ALI). Mitophagy, a cellular mechanism that removes damaged mitochondria, plays a vital role in maintaining mitochondrial health during sepsis. In this study, we investigated the protective effects of Urolithin-A against ALI and sepsis. In LPS-stimulated RAW264.7 macrophages, Urolithin-A significantly reduced mitochondrial dysfunction, Reactive Oxygen Species (ROS), Nitric Oxide (NO) production, and apoptosis. Additionally, it enhanced mitophagy by upregulating PINK1, Parkin, and LC3-II, which helped preserve mitochondrial function. In vivo, Urolithin-A treatment in mouse models of ALI and sepsis reduced lung injury and inflammation, as shown by improved ALI scores, decreased wet/dry lung weight ratios, and lower levels of inflammatory markers such as iNOS, IL-1β, and MPO. Urolithin-A also improved epithelial barrier integrity and upregulated anti-apoptotic markers, demonstrating its ability to alleviate sepsis-induced lung damage. These findings suggest that Urolithin-A holds significant promise as a therapeutic agent for managing inflammatory lung conditions associated with sepsis.
Collapse
Affiliation(s)
- Mohd Mohsin
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Almaz Zaki
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Gulnaz Tabassum
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Salman Khan
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi, India.
| | - Shakir Ali
- Department of Biochemistry, School of Chemical and Life Sciences, Jamia Hamdard University, New Delhi, India.
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advance Research and Studies, Jamia Millia Islamia, New Delhi, India.
| | - Mansoor Ali Syed
- Department of Biotechnology, Faculty of Life Sciences, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
9
|
Almutary KH, Zaghloul MS, Nader MA, Elsheakh AR. Mechanistic insights into the protective potential of ambrisentan against L-arginine induced acute pancreatitis and multiorgan damage (role of NRF2/HO-1 and TXNIP/NLRP3 pathways). Biomed Pharmacother 2025; 187:118119. [PMID: 40319659 DOI: 10.1016/j.biopha.2025.118119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025] Open
Abstract
Acute pancreatitis (AP) is an abrupt inflammation of the pancreatic tissue. The severity of AP varies from mild and self-limiting to severe, potentially fatal, and can affect several organ systems. The most severe type of AP causes multiple organ damage (MOD) due to systemic inflammation. In this study, ambrisentan (AMB), an endothelin A receptor antagonist (ETA), was investigated for its potential to ameliorate L-arginine (L-Arg) induced AP and MOD in rats. AP was induced using L-Arg (100 mg/100 g). Two doses of AMB were tested and compared to N-acetylcystiene (NAC) effect. AMB restored the normal structure of the pancreatic, hepatic, pulmonary, and renal tissues. In addition, it normalized the levels of pancreatic enzymes, lactate dehydrogenase (LDH), serum liver enzymes, and kidney biomarkers. Furthermore, AMB corrected the imbalance in the levels of oxidants/antioxidants caused by L-Arg. In contrast, AMB (5 mg/kg) significantly upregulated the protein levels of adenosine monophosphate protein kinase (AMPK), nuclear factor erythroid 2-related factor 2 (NRF2), heme oxidase-1(HO-1) and thioredoxin reductase 1 (TXNRD1) by approximately 69.59 %, 85.14 %, 688 % and 96 % respectively, compared with those in rats treated with L-Arg. Furthermore, AMB (5 mg/kg) significantly lowered the thioredoxin-interacting protein (TXNIP), nod-like Receptor Protein 3 (NLRP3), glycogen synthase kinase-3β (GSK-3β), inducible nitric oxide synthase (iNOS), tumor necrosis factor-α (TNF-α), CD68, autophagic markers (P62 and LC3) and apoptotic marker caspase 3 by around 62.43 %, 73.56 %, 62.5 %,70 %, 80.3 %, 93 %, 96.7 %, 95 %, 39.6 % respectively, compared to the group treated with L-Arg. AMB effectively improved the AP and MOD produced by L-Arg through its anti-inflammatory and antioxidant properties. NRF2/HO-1 and TXNIP/NLRP3 pathways play major roles in these protective effects.
Collapse
Affiliation(s)
- Khaled H Almutary
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Majmaah University, P.O.Box 66, Majmaah 11952, Saudi Arabia
| | - Marwa S Zaghloul
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa 7731168, Egypt.
| | - Manar A Nader
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa 7731168, Egypt
| | - Ahmed R Elsheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura National University, Gamasa 7731168, Egypt; Future Studies and Risks Management & National Committee of Drugs, Academy of Scientific Research, Ministry of Higher Education, Elsayeda Zeinab, Egypt
| |
Collapse
|
10
|
Lim EY, Kim GD, Kim HJ, Eom JE, Song HJ, Shin DU, Kim YI, Kim HJ, Lee SY, Shin HS. Cirsium japonicum leaf extract attenuated lipopolysaccharide-induced acute respiratory distress syndrome in mice via suppression of the NLRP3 and HIF1α pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 140:156601. [PMID: 40064116 DOI: 10.1016/j.phymed.2025.156601] [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: 09/10/2024] [Revised: 12/20/2024] [Accepted: 03/01/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Acute respiratory distress syndrome (ARDS) is a severe inflammatory disorder characterized by acute respiratory failure, alveolar barrier dysfunction, edema, and dysregulated alveolar macrophage-mediated pulmonary inflammation. Despite advancements in treatment strategies, the mortality rate in patients with ARDS remains high, ranging from 40-60 %. Current approaches are limited to supportive care, necessitating the exploration of effective therapeutic options such as suppressing broad inflammatory responses. Although Cirsium japonicum leaves possess anti-inflammatory properties, their specific effects on ARDS have not yet been investigated. METHODS The anti-inflammatory activity of Cirsium japonicum extract (CJE) was investigated in a lipopolysaccharide (LPS)-induced ARDS model. RESULTS CJE significantly attenuated LPS-induced lung injury, including reduced alveolar wall thickness, inflammatory cell infiltration, proteinaceous debris, and hyaline membranes. Moreover, CJE repressed infiltration of inflammatory cells and pro-inflammatory gene expression in bronchoalveolar lavage fluid. Concordantly, CJE mitigated alveolar macrophage activation, which consequently reduced neutrophil chemoattractic infiltration. Additionally, CJE suppressed NLRP3 and HIF1α expression in the lungs of the ARDS mouse. Similarly, LPS-induced NLRP3 and HIF1α pathway-associated inflammatory and glycolytic gene expressions significantly diminished by CJE in murine alveolar macrophage cell line, MH-S cells, and bone marrow-derived macrophages. CONCLUSION CJE suppressed multiple inflammatory responses through the regulation of NLRP3 and HIF1α signaling-related gene expression in macrophages of LPS-induced ARDS mice. These results suggest that CJE has therapeutic potential for treating patients with ARDS via macrophage regulation.
Collapse
Affiliation(s)
- Eun Yeong Lim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Gun-Dong Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Ha-Jung Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Ji-Eun Eom
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Hyeon-Ji Song
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Science and Technology, Jeonbuk National University, Jeonju 54896, South Korea
| | - Dong-Uk Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Young In Kim
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea
| | - Hyun-Jin Kim
- Department of Food Science and Technology, Gyeongsang National University, Jinju 52828, South Korea
| | - So-Young Lee
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, South Korea
| | - Hee Soon Shin
- Division of Food Functionality Research, Korea Food Research Institute (KFRI), Wanju, 55365, South Korea; Department of Food Biotechnology, Korea University of Science and Technology (UST), Daejeon 34113, South Korea.
| |
Collapse
|
11
|
Huang Q, Wang Y, Zhang Z, Wu M, Liu J, Chen J, Li J, Yao Y, Guo C, Zhao D, Qi W, Li X. Organ dysfunction induced by hemorrhagic shock: From mechanisms to therapeutic medicines. Pharmacol Res 2025; 216:107755. [PMID: 40315969 DOI: 10.1016/j.phrs.2025.107755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/03/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Hemorrhagic shock (HS) leads to organ dysfunction, which increases the incidence of unfavorable outcomes in patients. However, adjuvant drug therapy for HS has not been widely accepted, and the benefits of vasopressors are generally considered to have insufficient evidence. Energy homeostasis disruption and excessive immune system activation are the main mechanisms underlying HS-induced organ dysfunction. Recent reports on HS animal models and clinical trials have revealed potential drugs that target the immune response, oxidative damage, and energy homeostasis in HS, providing new insights for the treatment of HS-induced organ dysfunction. In this review, we first discuss the pathophysiology of organ dysfunction involved in HS injury and then systematically review potential drugs that regulate immunity, the inflammatory response, oxidative damage, energy homeostasis, and cell death. We also review the available drugs with clinical evidence of HS-induced organ dysfunction efficacy. Treatment strategies combined with an improved understanding of the organ injury mechanisms of HS may help identify and develop targeted therapeutic modalities that mitigate severe organ dysfunction and reduce mortality caused by HS injury.
Collapse
Affiliation(s)
- Qingxia Huang
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Yisa Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Zepeng Zhang
- Research Center of Traditional Chinese Medicine, College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130021, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Mingxia Wu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Jiaqi Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Jinjin Chen
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Jing Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Yao Yao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Chen Guo
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Wenxiu Qi
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China.
| | - Xiangyan Li
- Northeast Asia Research Institute of Traditional Chinese Medicine, Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Jilin Provincial Key Laboratory of Bio-Macromolecules of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China.
| |
Collapse
|
12
|
Bronowicka-Adamska P, Szlęzak D, Bentke-Imiolek A, Kaszuba K, Majewska-Szczepanik M. The modulation of low molecular weight sulfur compounds levels in visceral adipose tissue of TLR2-deficient mice on a high-fat diet. Biochimie 2025; 232:66-73. [PMID: 39870157 DOI: 10.1016/j.biochi.2025.01.008] [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: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025]
Abstract
Obesity treatment requires an individualized approach, emphasizing the need to identify metabolic pathways of diagnostic relevance. Toll-like receptors (TLRs), particularly TLR2 and TLR4, play a crucial role in metabolic disorders, as receptor deficiencies improves insulin sensitivity and reduces obesity-related inflammation. Additionally, hydrogen sulfide (H2S) influences lipolysis, adipogenesis, and adipose tissue browning through persulfidation. This study investigates the impact of a high-fat diet (HFD) on low molecular weight sulfur compounds in the visceral adipose tissue (VAT) of C57BL/6 and TLR2-deficient mice. It focuses on key enzymes involved in H2S metabolism: cystathionine beta-synthase (CBS), cystathionine gamma-lyase (CGL), 3-mercaptopyruvate sulfurtransferase (MPST), and thiosulfate sulfurtransferase (TST). In C57BL/6 mice on HFD, MPST activity decreased, while CBS level increased, potentially compensating for H2S production. In contrast, TLR2-deficient mice on HFD exhibited higher MPST activity but reduced level of CBS and CGL activity, suggesting that TLR2 deficiency mitigates HFD-induced changes in sulfur metabolism. TST activity was lower in TLR2-deficient mice, indicating an independent regulatory role of TLR2 in TST activity. Elevated oxidative stress, reflected by increased glutathione levels, was observed in wild-type mice. Interestingly, cysteine and cystine were detectable only in the VAT of the C57BL/6 ND group and were absent in all other groups. The capacity for hydrogen sulfide production in tissues from TLR2-/-B6 HFD group was significantly lower than in the C57BL/6 HFD group. In conclusion, TLR2 modulates sulfur metabolism, oxidative stress, and inflammation in obesity. TLR2 deficiency disrupts H2S production and redox balance, potentially contributing to metabolic dysfunction, highlighting TLR2 as a potential therapeutic target for obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Patrycja Bronowicka-Adamska
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, 7c Kopernika st., 31-034, Cracow, Poland.
| | - Dominika Szlęzak
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, 7c Kopernika st., 31-034, Cracow, Poland
| | - Anna Bentke-Imiolek
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, 7c Kopernika st., 31-034, Cracow, Poland
| | - Kinga Kaszuba
- Jagiellonian University Medical College, Faculty of Medicine, Chair of Medical Biochemistry, 7c Kopernika st., 31-034, Cracow, Poland
| | - Monika Majewska-Szczepanik
- Jagiellonian University Medical College, Faculty of Health Sciences, Department of Medical Physiology, Chair of Biomedical Sciences, 12 Michalowskiego st., 33-332, Cracow, Poland
| |
Collapse
|
13
|
Bruschi M, Masini S, Biancucci F, Piersanti G, Canonico B, Menotta M, Magnani M, Fraternale A. Redox modulation via a synthetic thiol compound reshapes energy metabolism in endothelial cells and ameliorates angiogenic expression in a co-culture study with activated macrophages. Biochim Biophys Acta Gen Subj 2025; 1869:130803. [PMID: 40187375 DOI: 10.1016/j.bbagen.2025.130803] [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: 12/19/2024] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
The vascular endothelium is the first interface exposed to circulating compounds and oxidative as well as pro-inflammatory stimuli. Nowadays, cysteine pro-drugs are emerging as new and potential therapies in cardiovascular and inflammatory diseases due to their cytoprotective effects. In this study, the effects of redox modulation by a synthetic thiol compound, i.e., I-152, a precursor of N-acetylcysteine (NAC) and cysteamine (MEA), were evaluated after 6 h and 24 h treatment on human umbilical cord endothelial cell (HUVECs) energy metabolism. Following I-152 treatment, higher cysteine and glutathione (GSH) content were detected via HPLC, in concomitance with I-152 derivatives, i.e., NAC and MEA. Untargeted metabolomics confirmed GSH upregulation and NAC presence in addition to I-152 itself and other metabolites, such as dithiol compound (NACMEAA) and triacetylated I-152. Mass spectrometry revealed that I-152 boosted ATP production, specifically through the mitochondrial OXPHOS, as determined via Seahorse assay without inducing oxidative stress. Additionally, I-152 treatment of HUVECs before co-culture with LPS-stimulated macrophages provided GSH and cysteine sustainment and attenuated the transcription of adhesion molecules as well as iNOS expression. Identifying the impact of redox regulation in physiological conditions and the possible metabolic targets could aid the application of novel thiol-based therapeutics.
Collapse
Affiliation(s)
- Michela Bruschi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Sofia Masini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Federica Biancucci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Barbara Canonico
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Michele Menotta
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| | - Alessandra Fraternale
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, PU, Italy.
| |
Collapse
|
14
|
Ma J, Yang P, Zhou Z, Song T, Jia L, Ye X, Yan W, Sun J, Ye T, Zhu L. GYY4137-induced p65 sulfhydration protects synovial macrophages against pyroptosis by improving mitochondrial function in osteoarthritis development. J Adv Res 2025; 71:173-188. [PMID: 38844123 DOI: 10.1016/j.jare.2024.05.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
INTRODUCTION Osteoarthritis (OA) is the most common arthritis that is characterized by the progressive synovial inflammation and loss of articular cartilage. Although GYY4137 is a novel and slow-releasing hydrogen sulfide (H2S) donor with potent anti-inflammatory properties that may modulate the progression of OA, its underlying mechanism remains unclear. OBJECTIVES In this study, we validated the protective role of GYY4137 against OA pathological courses and elucidated its underlying regulatory mechanisms. METHODS Cell transfection, immunofluorescence staining, EdU assay, transmission electron microscopy, mitochondrial membrane potential measurement, electrophoretic mobility shift assay, sulfhydration assay, qPCR and western blot assays were performed in the primary mouse chondrocytes or the mouse macrophage cell line raw 264.7 for in vitro study. DMM-induced OA mice model and Macrophage-specific p65 knockout (p65f/f LysM-CreERT2) mice on the C57BL/6 background were used for in vivo study. RESULTS We found that GYY4137 can alleviate OA progress by suppressing synovium pyroptosis in vivo. Moreover, our in vitro data revealed that GYY4137 attenuates inflammation-induced NLRP3 and caspase-1 activation and results in a decrease of IL-1β production in macrophages. Mechanistically, GYY4137 increased persulfidation of NF-kB p65 in response to inflammatory stimuli that results in a decrease of cellular reactive oxygen species (ROS) accumulation and ameliorates mitochondrial dysfunctions. Using site-directed mutagenesis, we showed that H2S persulfidates cysteine38 in p65 protein and hampers p65 transcriptional activity, and p65 mutant impaired macrophage responses to GYY4137. CONCLUSION These findings suggest a mechanism by which GYY4137 through redox modification of p65 participates in inhibiting NLRP3 activation by OA to regulate inflammatory responses. Thus, we propose that GYY4137 represents a promising novel therapeutic strategy for the treatment of OA.
Collapse
Affiliation(s)
- Jun Ma
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China; Department of Health Statistics, Naval Medical University, Shanghai, China; Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Peng Yang
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China; Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Zhibin Zhou
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China; Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Tengfei Song
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Liang Jia
- College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Naval Medical University, Shanghai, China
| | - Wei Yan
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China
| | - Jiuyi Sun
- Department of Orthepaedics, Naval Medical Center of PLA, Naval Medical University, Shanghai, China.
| | - Tianwen Ye
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| | - Lei Zhu
- Department of Orthopaedic Trauma Surgery, Shanghai Changzheng Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
15
|
Wang P, Sun Y, Zhang R, Guo Y, Zhang Y, Guo S, Wang Y, Gao J, Yang P, Deng Z. Salvianolic Acid A Attenuates Lipopolysaccharide-Induced Acute Lung Injury by Activating AMPK/SIRT1/Nrf2 Signaling Pathway. J Biochem Mol Toxicol 2025; 39:e70282. [PMID: 40272051 DOI: 10.1002/jbt.70282] [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: 04/11/2024] [Revised: 12/11/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Salvianolic acid A (Sal A) has been reported to have anti-inflammatory and antioxidant properties. The present study aimed to explore the potential mechanisms of Sal A on lipopolysaccharide (LPS)-induced acute lung injury (ALI). The results indicated that Sal A pretreatment attenuated LPS induced lung injury, shown by alleviated histopathological damage and alveolar-capillary barrier dysfunction, as well as reduced inflammatory response and oxidative stress. Moreover, Sal A pretreatment effectively increased the expression of p-AMPK and SIRT1 and promoted Nrf2 nuclear translocation in lung tissues. However, these effects were remarkably blunted by Compound C. Molecular docking experiments further confirmed that Sal A bound well to the active sites of AMPK and SIRT1. In conclusion, these results indicated that Sal A exerted its protective effects on LPS-induced ALI through suppressing inflammation and oxidative stress, which was mainly dependent on the activation of AMPK/SIRT1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Pengwei Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Clinical Pharmacy, Henan Province Key subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Ru Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Life Science Research Center, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yongli Guo
- Department of Ultrasound, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yongheng Zhang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Shengjie Guo
- Department of Endocrinology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Yemin Wang
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Jianlian Gao
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Xinxiang, China
| | - Zhijian Deng
- Department of Clinical Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Xinxiang Key Laboratory for Individualized Drug Use Research for Immune Diseases, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
- Clinical Pharmacy, Henan Province Key subjects of Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, China
| |
Collapse
|
16
|
Bertola L, Pepe G, Dolce A, Lecchi C, Borroni EM, Savino B, Canesi S, Sala L, Moretti P, Giordano A, Ressel L, Scanziani E, Vegeto E, Recordati C. Sex-dependent modulation of caerulein-induced acute pancreatitis in C57BL/6J mice. Vet Pathol 2025; 62:382-396. [PMID: 39878085 DOI: 10.1177/03009858241312606] [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] [Indexed: 01/31/2025]
Abstract
Acute pancreatitis (AP) is a life-threatening condition, with a higher mortality rate in men than women and in which estrogens might play a protective role. This study aimed to investigate sex-dependent differences in a mouse model of caerulein-induced AP. Thirty-six C57BL/6J mice (19 females and 17 males) were treated intraperitoneally with phosphate-buffered saline or caerulein, and sacrificed 12 hours, 2 days, or 7 days after the last injection. Blood was collected for amylase, lipase, and glucose determination. Severity and extent of inflammation, apoptosis, and acinar to ductal metaplasia (ADM) in pancreatic tissue were scored histologically and total macrophages, major histocompatibility complex (MHC)-II+ cells, M2 macrophages, T and B cells, neutrophils, apoptosis, and ADM were marked immunohistochemically and quantified by digital image analysis. Serum amylase had a peak at 12 hours, without differences between the sexes. In females, pancreatitis reached a peak at 12 hours with a fast recovery while, in males, the peak was delayed to day 2 with residual apoptosis still present. Macrophages were the main inflammatory cell population, followed by T cells, B cells and neutrophils, without differences between sexes. In males, CD206+ cells and apoptosis were higher at both days 2 and 7, and cytokeratin-19+ (CK19+) ADM was higher at day 7 compared with females. The results of this study revealed a faster onset and resolution of caerulein-induced AP in female mice compared with male mice, supporting a sex-dependent modulation of acute pancreatitis.
Collapse
Affiliation(s)
- Luca Bertola
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | | | | | | | - Elena Monica Borroni
- University of Milan, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Benedetta Savino
- University of Milan, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano, MI, Italy
| | - Simone Canesi
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | - Laura Sala
- University of Milan, Lodi, Italy
- Fondazione UNIMI, Milan, Italy
| | | | | | | | | | | | | |
Collapse
|
17
|
Wang Y, Dai GF, Xiao WB, Shi JS, Lin BW, Lin JD, Xiao XJ. Effects of continuous venous-venous hemofiltration with or without hemoperfusion on patients with hypertriglyceride acute pancreatitis. Clin Res Hepatol Gastroenterol 2025; 49:102572. [PMID: 40107658 DOI: 10.1016/j.clinre.2025.102572] [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: 01/28/2025] [Revised: 02/18/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND The role of continuous venous-venous hemofiltration (CVVH) and combined CVVH with hemoperfusion (HP) in patients with acute pancreatitis (AP) is diverse. We hypothesized HP+CVVH, rather than CVVH alone, could have significant benefits in hypertriglyceridemia (HTG)-AP patients. METHODS This single-center retrospective study included 347 patients with hypertriglyceride (HTH) -AP treated from January 2020 to December 2023. We assessed the association of short- and long-term outcomes (including incidence of systemic and local complications, length of ICU and hospital stays, and costs) between the HP+CVVH and CVVH groups. A subgroup analysis was performed to explore the effects of heterogeneity upon the incidence of severe AP (SAP). RESULTS Among 86 included patients, 40 received HP+CVVH therapy, and 46 received CVVH. Subgroup analysis revealed a lower incidence of severe AP after HP+CVVH therapy in patients with high procalcitonin, C-reactive protein, and interleukin-6 levels (46.4 % vs. 80.0 %, p = 0.019; 33.3 % vs. 72.7 %, p = 0.010; 37.5 % vs. 79.2 %, respectively). A significantly decreased hospital length of stay (LOS) in the HP+CVVH group was observed (10.40 [8.63-12.17] vs. 15.48 [13.02-17.94] days, p = 0.001). Furthermore, HP+CVVH showed a tendency towards lower hospital costs than CVVH ($5128 [4312-5943] vs. $8168 [6416-9920], p = 0.001). No significant differences were observed in the incidence of systemic or local complications, recurrence rates, or quality of life. CONCLUSIONS The use of HP+CVVH yielded superior outcomes in terms of the incidence of SAP compared to that of CVVH, for HTG-AP patients with a high inflammatory burden.
Collapse
Affiliation(s)
- Ying Wang
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Gao-Fan Dai
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Wen-Biao Xiao
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Jing-Shi Shi
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Bing-Wen Lin
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Jian-Dong Lin
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| | - Xiong-Jian Xiao
- Department of Intensive Care Units, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; Department of Intensive Care Units, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
18
|
Kocot N, Pękala E, Koczurkiewicz-Adamczyk P, Chłoń-Rzepa G, Łapa A, Wójcik-Pszczoła K. Airway and cardiovascular remodeling in chronic obstructive pulmonary disease (COPD) as a target for transient receptor potential ankyrin 1 (TRPA1) channel modulators. Bioorg Chem 2025; 158:108301. [PMID: 40058223 DOI: 10.1016/j.bioorg.2025.108301] [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: 11/08/2024] [Revised: 02/06/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by chronic inflammation, which leads to airway remodeling (AR). AR refers to various structural changes occurring in the airway wall, resulting in thickening, and narrowing of the airways. Apart from airways, and lung tissue, pulmonary vasculature also undergoes remodeling. Thus, the pressure in vascular bed is increased, leading to pulmonary hypertension and further right and left ventricle hypertrophy, as well as myocardial fibrosis. Currently, there is lack of effective treatment directly targeting airway and cardiovascular remodeling in the course of COPD. Due to a lot of research showing involvement of transient receptor potential ankyrin 1 (TRPA1) in respiratory disorders, it seems reasonable to consider this ion channel as a molecular target in treatment of remodeling consequences of COPD. The aim of this review is to summarize current knowledge of its role in this case and to identify areas requiring further research. Moreover, we provide few patented structures intended to treat chronic respiratory diseases, which may be worth investigating in the context of airway remodeling.
Collapse
Affiliation(s)
- Natalia Kocot
- Jagiellonian University, Doctoral School of Medical and Health Sciences, Łazarza 16, 31-530 Kraków, Poland; Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Elżbieta Pękala
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Paulina Koczurkiewicz-Adamczyk
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Grażyna Chłoń-Rzepa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Medicinal Chemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Aleksandra Łapa
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| | - Katarzyna Wójcik-Pszczoła
- Jagiellonian University Medical College, Faculty of Pharmacy, Department of Pharmaceutical Biochemistry, Medyczna 9, 30-688 Kraków, Poland.
| |
Collapse
|
19
|
Lou J, Xiang Z, Zhu X, Fan Y, Li J, Jin G, Cui S, Huang N. A bidirectional mendelian-randomization analyses of genetically predicted circulating levels of systemic inflammatory regulators with risk of sepsis. Medicine (Baltimore) 2025; 104:e42199. [PMID: 40295284 PMCID: PMC12040038 DOI: 10.1097/md.0000000000042199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 03/21/2025] [Accepted: 04/04/2025] [Indexed: 04/30/2025] Open
Abstract
Whether there is a causal relationship between circulating levels of systemic inflammatory regulators and sepsis remains unclear. To determine whether genetically predicted circulating levels of cytokines are associated with risk of sepsis, a bidirectional two-sample Mendelian randomization (MR) analysis based on the a STROBE-compliant cross-sectional observational study was conducted utilizing gene-wide association study (GWAS) data. Selected with rigor, single-nucleotide polymorphisms served as instrumental variables for subsequent MR analysis. The preferred method for the MR analysis was the inverse-variance weighted approach. However, for comprehensive sensitivity analyses, 6 additional MR methods were employed. Cochrane's Q test was performed to examine heterogeneity. A leave-one-out method ensured the stability of MR results. Our findings suggest an inverse association between the levels of beta-nerve growth factor (BNGF) and the risk of sepsis development (OR = 0.769, 95% CI = 0.599-0.987, P = .039). In contrast, higher levels of TNF-related apoptosis-inducing ligand and vascular endothelial growth factor A (VEGF-A) are positively correlated with sepsis risk (OR = 1.094, 95% CI = 1.012-1.183, P = .025; OR = 1.182, 95% CI = 1.016-1.375, P = .031, respectively). Reverse MR Analysis indicated that sepsis risk is linked with lower circulating levels of adenosine deaminase and Interleukin-17A (β = -0.043, 95% CI = -0.085 to -0.002, P = .042; β = -0.061, 95% CI = -0.108 to -0.013, P = .012, respectively), and also with higher circulating levels of BNGF, delta/notchlike epidermal growth factor-related receptor, fibroblast growth factor 23, leukemia inhibitory factor, monocyte chemoattractant protein-1, and osteoprotegerin (β = 0.056, 95% CI = 0.015-0.096, P = .007; β = 0.137, 95% CI = 0.035-0.240, P = .009; β = 0.118, 95% CI = 0.020-0.216, P = .018; β = 0.136, 95% CI = 0.020-0.252, P = .022; β = 0.143, 95% CI = 0.043-0.242, P = .005; β = 0.116, 95% CI = 0.010-0.222, P = .031, respectively). Sum up, our study provides evidence supporting a bidirectional causal relationship between sepsis and genetically predicted circulating levels of systemic inflammatory regulators.
Collapse
Affiliation(s)
- Jiaqi Lou
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Ziyi Xiang
- Institute of Pathology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Xiaoyu Zhu
- Health Science Center, Ningbo University, Ningbo, China
| | - Youfen Fan
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Jiliang Li
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Guoying Jin
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Shengyong Cui
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| | - Neng Huang
- Burn Department, Ningbo No. 2 Hospital, Ningbo, China
| |
Collapse
|
20
|
Huang L, Lv F, Bin Y, Zhao J, Li C, Zhao S, Hu S, Zhang L. A Hydrogen Sulfide-Activated NIR-II Fluorescence/NIR-I Photoacoustic Dual-Ratiometric Nanoprobe With Unique Recognition Reaction for Precise Visual Diagnosis of Hepatitis Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2501269. [PMID: 40270361 DOI: 10.1002/smll.202501269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/11/2025] [Indexed: 04/25/2025]
Abstract
Hydrogen sulfide (H2S) is a vital gaseous signaling molecule that plays a central role in various physiological and pathological processes. Given the complementary advantages of fluorescence (FL) and photoacoustic (PA) imaging, there is a growing demand for dual-ratiometric probes that enable precise in vivo monitoring of H2S levels. In this study, the use of 2-mercapto-1,3,4-thiadiazole (MTD) as a novel recognition group of H2S is presented for the first time, following conjugation with cyanine dyes to obtain a new PA probe Cy-MTD. To achieve dual-ratiometric imaging, Cy-MTD is incorporated into down-conversion nanoparticle (DCNP), resulting in the creation of a pioneering NIR-II FL/NIR-I PA dual-ratiometric nanoprobe DCNP@Cy-MTD. Cy-MTD undergoes the blueshift in absorption from 840 to 670 nm after reaction with H2S, enabling NIR-I ratiometric PA imaging of H2S by measuring the ratio of PA signal at 670 and 840 nm (PA670/PA840). In addition, due to the strong absorption of Cy-MTD ≈840 nm and the overlapping between the absorption spectrum of Cy-MTD and 808 nm excitation band of DCNP, the 808 nm-excited FL emission (F1550 nm,808Ex) of DCNP in DCNP@Cy-MTD nanoprobe is quenched through the competitive absorption, while it is restored upon the interaction with H2S because of the blueshift in absorption of Cy-MTD. Using the stable FL emission of DCNP under 980 nm excitation (F1550 nm,980Ex) as the reference signal, NIR-II ratiometric FL imaging (F1550 nm,808Ex/F1550 nm,980Ex) of H2S is achieved. The dual-ratiometric response features of the DCNP@Cy-MTD nanoprobe offer a significant advancement over traditional single-signal or single-modality imaging techniques. By providing enhanced accuracy and reliability, this probe allows for the diagnosis of hepatitis by monitoring the H2S, surpassing the capabilities of conventional histopathological methods, which provides a new way for more effective diagnostic strategies for liver diseases.
Collapse
Affiliation(s)
- Lixian Huang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Fei Lv
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Yidong Bin
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Jingjin Zhao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Caiying Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Shulin Zhao
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Shengqiang Hu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| | - Liangliang Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, 541004, China
| |
Collapse
|
21
|
Wang QY, Xu BY, Wang Y, Lin YM, Zheng LF, Liu G, Li DZ, Jiang CS, Wang W, Zeng XP. Sodium aescinate promotes apoptosis of pancreatic stellate cells and alleviates pancreatic fibrosis by inhibiting the PI3K/Akt/FOXO1 signaling pathways. Front Pharmacol 2025; 16:1554260. [PMID: 40331192 PMCID: PMC12052937 DOI: 10.3389/fphar.2025.1554260] [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: 01/01/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Chronic pancreatitis (CP) is an inflammatory disease of progressive pancreatic fibrosis, and pancreatic stellate cells (PSCs) are key cells involved in pancreatic fibrosis. To date, there are no clinical therapies available to reverse inflammatory damage or pancreatic fibrosis associated with CP. Sodium Aescinate (SA) is a natural mixture of triterpene saponins extracted from the dried and ripe fruits of horse chestnut tree. It has been shown to have anti-inflammatory and anti-edematous effects. This study aims to explore the therapeutic potential of SA in CP and the molecular mechanism of its modulation. Through in vivo animal models and experiments, we found that SA significantly alleviated pancreatic inflammation and fibrosis in caerulein-induced CP mice model. In addition, SA inhibited the proliferation, migration and activation of PSCs as well as promoted apoptosis of PSCs through a series of experiments on cells in vitro including CCK-8 assay, Western blotting, immunofluorescence staining, wound-healing assay, Transwell migration assays, flow cytometric analysis, etc. Further RNA sequencing and in vitro validation assays revealed that inhibition of the PI3K/AKT/FOXO1 signaling pathway was involved in the SA mediated promotion of PSCs apoptosis, thus alleviating pancreatic fibrosis. In conclusion, this study revealed that SA may have promising potential as therapeutic agent for the treatment of CP, and the PI3K/AKT/FOXO1 pathway is a potential therapeutic target for pancreatic inflammation and fibrosis.
Collapse
Affiliation(s)
- Qing-Yun Wang
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Bai-Yan Xu
- Department of Digestive Diseases, Huian County Hospital, Quanzhou, China
| | - Yi Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan-Mei Lin
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Lin-Fu Zheng
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Gang Liu
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Da-Zhou Li
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Chuan-Shen Jiang
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Wen Wang
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| | - Xiang-Peng Zeng
- Department of Digestive Diseases, Dongfang Hospital of Xiamen University, School of Medicine, Xiamen University, Fuzhou, China
- Department of Digestive Diseases, 900th Hospital of PLA Joint Logistic Support Force, Fuzhou, China
- Department of Digestive Diseases, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
22
|
Bin Y, Peng R, Lee Y, Lee Z, Liu Y. Efficacy of Xuebijing injection on pulmonary ventilation improvement in acute pancreatitis: a systematic review and meta-analysis. Front Pharmacol 2025; 16:1549419. [PMID: 40308770 PMCID: PMC12041077 DOI: 10.3389/fphar.2025.1549419] [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: 12/21/2024] [Accepted: 03/14/2025] [Indexed: 05/02/2025] Open
Abstract
Background Xuebijing injection (XBJI), as a Chinese patent medicine injection, consists of five botanical drugs for anti-inflammatory treatment. Acute pancreatitis (AP) is induced by localized inflammation, potentially resulting in multiple organ dysfunction syndromes, specifically including acute lung injury (ALI) and acute respiratory distress syndrome (ARDS). Recent studies suggest that XBJI effective in alleviating potentially easing ALI and ARDS. Objective We illustrated the efficacy and safety of XBJI for pulmonary function of AP by conducting a systematic literature review and meta-analysis. Methods We conducted searches across eight databases, including PubMed, Embase, and the Cochrane Library, up to September 2024. Two independent investigators screened and selected the literature based on predefined inclusion and exclusion criteria, followed by data extraction. The quality of the selected studies was assessed using the Cochrane Collaboration's Risk of Bias 2.0 tool. The data were then qualitatively analyzed and synthesized by using Review Manager software, in accordance with the PRISMA guidelines and the Cochrane Handbook. Conclusion This study showed that using conventional therapy combined with XBJI might increase the oxygenation index, lower the respiratory rate, and improve APACHE II scores and inflammatory biomarkers. However, there is a high risk of bias and the quality of the included studies is low. More well-designed, large-sample, and high-quality trials are needed to be conducted in multiple centers.
Collapse
Affiliation(s)
- Yuling Bin
- Intensive Care Medicine Department of Hengyang Central Hospital, Hunan, China
| | - Rumei Peng
- Department of Pathology, Changsha Medical University, Hunan, China
| | - Yaqian Lee
- Intensive Care Medicine Department of Hengyang Central Hospital, Hunan, China
| | - Zhijie Lee
- Intensive Care Medicine Department of Hengyang Central Hospital, Hunan, China
| | - Yang Liu
- Intensive Care Medicine Department of Hengyang Central Hospital, Hunan, China
| |
Collapse
|
23
|
Li J, Jia YC, Lu J, Zhang H, Wang Z, Xie X, Cao F, Li F. Inhibition of Zbp1-PANoptosome-mediated PANoptosis effectively attenuates acute pancreatitis. Cell Death Discov 2025; 11:180. [PMID: 40240343 PMCID: PMC12003674 DOI: 10.1038/s41420-025-02451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Early acute pancreatitis is an acute inflammatory disease that involves multiple modes of cell death, including apoptosis, necrotic apoptosis, and pyroptosis in its disease process. PANoptosis, a type of cell death that includes pyroptosis, apoptosis, and necroptosis, has had an important role in a variety of infectious and inflammatory diseases in recent years. To judge the relationship between PANoptosis and AP, we first analyzed the data from pancreatic transcriptome data by bioinformatics techniques, and we found the enrichment of PANoptosis pathway in AP. Next, we screened the genes and identified differentially expressed genes (DEGs) associated with AP and PANoptosis. Finally, we found that Zbp1 may have a major role in the process of PANoptosis. For this purpose, we constructed AP models in mice and in vitro cell line 266-6 and intervened by inhibiting Zbp1. The final results showed that the PANoptosis in mice was significantly suppressed after inhibition of Zbp1. In conclusion, inflammatory injury in AP can be significantly improved by inhibiting Zbp1- PANoptosome-mediated PANoptosis.
Collapse
Affiliation(s)
- Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Yu-Chen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Jiongdi Lu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China.
| |
Collapse
|
24
|
Ahmed F, Abu-El-Haija M. Acute Pancreatitis in Children: It's Not Just a Simple Attack. Gastroenterology 2025:S0016-5085(25)00633-X. [PMID: 40228704 DOI: 10.1053/j.gastro.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 04/16/2025]
Abstract
Acute pancreatitis (AP) in children presents unique challenges distinct from adult manifestations, requiring specialized diagnostic and therapeutic approaches. Compared with adults, pediatric AP has lower mortality rates but still carries significant morbidity and potential long-term complications. This review examines current evidence on pediatric AP, highlighting recent advances in diagnosis, risk stratification, and management strategies. Current diagnostic approaches use serum lipase and amylase testing, along with various imaging modalities that have different diagnostic values. Recent research has identified promising biomarkers for predicting severe AP, including blood urea nitrogen, C-reactive protein, and specific cytokine signals. Emerging evidence suggests a role of gut microbiome dysbiosis in disease pathogenesis, opening new therapeutic possibilities targeting the gut-pancreas axis. Genetic factors, specifically pancreatitis risk genes, influence disease progression to recurrent and chronic pancreatitis. In this review, we summarize the consequences of an isolated AP episode in children. Our review highlights for the first time how AP can lead to significant long-term sequelae, including exocrine/nutritional deficiencies, endocrine pancreatic dysfunction, diabetes, recurrent pain, and decreased quality of life compared with healthy population controls. The goal of this review is to summarize advances in understanding of pediatric AP and to emphasize the importance of early recognition, appropriate risk stratification, and comprehensive follow-up after the first pediatric AP episode, while highlighting areas requiring future research to optimize patient outcomes.
Collapse
Affiliation(s)
- Faizan Ahmed
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Maisam Abu-El-Haija
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
25
|
Wan T, Wei A, Ding Z, Gonzalez SC, Wang J, Hou X, Luo X, He L, Song Z. Inflammation and macrophage infiltration exacerbate adult incision response by early life injury. BMC Anesthesiol 2025; 25:165. [PMID: 40211125 PMCID: PMC11983940 DOI: 10.1186/s12871-025-03029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/26/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Neonatal hindpaw incision can evoke long-lasting changes in nociceptive processing following repeat injury in adulthood. Studies have focused on the effects and mechanisms in the spinal cord and brain, however changes in inflammation and macrophages in the periphery, especially at the site of early life injury, remain poorly defined. In this paper, we investigated the role of macrophages in the injured tissue in pain hypersensitivity caused by repeat hindpaw incisions and primed by neonatal injury. METHODS Hindpaw incision was performed in anesthetized adult rats. Among them, some had neonatal hindpaw incisions on postnatal day 3. To assess the role of inflammatory response in the priming of adult incision pain, the rats were treated with clodronate liposome, a macrophage depletion agent, and ketorolac tromethamine, the commonly used anti-inflammatory drug following surgery. Their mechanical pain sensitivity was measured via von Frey filaments. Inflammation induced by hindpaw incision was evaluated via Enzyme-linked Immunosorbent Assay, H&E, and immunofluorescence staining. The phenotypes of macrophages were examined by analyzing their surface markers by flow cytometry. RESULTS Mechanical pain hypersensitivity caused by the hindpaw incision in the adult rats was enhanced by previous neonatal injury, which also significantly increased microglial activation in the spinal dorsal horn, aggravation of inflammation, and infiltration of both M1 and M2 macrophages in damaged hindpaw tissue after the repeat incision in the adult rats on POD 5. Intraperitoneal injection of clodronate liposome alleviates nociceptive and inflammatory responses in neonatal injured rats. Intramuscular injection of ketorolac tromethamine decreased mechanical hyperalgesia and inflammatory responses primed by prior neonatal injury. CONCLUSIONS Neonatal tissue injury exacerbated mechanical hypersensitivity, infiltration, and activation of macrophages evoked by repeat hindpaw incision in adulthood.
Collapse
Affiliation(s)
- Tong Wan
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Anqi Wei
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
- Department of Anesthesiology, Guiqian International General Hospital, Guiyang, 550024, China
| | - Zhuofeng Ding
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China.
| | - Sarel Chavarria Gonzalez
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Jian Wang
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Xinran Hou
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Xiao Luo
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Liqiong He
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China
| | - Zongbin Song
- 1Department of Anesthesiology, Xiangya Hospital Central South University, Changsha, 410008, China.
- Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, 410008, China.
| |
Collapse
|
26
|
Overton E, Emelyanova A, Bunik VI. Thiamine, gastrointestinal beriberi and acetylcholine signaling. Front Nutr 2025; 12:1541054. [PMID: 40271433 PMCID: PMC12014454 DOI: 10.3389/fnut.2025.1541054] [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: 12/06/2024] [Accepted: 03/20/2025] [Indexed: 04/25/2025] Open
Abstract
Research has highlighted numerous detrimental consequences of thiamine deficiency on digestive function. These range from impaired gastric and intestinal motility to aberrant changes in pancreatic exocrine function, gastric acidity and disturbances in gut barrier integrity and inflammation. Thiamine and its pharmacological forms, as a primary or adjunctive therapy, have been shown to improve symptoms such as nausea, constipation, dysphagia and intestinal dysmotility, in both humans and animals. This review aims to explore molecular mechanisms underlying the therapeutic action of thiamine in gastrointestinal dysfunction. Our analysis demonstrates that thiamine insufficiency restricted to the gastrointestinal system, i.e., lacking well-known symptoms of dry and wet beriberi, may arise through (i) a disbalance between the nutrient influx and efflux in the gastrointestinal system due to increased demands of thiamine by the organism; (ii) direct exposure of the gastrointestinal system to oral drugs and gut microbiome, targeting thiamine-dependent metabolism in the gastrointestinal system in the first line; (iii) the involvement of thiamine in acetylcholine (ACh) signaling and cholinergic activity in the enteric nervous system and non-neuronal cells of the gut and pancreas, employing both the coenzyme and non-coenzyme actions of thiamine. The coenzyme action relies on the requirement of the thiamine coenzyme form - thiamine diphosphate - for the production of energy and acetylcholine (ACh). The non-coenzyme action involves participation of thiamine and/or derivatives, including thiamine triphosphate, in the regulation of ACh synaptic function, consistent with the early data on thiamine as a co-mediator of ACh in neuromuscular synapses, and in allosteric action on metabolic enzymes. By examining the available evidence with a focus on the gastrointestinal system, we deepen the understanding of thiamine's contribution to overall gastrointestinal health, highlighting important implications of thiamine-dependent mechanisms in functional gastrointestinal disorders.
Collapse
Affiliation(s)
| | - Alina Emelyanova
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| | - Victoria I. Bunik
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- Department of Biochemistry, Sechenov University, Moscow, Russia
| |
Collapse
|
27
|
Kalkan AE, BinMowyna MN, Raposo A, Ahmad MF, Ahmed F, Otayf AY, Carrascosa C, Saraiva A, Karav S. Beyond the Gut: Unveiling Butyrate's Global Health Impact Through Gut Health and Dysbiosis-Related Conditions: A Narrative Review. Nutrients 2025; 17:1305. [PMID: 40284169 PMCID: PMC12029953 DOI: 10.3390/nu17081305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/03/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
Short-chain fatty acids (SCFAs), mainly produced by gut microbiota through the fermentation process of dietary fibers and proteins, are crucial to human health, with butyrate, a famous four-carbon SCFA, standing out for its inevitably regulatory impact on both gut and immune functions. Within this narrative review, the vital physiological functions of SCFAs were examined, with emphasis on butyrate's role as an energy source for colonocytes and its ability to enhance the gut barrier while exhibiting anti-inflammatory effects. Knowledge of butyrate synthesis, primarily generated by Firmicutes bacteria, can be influenced by diets with specifically high contents of resistant starches and fiber. Butyrate can inhibit histone deacetylase, modulate gene expression, influence immune functionality, and regulate tight junction integrity, supporting the idea of its role in gut barrier preservation. Butyrate possesses systemic anti-inflammatory properties, particularly, its capacity to reduce pro-inflammatory cytokines and maintain immune homeostasis, highlighting its therapeutic potential in managing dysbiosis and inflammatory diseases. Although butyrate absorption into circulation is typically minimal, its broader health implications are substantial, especially regarding obesity and type 2 diabetes through its influence on metabolic regulation and inflammation. Furthermore, this narrative review thoroughly examines butyrate's growing recognition as a modulator of neurological health via its interaction with the gut-brain axis. Additionally, butyrate's neuroprotective effects are mediated through activation of specific G-protein-coupled receptors, such as FFAR3 and GPR109a, and inhibition of histone deacetylases (HDACs). Research indicates that butyrate can alleviate neurological disorders, including Alzheimer's, Parkinson's, autism spectrum disorder, and Huntington's disease, by reducing neuroinflammation, enhancing neurotransmitter modulation, and improving histone acetylation. This focus will help unlock its full therapeutic potential for metabolic and neurological health, rather than exclusively on its well-known benefits for gut health, as these are often interconnected.
Collapse
Affiliation(s)
- Arda Erkan Kalkan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| | - Mona N. BinMowyna
- College of Education, Shaqra University, Shaqra 11911, Saudi Arabia;
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Md Faruque Ahmad
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Faiyaz Ahmed
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia;
| | - Abdullah Y. Otayf
- Department of Clinical Nutrition, College of Nursing and Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; (M.F.A.); (A.Y.O.)
| | - Conrado Carrascosa
- Department of Animal Pathology and Production, Bromatology and Food Technology, Faculty of Veterinary, Universidad de Las Palmas de Gran Canaria, Trasmontaña s/n, 35413 Arucas, Spain;
| | - Ariana Saraiva
- Research in Veterinary Medicine (I-MVET), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal;
- Veterinary and Animal Research Centre (CECAV), Faculty of Veterinary Medicine, Lisbon University Centre, Lusófona University, Campo Grande 376, 1749-024 Lisboa, Portugal
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17100, Turkey;
| |
Collapse
|
28
|
Cai W, Wu A, Lin Z, Cao W, Pathak JL, Jaspers RT, Li R, Li X, Zheng K, Lin Y, Zhou N, Zhang X, Zhu Y, Zhang Q. S-propargyl-cysteine attenuates temporomandibular joint osteoarthritis by regulating macrophage polarization via Inhibition of JAK/STAT signaling. Mol Med 2025; 31:128. [PMID: 40197110 PMCID: PMC11974036 DOI: 10.1186/s10020-025-01186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 03/26/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Temporomandibular joint osteoarthritis (TMJ-OA) is a disease characterized by cartilage degradation and synovial inflammation, with limited effective treatment currently. Synovial macrophage polarization is pivotal in TMJ-OA progression, making it a promising therapeutic aspect. This study investigated the effects of S-propargyl-cysteine (SPRC), an endogenous H2S donor, on macrophage polarization and its therapeutic potential in alleviating TMJ-OA. METHODS A MIA-induced TMJ-OA rat model and LPS-stimulated RAW264.7 macrophages were employed to evaluate the effects of SPRC in vivo and in vitro. TMJ bone and cartilage were analyzed via micro-CT and histological methods, while macrophage polarization markers expression were assessed via RT-qPCR, western blot, and immunofluorescence. RNA sequencing was performed on macrophages, and the JAK2/STAT3 signaling pathway was validated using the JAK2-specific inhibitor AG490. The direct effects of SPRC on rat primary condylar chondrocytes were examined by evaluating ECM synthesis and degradation. Co-culture experiments further assessed macrophage-chondrocyte interactions. RESULTS SPRC significantly alleviated cartilage and bone damage in the TMJ-OA rat model, as demonstrated by improved bone volume and cartilage structure. SPRC reduced pro-inflammatory M1 macrophage infiltration and enhanced anti-inflammatory M2 macrophage polarization. SPRC effectively inhibited the JAK2/STAT3, leading to reduction of inflammatory markers, including TNF-α, IL-6, and iNOS. Co-culture experiments revealed that SPRC-treated macrophage-conditioned medium improved chondrocyte metabolic activity and restored ECM integrity. CONCLUSIONS SPRC-modulated macrophage polarization alleviates TMJ-OA via JAK/STAT downregulation, thereby reducing synovial inflammation and cartilage degradation. These findings position SPRC as a promising therapeutic candidate for TMJ-OA and provide insights into novel strategies targeting macrophage polarization and synovium-cartilage crosstalk.
Collapse
Affiliation(s)
- Wenyi Cai
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Antong Wu
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Zhongxiao Lin
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Wei Cao
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Janak L Pathak
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Richard T Jaspers
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, The Netherlands
| | - Rui Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Xin Li
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Kaihan Zheng
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Yufu Lin
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China
| | - Na Zhou
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
| | - Xin Zhang
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China
| | - Yizhun Zhu
- School of Pharmacy, State Key Laboratory of Quality Research in Chinese Medicines and Laboratory of Drug Discovery from Natural Resources and Industrialization, Macau University of Science and Technology, Room 210, Block E, Avenida Wai Long, Taipa, Macau, China.
| | - Qingbin Zhang
- Department of Temporomandibular Joint, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, 195 Dongfeng Road (West), Yuexiu District, Guangzhou, Guangdong, 510140, China.
| |
Collapse
|
29
|
Li J, Zhang QY, Zhang MH, Jiang SY. Maternal and fetal death associated with acute pancreatitis during pregnancy: A case report. World J Clin Cases 2025; 13:102011. [PMID: 40191677 PMCID: PMC11670032 DOI: 10.12998/wjcc.v13.i10.102011] [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: 10/05/2024] [Revised: 11/10/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Acute pancreatitis in pregnancy is a rare but serious condition that can lead to high maternal mortality and fetal loss. Instances of pregnancy complicated by severe acute pancreatitis, particularly with subsequent respiratory and cardiac arrest, are rarely reported. CASE SUMMARY We present the case of a 35-year-old woman, at 36 + 5 weeks of gestation, who presented with paroxysmal epigastric pain accompanied by low back pain, nausea, and vomiting. According to the clinical symptoms, B-ultrasound imaging and biochemical indicators, the patient was diagnosed with acute pancreatitis and initially managed conservatively. However, 3 hours after admission, the patient experienced respiratory and cardiac arrest, and the fetus died. In this case, the adverse outcomes occurred due to the lack of aggressive fluid resuscitation and an active surgical intervention. CONCLUSION Implementing aggressive fluid resuscitation to sustain tissue perfusion, alongside the proactive evaluation of pharmacological agents that suppress gastric acid secretion and inhibit pancreatic enzyme activity, may be beneficial in mitigating the risk of a severely adverse prognosis. Effective management of acute pancreatitis during pregnancy requires careful timing of surgical intervention, a thorough evaluation of the risks and benefits regarding the continuation or termination of pregnancy, and a focus on safeguarding both maternal and fetal health.
Collapse
Affiliation(s)
- Jin Li
- Department of Anesthesiology, Kaihua County People’s Hospital, Quzhou 324300, Zhejiang Province, China
| | - Qing-Yan Zhang
- Department of Anesthesiology, Kaihua County People’s Hospital, Quzhou 324300, Zhejiang Province, China
| | - Mei-Hong Zhang
- Department of Anesthesiology, Kaihua County People’s Hospital, Quzhou 324300, Zhejiang Province, China
| | - Shan-Yun Jiang
- Center for Clinical Inspection and Quarantine, Kaihua County Center for Clinical Inspection and Quarantine, Quzhou 324300, Zhejiang Province, China
| |
Collapse
|
30
|
Szabo C. Role of cystathionine-β-synthase and hydrogen sulfide in down syndrome. Neurotherapeutics 2025:e00584. [PMID: 40187942 DOI: 10.1016/j.neurot.2025.e00584] [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: 01/23/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/07/2025] Open
Abstract
Down syndrome (DS) is a genetic condition where the person affected by it is born with an additional - full or partial - copy of chromosome 21. DS presents with characteristic morphological features and is associated with a wide range of biochemical alterations and maladaptations. Cystathionine-β-synthase (CBS) - one of the key mammalian enzymes responsible for the biogenesis of the gaseous transmitter hydrogen sulfide (H2S) - is located on chromosome 21, and people with DS exhibit a significant upregulation of this enzyme in their brain and other organs. Even though 3-mercaptopyruvate sulfurtransferase - another key mammalian enzyme responsible for the biogenesis of H2S and of reactive polysulfides - is not located on chromosome 21, there is also evidence for the upregulation of this enzyme in DS cells. The hypothesis that excess H2S in DS impairs mitochondrial function and cellular bioenergetics was first proposed in the 1990s and has been substantiated and expanded upon over the past 25 years. DS cells are in a state of metabolic suppression due to H2S-induced, reversible inhibition of mitochondrial Complex IV activity. The impairment of aerobic ATP generation in DS cells is partially compensated by an upregulation of glycolysis. The DS-associated metabolic impairment can be reversed by pharmacological CBS inhibition or CBS silencing. In rodent models of DS, CBS upregulation and H2S overproduction contribute to the development of cognitive dysfunction, alter brain electrical activity, and promote reactive gliosis: pharmacological inhibition or genetic correction of CBS overactivation reverses these alterations. CBS can be considered a preclinically validated drug target for the experimental therapy of DS.
Collapse
Affiliation(s)
- Csaba Szabo
- Section of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Switzerland.
| |
Collapse
|
31
|
Gupta M, Das N, Mohanty S, Shunmugam R. Hydrogen Sulfide Induced Unique Twisted-Rod Assemblies from Norbornene-Based Polymer in Aqueous Environment as Gasotransmitter Regulators. Chem Asian J 2025; 20:e202401571. [PMID: 39946149 DOI: 10.1002/asia.202401571] [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: 11/06/2024] [Revised: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Hydrogen sulfide, being a poisonous pollutant, (H2S) is known for its pungent smell. Although it is having significant role in the biological and physiological processes as a potential biomarker in the present day. The over-expression of H2S leads to several biological disorders and diseases, therefore, monitoring the level of H2S in the biological pH is important. Optical sensors to detect hydrogen sulfide have gained popularity due to their cost-effectiveness, portability, fast response, etc. Here, we are designing a norbornene-based polymeric bio-sensor for detecting hydrogen sulfide in physiological pH. Hydrophilic and hydrophobic monomers are strategically crafted to permeate polymeric sensors with water solubility and the ability to detect hydrogen sulfide, respectively. These monomers are polymerized by ring-opening metathesis polymerization (ROMP). The probe is characterized by nuclear magnetic resonance (NMR) spectroscopy, fluorescence, and UV-visible absorbance spectra. The polymeric sensor can selectively detect hydrogen sulfide with high sensitivity by turn-on fluorescence responses. Moreover, this probe efficiently conducts the morphological changes associated with sensing phenomena. The collapsed vesicles are nicely converted into twisted rods. One dimentional structures (such as twisted rods, etc.) are becoming very interesting as they have fascinating structural and functional properties. Having application in biomedical fields such as targeted drug delivery, imaging, and scaffolds for tissue engineering, it also can exhibits improved mechanical strength, flexibility, unique optical properties, increased surface area, etc. This probe is biocompatible and has the scopes for bio-imaging. Therefore, this water-soluble polymeric probe can open new bio-medical applications for detecting analytes.
Collapse
Affiliation(s)
- Moumita Gupta
- Polymer Research Center, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Kolkata, West Bengal, 741246, India
| | - Narayan Das
- Polymer Research Center, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Kolkata, West Bengal, 741246, India
| | - Srujana Mohanty
- Polymer Research Center, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Kolkata, West Bengal, 741246, India
| | - Raja Shunmugam
- Polymer Research Center, Department of Chemical Sciences, Indian Institute of Science Education and Research (IISER), Kolkata, West Bengal, 741246, India
| |
Collapse
|
32
|
Sickinger M, Jörling J, Büttner K, Roth J, Wehrend A. Association of Stress and Inflammatory Diseases with Serum Ferritin and Iron Concentrations in Neonatal Calves. Animals (Basel) 2025; 15:1021. [PMID: 40218414 PMCID: PMC11987952 DOI: 10.3390/ani15071021] [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: 03/06/2025] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025] Open
Abstract
This study investigated the effects of iron supplementation and inflammatory disease on cortisol, white blood cell (WBC) count, total protein (TP), lactate, interleukin 1 β (IL1β), interleukin 6 (IL6), substance P (SP), hepcidin, haptoglobin, and ferric-reducing ability of plasma (FRAP) in calves. Correlation analyses for the aforementioned parameters with serum iron and ferritin were performed in 40 neonatal calves over the first 10 days of life. Neither iron supplementation, disease status, nor sex had statistically significant effects on the areas under the curve of ferritin, WBC, TP, IL1β, IL6, SP, hepcidin, haptoglobin, or FRAP. However, cortisol concentrations were influenced by disease development. Cortisol concentrations were higher at birth (44.1 ± 1.95 ng/mL) than on day 2 (38.8 ± 1.87 ng/mL) (p = 0.0477), and healthy animals exhibited lower cortisol concentrations than diseased calves (p = 0.0028). Correlation analyses indicated weak positive correlations between ferritin and IL1β (p = 0.0015; ρ = 0.49) and IL6 (p = 0.0011; ρ = 0.50), respectively. The clinical significance of these findings and resulting therapeutic consequences, especially with respect to iron supplementation, should be further investigated in calves and adult cattle.
Collapse
Affiliation(s)
- Marlene Sickinger
- Clinic for Ruminants and Herd Health Management, Justus-Liebig-University of Giessen, 35392 Giessen, Germany
| | | | - Kathrin Büttner
- Department for Biomathematics and Data Processing, Justus-Liebig-University of Giessen, 35392 Giessen, Germany;
| | - Joachim Roth
- Institute for Veterinary Physiology, Justus-Liebig-University of Giessen, 35390 Giessen, Germany;
| | - Axel Wehrend
- Veterinary Clinic for Reproduction and Neonatology, Justus-Liebig-University of Giessen, 35392 Giessen, Germany;
| |
Collapse
|
33
|
Bhattacharya R, Avdieiev SS, Bukkuri A, Whelan CJ, Gatenby RA, Tsai KY, Brown JS. The Hallmarks of Cancer as Eco-Evolutionary Processes. Cancer Discov 2025; 15:685-701. [PMID: 40170539 DOI: 10.1158/2159-8290.cd-24-0861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/19/2024] [Accepted: 01/28/2025] [Indexed: 04/03/2025]
Abstract
SIGNIFICANCE Viewing the hallmarks as a sequence of adaptations captures the "why" behind the "how" of the molecular changes driving cancer. This eco-evolutionary view distils the complexity of cancer progression into logical steps, providing a framework for understanding all existing and emerging hallmarks of cancer and developing therapeutic interventions.
Collapse
Affiliation(s)
- Ranjini Bhattacharya
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Cancer Biology, University of South Florida, Tampa, Florida
| | - Stanislav S Avdieiev
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Anuraag Bukkuri
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania
- Center for Evolutionary Biology and Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Christopher J Whelan
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Metabolism and Physiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Robert A Gatenby
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Kenneth Y Tsai
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Tumor Microenvironment & Metastasis, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Joel S Brown
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
34
|
Wang Q, Liu X, Du Z, Zheng Y, Meng Z, Lv Z, Wang L, Xue D. Astragalus polysaccharide reduces the severity of acute pancreatitis under a high-fat diet through enriching L. reuteri and propionate. Int J Biol Macromol 2025; 298:140021. [PMID: 39837455 DOI: 10.1016/j.ijbiomac.2025.140021] [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: 10/31/2024] [Revised: 12/21/2024] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
Acute pancreatitis (AP) is a severe digestive disorder, worsened by a high-fat diet (HFD) through inflammation and gut microbiota disruption. Astragalus polysaccharides (APS), known for their anti-inflammatory properties, may alleviate HFD-induced exacerbation of AP by modulating gut microbiota. This study investigates the effect of APS on AP severity under a HFD (HAP). Results show that HFD significantly worsens AP, with elevated serum enzymes, pro-inflammatory cytokines, and pancreatic damage. Single-cell RNA sequencing revealed increased ICAM1+ neutrophils and activation of the NF-κB/necroptosis pathway in HAP mice. Treatment with APS reduced neutrophil infiltration, downregulated NF-κB, and suppressed necroptosis. APS also restored gut microbiota balance, boosting Lactobacillus reuteri (L. reuteri) and propionate (PA) levels. Interventions with L. reuteri or PA reduced HAP severity, with combined treatment showing synergistic effects. These findings suggest that the protective effect of APS is mediated by microbiota-dependent mechanisms, highlighting the gut-pancreas axis as a potential therapeutic target for AP.
Collapse
Affiliation(s)
- Qiang Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Xuxu Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiwei Du
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yi Zheng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ziang Meng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhenyi Lv
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Liyi Wang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Dongbo Xue
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
35
|
An Y, Tu Z, Wang A, Gou W, Yu H, Wang X, Xu F, Li Y, Wang C, Li J, Zhang M, Xiao M, Di Y, Hou W, Cui Y. Qingyi decoction and its active ingredients ameliorate acute pancreatitis by regulating acinar cells and macrophages via NF-κB/NLRP3/Caspase-1 pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156424. [PMID: 40020626 DOI: 10.1016/j.phymed.2025.156424] [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: 10/29/2024] [Revised: 01/08/2025] [Accepted: 01/23/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND AND PURPOSE Macrophage infiltration and activation is a critical step during acute pancreatitis (AP). NLRP3 inflammasomes in macrophages plays a critical role in mediating pancreatic inflammatory responses. Qing-Yi Decoction(QYD)has been used for many years in clinical practice of Nankai Hospital combined with traditional Chinese and western medicine treatment of acute pancreatitis. Although QYD has a well-established clinical efficacy, little is known about its bioactive ingredients, how they interact with different therapeutic targets and the pathways to produce anti-inflammatory effects. Here, we elucidate the therapeutic effects of QYD against acute pancreatitis and reveal its mechanism of action. METHODS The main components of QYD were identified using UHPLC-Q-Orbitrap MS. Network pharmacology was employed to predict potential therapeutic targets and their mechanisms of action. C57BL/6 mice were randomly divided into control group, model group, low, medium and high dose (6, 12, 24 g/kg) QYD groups, with 10 mice in each group. The therapeutic effect of QYD on cerulein-induced acute pancreatitis. (CER-AP) was evaluated by histopathological score, immunohistochemistry, serum amylase and cytokines detection by ELISA. The protein expressions of MyD88/NF-κB/NLRP3 signaling pathway were detected by Western blotting. Along with molecular docking of key bioactive compounds and targets, RAW264.7 cells stimulated with 1μg/ml LPS is used to screen components with more potent effects on target proteins. AR42 J cells were stimulated with 100 nM dexamethasone (dexa) combined with 10 nM cerulein (CN) as s a cell-culture model of acute pancreatitis. Inhibitory effects of the main chemical composition Wogonoside on NLRP3 inflammasomes were analyzed by qRT-PCR and Western blots. RESULTS Using UHPLC-Q-Orbitrap MS, 217 compounds were identified from QYD, including Wogonoside, Catechins, Rhein, etc. A visualization network of QYD-compounds-key targets-pathways-AP show that QYD may modulate PI3K-Akt signaling pathway, NOD-like receptor signaling pathway, MAPK signaling pathway, Ras signaling pathway and Apoptosis signaling pathway by targeting TNF, IL1β, AKT1, TP53 and STAT3 exerting a therapeutic effect on AP. QYD administration effectively mitigated CER-induced cytokine storm, pancreas edema and serum amylase. QYD (12 mg/kg) showed better effect. The protein expression levels of MyD88, NF-κB, NLRP3, Caspase-1 and GSDMD in pancreatic tissue were significantly decreased. Through molecular docking and LPS-RAW264.7 inflammation model, the selected Wogonoside significantly decreased IL-1β mRNA. The expression levels of NLRP3/Caspase-1/GSDMD pathway-related proteins were also decreased on AR42J-AP. CONCLUSION The results of network pharmacology indicate that QYD can inhibit AP through multiple pathways and targets. This finding was validated through in vivo tests, which demonstrated that QYD can reduce AP by inhibiting NLRP3 inflammasomes, additionally, it should be noted that 12mg/kg was a relatively superior dose. One of the main chemical compositions Wogonoside regulated NLRP3 inflammasome activation to protect against AP. This study is the first to verify the intrinsic molecular mechanism of QYD in treating AP by combining network pharmacology and animal experiments. The findings can provide evidence for subsequent clinical research and drug development.
Collapse
Affiliation(s)
- Yu An
- Tianjin Medical University, Tianjin, China
| | - Zhengwei Tu
- Tianjin Nankai Hospital, Tianjin, China; Department of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Nankai Clinical School of Medicine, Tianjin Medical University, Tianjin, China
| | - Ao Wang
- Tianjin Medical University, Tianjin, China
| | - Wenfeng Gou
- Peking Union Medical College & Institute of Radiological Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Huijuan Yu
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | | | - Feifei Xu
- Peking Union Medical College & Institute of Radiological Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Yanli Li
- Peking Union Medical College & Institute of Radiological Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China
| | - Cong Wang
- Tianjin Medical University, Tianjin, China
| | - Jinan Li
- Tianjin Medical University, Tianjin, China
| | - Mengyue Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; State Key Laboratory of Chinese Medicine Modernization, State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | | | - Ying Di
- Tianjin Medical University, Tianjin, China
| | - Wenbin Hou
- Peking Union Medical College & Institute of Radiological Medicine, Chinese Academy of Medical Sciences, Tianjin 300192, China.
| | - Yunfeng Cui
- Tianjin Medical University, Tianjin, China; Tianjin Nankai Hospital, Tianjin, China; Department of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Nankai Clinical School of Medicine, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
36
|
Paquet A, Bahlouli N, Coutel X, Leterme D, Delattre J, Gauthier V, Miellot F, Delplace S, Rouge-Labriet H, Bertheaume N, Chauveau C, Benachour H. Obesity and insulinopenic type 2 diabetes differentially impact, bone phenotype, bone marrow adipose tissue, and serum levels of the cathelicidin-related antimicrobial peptide in mice. Bone 2025; 193:117387. [PMID: 39742907 DOI: 10.1016/j.bone.2024.117387] [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: 07/31/2024] [Revised: 12/20/2024] [Accepted: 12/28/2024] [Indexed: 01/04/2025]
Abstract
Obesity is a risk factor of developing type 2 diabetes (T2D) and metabolic complications, through systemic inflammation and insulin resistance. It has also been associated with increased bone marrow adipocytes along with increased bone fragility and fracture risk. However, the differential effects of obesity and T2D on bone fragility remain unclear. The cathelicidin-related antimicrobial peptide (CRAMP) is a multifunctional modulator of the innate immunity that has emerged as biomarker of cardiometabolic diseases. The aims of this study were i) to assess the differential impact between hyperinsulinemic obesity versus insulinopenic T2D, on bone phenotype and bone marrow adipose tissue (BMAT), and ii) to analyse the link with CRAMP expression and its circulating levels in the context of obesity and T2D. We used C57BL/6 J male mice models of obesity induced by high-fat diet (HFD), and of insulinopenic T2D induced by streptozotocin (STZ) treatment combined with HFD, reflecting the metabolic heterogeneity of the diseases. As compared to low-fat diet (LFD) control group after 16 weeks of feeding, the HFD mice exhibit a significant weight gain, moderate hyperglycaemia, impaired glucose tolerance and insulin sensitivity, and significant increase in serum insulin levels. This hyperinsulinemic obesity led to decreased trabecular (Tb.Th) and cortical thickness (Ct.Th) in the tibia, associated with significant BMAT expansion, in addition to increased subcutaneaous (SCAT) and visceral adipose tissue (VAT). No changes were observed in the circulating levels of CRAMP peptide neither in other bone parameters. While, STZ treatment in HFD/STZ group induced a more severe hyperglycaemia, glucose intolerance and insulin resistance, and hypoinsulinemia. We also observed a negative effect on the expansion of both SCAT and VAT, as well as lower increase in BMAT as compared to HFD group. However, these mice with insulinopenic T2D exhibit early decrease in trabecular number (Tb.N) in proximal tibia, progressively from 8 to 16 weeks of protocol, and impaired femoral biomechanical stiffness. These alterations are also accompanied with decreased circulating levels of the CRAMP peptide in the HFD/STZ mice. The CRAMP mRNA levels decreased in VAT of both HFD and HFD/STZ groups. Overall, these results provide novel insights into the differential negative impact of obesity versus T2D on bone microenvironment, and suggest a link between hyperglycaemia-induced bone quality alterations during insulinopenia, and impaired regulation of the cathelicidin peptide of the innate immunity. Further investigations are needed to elucidate this relationship.
Collapse
Affiliation(s)
- Amélie Paquet
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Nadia Bahlouli
- ICube, Université de Strasbourg, CNRS, 2-4 Rue Boussingault, Strasbourg 67000, France
| | - Xavier Coutel
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Damien Leterme
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Jérôme Delattre
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Véronique Gauthier
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Flore Miellot
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Séverine Delplace
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Hélène Rouge-Labriet
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Nicolas Bertheaume
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Hamanou Benachour
- Marrow Adiposity and Bone Lab, MABLab-ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
37
|
Wang W, Gan Y, Jiang H, Fang M, Wu Z, Zhu W, Li C. A Novel DNBS-based Fluorescent Probe for the Detection of H 2S in Cells and on Test Strips. J Fluoresc 2025; 35:2109-2120. [PMID: 38502406 DOI: 10.1007/s10895-024-03660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/11/2024] [Indexed: 03/21/2024]
Abstract
Hydrogen sulfide (H2S) plays a key role in the physiology and pathology of organisms, and H2S in the environment is easily absorbed and harmful to health. It is of great significance to develop a probe with good selectivity, high sensitivity and good stability that can detect hydrogen sulfide inside and outside organisms. In this work, we designed a novel "turn-on" fluorescent probe CIM-SDB for the detection of H2S. The probe CIM-SDB used indene-carbazole as the fluorophore and 2,4-dinitrobenzenesulfonyl as the recognition site. The probe CIM-SDB exhibited high selectivity and sensitivity to H2S (detection limit as low as 123 nM). Moreover, the probe CIM-SDB was successfully applied to the detection of intracellular exogenous and endogenous H2S, and the test strips prepared by the probe CIM-SDB could realize the convenient and rapid detection of H2S.
Collapse
Affiliation(s)
- Wenxiang Wang
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, PR China
| | - Yudie Gan
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, PR China
| | - Huaqin Jiang
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, PR China
| | - Min Fang
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, PR China.
- Anhui Province Key Laboratory of Environment-friendly Polymer Materials, Anhui University, Hefei, 230601, PR China.
| | - Zhenyu Wu
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, PR China
| | - Weiju Zhu
- School of Chemistry and Chemical Engineering, Anhui University, Hefei, 230601, PR China
- Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei, 230601, PR China
| | - Cun Li
- School of Materials Science and Engineering, Anhui University, Hefei, 230601, PR China
| |
Collapse
|
38
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [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] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
39
|
Zhou H, Lai Y, Zhu Y, Shao F, Ma G, Yang N, Ma X, Sun Y, Shi Q. Quercetin improves airway remodeling in COPD rats by suppressing phenotypic switch of ASMCs via inhibiting the Wnt5a/β-catenin pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156491. [PMID: 39955824 DOI: 10.1016/j.phymed.2025.156491] [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: 10/12/2024] [Revised: 02/02/2025] [Accepted: 02/08/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND AND PURPOSE Airway remodeling in chronic obstructive pulmonary disease (COPD) is a contributor to airflow limitation, promotes disease progression, and affects disease outcome and prognosis. Quercetin has been identified as a potential therapeutic agent for COPD. Currently, there is insufficient research providing direct evidence to support this hypothesis. The present study investigates the therapeutic effects and the underlying mechanisms of quercetin in the alleviation of airway remodeling in rat models of COPD. EXPERIMENTAL STEPS This study used a network pharmacology approach to predict, for the first time, the potential molecular targets of quercetin in COPD. The effects of quercetin on phenotypic switching and mitochondrial function of ASMCs were assessed in vitro using CCK-8, EdU staining, migration assays, western blotting, and JC-1 staining. Additionally, the interaction between Wnt5a and quercetin was analyzed via molecular docking, and findings were experimentally confirmed using the cellular thermal shift assay (CETSA). Quercetin's influence on airway remodeling in COPD was examined in vivo through pulmonary function evaluation, H&E staining, and Modified Sirius Red staining. Molecular alterations associated with phenotypic switching, oxidative stress, autophagy and Wnt5a/β-Catenin pathway were examined by Western blotting, immunofluorescence, immunohistochemistry, DHE staining and ELISA. KEY RESULTS The results showed that quercetin has a beneficial therapeutic effect on COPD. Its ability to mitigate airway remodeling is linked to modulating autophagy levels, reducing oxidative stress, alleviating mitochondrial damage, and influencing the phenotypic switch in ASMCs. By increasing oxidative stress tolerance, quercetin reduces mitochondrial damage and regulates the phenotypic switch in ASMCs. Furthermore, quercetin suppresses autophagy hyperactivation, which subsequently lowers oxidative stress levels in ASMCs. Notably, quercetin modulates autophagy through the regulation of the Wnt5a/β-catenin signaling pathway. CONCLUSION AND IMPLICATIONS In conclusion, quercetin demonstrates potential therapeutic effects in COPD by suppressing the Wnt5a/β-cateninsignaling pathway, autophagy as well as oxidative stress, and thereby alleviating mitochondrial damage and the phenotypic switch in ASMCs. These findings may have clinical applications and offer new insights for the development of COPD treatments.
Collapse
Affiliation(s)
- Hui Zhou
- College of Traditional Chinese Medicine/College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Yingying Lai
- Department of Anesthesiology, Affiliated Hospital of Jiaxing University, Jiaxing 314001, PR China
| | - Yuanyuan Zhu
- College of Traditional Chinese Medicine/College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Feng Shao
- Key Laboratory of Innovation Drug and Efficient Energy- saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, PR China
| | - Guangqiang Ma
- College of Traditional Chinese Medicine/College of Life Sciences, Jiangxi University of Chinese Medicine, Nanchang 330004, PR China
| | - Ningning Yang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325027, PR China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Xianhui Ma
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, PR China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325027, PR China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325027, PR China
| | - Yinxiang Sun
- Qi Huang Chinese Medicine Academy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330025, PR China.
| | - Qiang Shi
- Qi Huang Chinese Medicine Academy, Jiangxi University of Traditional Chinese Medicine, Nanchang 330025, PR China.
| |
Collapse
|
40
|
Wang D, Xie A, Luo J, Li L, Zhang Z, Deng W, Yang B, Chang Y, Liang Y. Thiotaurine inhibits melanoma progression by enhancing Ca 2+ overload-induced cellular apoptosis. J Dermatol Sci 2025; 118:29-37. [PMID: 40189970 DOI: 10.1016/j.jdermsci.2025.03.001] [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: 11/06/2024] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Melanoma is the most dangerous type of skin cancer with poor therapy outcomes. Since malignant cells are more susceptible to Ca2+ overload than normal cells, activating Ca2+ overload-mediated apoptosis may be a promising strategy to inhibit melanoma progression. Hydrogen sulfide (H2S) donors can regulate Ca2+ channels, but their effects on melanoma cells remain unclear. OBJECTIVE To explore the effects of Thiotaurine (TTAU), an H2S donor, on melanoma cells and its underlying mechanisms. METHODS We tested the effect of TTAU by culturing melanoma cells in vitro and establishing the xenograft model of mice in vivo. Cell proliferation and apoptosis were assessed using the CCK-8 test and flow cytometry. Molecules involved in apoptosis or Ca2+-related signal transduction were analyzed by western blotting. Immunofluorescence was used to measure Ca2+ levels, mitochondrial damage, and reactive oxygen species (ROS). RESULTS TTAU significantly reduced melanoma cell viability and induced apoptosis both in vitro and in vivo. Mechanistically, TTAU increased intracellular Ca2+, upregulated transient receptor potential vanilloid 1(TRPV1), and decreased activating transcription factor 3(ATF3) by nuclear factor of activated T cell cytoplasmic 1(NFATc1). TTAU also caused mitochondrial damage and ROS overproduction, which also promoted apoptosis. CONCLUSION We first elucidate that TTAU inhibits melanoma progression by activating Ca2+ influx-NFATc1-ATF3 signaling and aggravating mitochondrial oxidative stress, in which TRPV1 may act as an amplifier for Ca2+ influx. Our research is expected to provide new ideas for the treatment of tumors such as melanoma, as well as the clinical application of reactive sulfur species-based drugs.
Collapse
Affiliation(s)
- Di Wang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Ansheng Xie
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Jialiang Luo
- Department of Medical Laboratory, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Lei Li
- Department of Clinical Laboratory, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhiwen Zhang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Deng
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Bin Yang
- Dermatology Hospital, Southern Medical University, Guangzhou, China
| | - Yuan Chang
- Department of Neurology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| | - Yunsheng Liang
- Dermatology Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
41
|
Erdal BDY, Erdal H. Evaluation of retinal structural and microvascular changes in patients with acute pancreatitis. Photodiagnosis Photodyn Ther 2025; 52:104522. [PMID: 39961459 DOI: 10.1016/j.pdpdt.2025.104522] [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: 01/19/2025] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND This study aimed to show the changes in retinal vascular densities and thicknesses in the peripapillary and macular regions in the acute period in patients with acute pancreatitis (AP). METHODS This prospective cross-sectional study included 57 eyes of 30 patients with AP and 58 eyes of 30 healthy people. Optical coherence tomography angiography (OCTA) was taken within 24-72 h of AP patients' hospitalization. OCTA was used to evaluate the retinal microvascular structure and retinal thickness. RESULTS Peripapillary retinal nerve fiber layer (pp-RNFL) and perifoveolar ganglion cell complex (pef-GCC) thickness in patients with AP were significantly higher than in the healthy control group (p = 0.020 and p = 0.039, respectively). While whole image vessel density (wiVD) and perifoveal vessel density (pefVD) were significantly lower in the deep capillary plexus (DCP), choriocapillaris flow area (CCFA) in the macula were significantly lower in each of the 1 mm and 3 mm radius areas in patients with AP (p = 0.014, p = 0.011; p = 0.011, and p = 0.035 respectively). In the univariable and multivariable linear regression analysis, it was observed that serum lipase and procalcitonin levels affected the thickness of pp-RNFL and pef-GCC (for pp-RNFL, β= 0.001, p = 0.002, β=24.992, p < 0.001, and for pef-GCC, β= 0.001 p = 0.014, β=17.107 p < 0.001 respectively). CONCLUSIONS There are significant microvascular and structural changes in the optic nerve and macula in patients with AP. The relationship between these changes and serum lipase and procalcitonin levels was shown. Clinicians should consider ocular involvement in AP patients with high serum lipase and procalcitonin levels.
Collapse
Affiliation(s)
| | - Harun Erdal
- Department of Gastroenterology, Ankara Etlik City Hospital, Ankara, Turkey.
| |
Collapse
|
42
|
Amiral J, Ferol R. Update on the measurement of "soluble angiotensin converting enzyme 2" in plasma and its emerging significance as a novel biomarker of cardiovascular and kidney diseases: A concise commentary. Transfus Apher Sci 2025; 64:104090. [PMID: 39923730 DOI: 10.1016/j.transci.2025.104090] [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] [Indexed: 02/11/2025]
Abstract
Angiotensin Converting Enzyme 2 has emerged as a major cell-surface enzyme receptor for controlling the Renin-Angiotensin-Aldosterone-System. The SARS-Cov-2 pandemics has focused a major interest on that cell-surface receptor. It is the virus entry door for cell infection, and when inside it can replicate and lead to cell destruction. In some physio-pathological conditions, ADAM 17 and TMPSSR2 enzymes can cleave ACE2 on the cell surface and release its extra-cellular domain into the blood circulation. Measurement of this soluble protein then becomes possible, preferentially in plasma, but also in serum. Clinical studies have shown that Soluble ACE2 is an emerging biomarker for cardiovascular and kidney diseases and it could be of prognostic value for heart failure and kidney dysfunctions. In Covid-19 its diagnostic value is controversial, and the various studies lead to different conclusions. Many laboratory assays have been reported for the measurement of this biomarker. They concern enzymatic assays, aptamer methods, or immunoassays, either chemiluminescent or ELISA. Normal and pathological plasma concentrations reported with the various assays yet lack standardization and are very heterogenous. Recently introduced immunoassays tend to yield more compliant results despite variations due to the assay design and calibration, or the antibody targeted epitopes and reactivity. This article reports an ELISA designed with affinity purified rabbit polyclonal antibodies, obtained with recombinant ACE2 and calibrated with the recombinant protein in plasma. This assay has a global reactivity with the various ACE2 protein epitopes. Assay performance characteristics, and values measured in normal populations are presented. Availability of optimized ELISAs can contribute to a better harmonization of sACE2 measurements in plasma, and confirm its clinical significance as biomarker.
Collapse
Affiliation(s)
- Jean Amiral
- Scientific Hemostasis, Franconville, France.
| | - Rémy Ferol
- Scientific Hemostasis, Franconville, France
| |
Collapse
|
43
|
Wang X, Qiu Y, Di Y, Shaohua H, Wu W, Wang W, Liu H, Li P. Potential causal association between gut microbiota, inflammatory cytokines, and acute pancreatitis: A Mendelian randomization study. JOURNAL OF INTENSIVE MEDICINE 2025; 5:185-192. [PMID: 40241835 PMCID: PMC11997579 DOI: 10.1016/j.jointm.2024.10.004] [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: 07/18/2024] [Revised: 09/27/2024] [Accepted: 10/23/2024] [Indexed: 04/18/2025]
Abstract
Background Acute pancreatitis (AP) ranks among the most frequently encountered gastrointestinal diseases in the emergency department. Recent studies have increasingly emphasized the substantial connection among gut microbiota, inflammatory cytokines, and AP. Methods A two-sample Mendelian randomization (MR) study was conducted using summary statistics of gut microbiota (GM) from the largest available meta-analysis of genome-wide association studies conducted by the MiBioGen consortium (n=18,340). For cytokines, the data were obtained from a study that investigated genome variant associations with 41 inflammatory cytokines and growth factors (n=8293). The summary statistics of AP were obtained from the FinnGen consortium version R5 data (3022 cases and 195,144 controls). The inverse variance weighted (IVW) method was used as the main analysis, with MR-Egger and weighted median as complementary analytical methods. Sensitivity analyses were performed using Cochran's Q-test, MR-Egger intercept test, leave-one-out analyses, and MR-PRESSO. In addition, we employed the reverse MR analysis and MR Steiger method to estimate the orientations of exposure and outcome. Result Among the 211 examined GM taxa, the IVW method revealed that Bacteroidales (odds ratio [OR]=1.412, 95% confidence interval [CI]:1.057 to 1.885, P=0.019), Eubacterium fissicatena group (OR=1.240, 95% CI:1.045 to 1.470, P=0.014), and Coprococcus3 (OR=1.481, 95 % CI:1.049 to 2.090, P=0.026) exhibited a positive association with AP. Conversely, Prevotella9 (OR=0.821, 95% CI:0.680 to 0.990, P=0.038), RuminococcaceaeUCG004 (OR=0.757, 95% CI:0.577 to 0.994, P=0.045), and Ruminiclostridium6 (OR=0.696, 95% CI:0.548 to 0.884, P=0.003) displayed a negative correlation with AP. Among the 41 inflammatory cytokines, only macrophage colony-stimulating factor (M_CSF, OR=0.894, 95% CI:0.847 to 0.943, P=0.037) exhibited a negative association with AP. Sensitivity analyses revealed no evidence of pleiotropy or heterogeneity. Nevertheless, the mediation analysis showed that M_CSF did not act as a mediating factor. Conclusion This two-sample MR study revealed causal associations between specific GM and inflammatory cytokines with AP, respectively. However, inflammatory cytokines did not appear to act as mediating factors in the pathway from GM to AP.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Qiu
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Di
- Intensive Care Unit, Shaanxi Provincial Rehabilitation Hospital, Xi'an, Shaanxi, China
| | - Hou Shaohua
- School of Life Sciences and Medicine, Northwest University, Xi ‘an, Shaanxi, China
| | - Wei Wu
- Department of Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Weiyi Wang
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Liu
- Department of Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| | - Pu Li
- Department of Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
44
|
Pessenda G, Ferreira TR, Paun A, Kabat J, Amaral EP, Kamenyeva O, Gazzinelli-Guimaraes PH, Perera SR, Ganesan S, Lee SH, Sacks DL. Kupffer cell and recruited macrophage heterogeneity orchestrate granuloma maturation and hepatic immunity in visceral leishmaniasis. Nat Commun 2025; 16:3125. [PMID: 40169598 PMCID: PMC11961706 DOI: 10.1038/s41467-025-58360-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: 03/13/2024] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
In murine models of visceral leishmaniasis (VL), the parasitization of resident Kupffer cells (resKCs) drives early Leishmania infantum growth in the liver, leading to granuloma formation and subsequent parasite control. Using the chronic VL model, we demonstrate that polyclonal resKCs redistributed to form granulomas outside the sinusoids, creating an open sinusoidal niche that was gradually repopulated by monocyte-derived KCs (moKCs) acquiring a tissue specific, homeostatic profile. Early-stage granulomas predominantly consisted of CLEC4F+KCs. In contrast, late-stage granulomas led to remodeling of the sinusoidal network and contained monocyte-derived macrophages (momacs) along with KCs that downregulated CLEC4F, with both populations expressing iNOS and pro-inflammatory chemokines. During late-stage infection, parasites were largely confined to CLEC4F-KCs. Reduced monocyte recruitment and increased resKCs proliferation in infected Ccr2-/- mice impaired parasite control. These findings show that the ontogenic heterogeneity of granuloma macrophages is closely linked to granuloma maturation and the development of hepatic immunity in VL.
Collapse
MESH Headings
- Animals
- Leishmaniasis, Visceral/immunology
- Leishmaniasis, Visceral/parasitology
- Leishmaniasis, Visceral/pathology
- Kupffer Cells/immunology
- Liver/parasitology
- Liver/immunology
- Liver/pathology
- Granuloma/immunology
- Granuloma/parasitology
- Granuloma/pathology
- Macrophages/immunology
- Macrophages/parasitology
- Mice
- Leishmania infantum/immunology
- Mice, Inbred C57BL
- Lectins, C-Type/metabolism
- Lectins, C-Type/genetics
- Disease Models, Animal
- Female
- Mice, Knockout
- Receptors, CCR2/metabolism
- Receptors, CCR2/genetics
- Monocytes/immunology
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Gabriela Pessenda
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tiago R Ferreira
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea Paun
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Eduardo P Amaral
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Pedro Henrique Gazzinelli-Guimaraes
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Microbiology, Immunology & Tropical Medicine School of Medicine & Health Sciences. The George Washington University, Washington DC, USA
| | - Shehan R Perera
- Department of Electrical and Computer Engineering, The Ohio State University, Columbus, OH, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Sang Hun Lee
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David L Sacks
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
45
|
Xu W, Hu L, Shi S, Gao J, Ye J, Lu Y. Prediction of Potential Drugs Targeting Acute Pancreatitis Based on the HLA-DR-Related Gene-Monocyte Infiltration Regulatory Network. Biomed Eng Comput Biol 2025; 16:11795972251328458. [PMID: 40165943 PMCID: PMC11956513 DOI: 10.1177/11795972251328458] [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: 10/02/2024] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Background Acute pancreatitis (AP) is a common disease of acute abdominal pain, the incidence of which is increasing annually, but its pathogenesis remains incompletely understood. Methods Gene expression profiles of AP were obtained from the Gene Expression Omnibus (GEO) database. R software was used to identify differentially expressed genes (DEGs) and perform functional analysis. The diagnostic value of HLA-DR-related genes was assessed by receiver operating characteristic (ROC) curves. Monocyte infiltration abundance in AP and normal groups was analyzed by Cibersort method, and the correlation between HLA-DR-related genes and monocyte abundance was analyzed. The modules highly correlated with HLA-DR-related genes were clarified by WGCNA modeling, and the core genes regulating HLA-DR were obtained by using LASSO regression. Finally, potential drugs targeting the above genes were analyzed by Enrichr database. Result A Total of 3 HLA-DR-related genes (HLA-DRA, HLA-DRB1, and HLA-DRB5) were identified, which were negatively correlated with the severity of AP and had excellent disease diagnostic value (AUC = 0.761, 0.761, and 0.718), were were positively correlated with monocyte abundance. We identified 110 genes that positively regulate HLA-DR and 130 genes that negatively regulate HLA-DR. LASSO regression identified UCP2, GK, and SAMHD1 as the core nodes of the regulated genes. Compared with the normal group, UCP2 and SAMHD1 were reduced in AP, and the opposite was true for GK, and SAMHD1 had better sensitivity and specificity in diagnosing AP. Drug sensitivity analysis predicted 12 drugs acting on HLA-DRA, HLA-DRB1, and HLA-DRB5 and 8 drugs acting on UCP2, GK, and SAMHD1. Conclusion We identified 3 HLA-DR-related genes (HLA-DRA, HLA-DRB1, and HLA-DRB5) and 3 coregulatory nodes (UCP2, GK, and SAMHD1), which were associated with AP severity and monocyte abundance. Based on these genes, we predicted 20 potential therapeutic agents for AP.
Collapse
Affiliation(s)
- Wei Xu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Hu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shengyi Shi
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Gao
- Division of Critical Care, Nanxiang Hospital of Jiading District, Shanghai, China
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Côte d’Azur University, Shanghai, China
- The State Key Laboratory of Medical Genomics, Pôle Sino-Français de Recherche en Sciences Du Vivant et Génomique, Shanghai, China
| | - Yiming Lu
- Department of Emergency, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Division of Critical Care, Nanxiang Hospital of Jiading District, Shanghai, China
- The State Key Laboratory of Medical Genomics, Pôle Sino-Français de Recherche en Sciences Du Vivant et Génomique, Shanghai, China
| |
Collapse
|
46
|
Boggio V, Gonzalez CD, Zotta E, Ropolo A, Vaccaro MI. VMP1 Constitutive Expression in Mice Dampens Pancreatic and Systemic Histopathological Damage in an Experimental Model of Severe Acute Pancreatitis. Int J Mol Sci 2025; 26:3196. [PMID: 40243995 PMCID: PMC11988950 DOI: 10.3390/ijms26073196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Acute pancreatitis (AP) an inflammatory condition caused by the premature activation of pancreatic proteases, leads to organ damage, systemic inflammation, and multi-organ failure. Severe acute pancreatitis (SAP) has high morbidity and mortality, affecting the liver, kidneys, and lungs. Autophagy maintains pancreatic homeostasis, with VMP1-mediated selective autophagy (zymophagy) preventing intracellular zymogen activation and acinar cell death. This study examines the protective role of VMP1 (Vacuole Membrane Protein 1)-induced autophagy using ElaI-VMP1 transgenic mice in a necrohemorrhagic SAP model (Hartwig's model). ElaI-VMP1 mice show significantly reduced pancreatic injury, including lower necrosis, edema, and inflammation, compared to wild-type (WT) mice. Biochemical markers (lactate dehydrogenase-LDH-, amylase, and lipase) and histopathology confirm that VMP1 expression mitigates pancreatic damage. Increased zymophagy negatively correlates with acinar necrosis, reinforcing its protective role. Beyond the pancreas, ElaI-VMP1 mice exhibit preserved liver, kidney, and lung histology, indicating reduced systemic organ damage. The liver maintains normal architecture, kidneys show minimal tubular necrosis, and lung inflammation features are reduced compared to WT mice. Our results confirm that zymophagy functions as a protective pathophysiological mechanism against pancreatic and extrapancreatic tissue injury in SAP. Further studies on the mechanism of VMP1-mediated selective autophagy in AP are necessary to determine its relevance and possible modulation to prevent the severity of AP.
Collapse
Affiliation(s)
- Veronica Boggio
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
| | - Claudio Daniel Gonzalez
- Centro de Educación Medica e Investigaciones Clínicas (CEMIC), Hospital Universitario Saavedra, Buenos Aires 1431, Argentina;
| | - Elsa Zotta
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
| | - Alejandro Ropolo
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
| | - Maria Ines Vaccaro
- Instituto de Bioquímica y Biología Molecular Prof. Alberto Boveris, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad de Buenos Aires, Buenos Aires 1113, Argentina; (V.B.); (E.Z.)
- Centro de Educación Medica e Investigaciones Clínicas (CEMIC), Hospital Universitario Saavedra, Buenos Aires 1431, Argentina;
| |
Collapse
|
47
|
Munteanu C, Galaction AI, Onose G, Turnea M, Rotariu M. Hydrogen Sulfide (H 2S- or H 2S n-Polysulfides) in Synaptic Plasticity: Modulation of NMDA Receptors and Neurotransmitter Release in Learning and Memory. Int J Mol Sci 2025; 26:3131. [PMID: 40243915 PMCID: PMC11988931 DOI: 10.3390/ijms26073131] [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: 02/09/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Hydrogen sulfide (H2S) has emerged as a pivotal gaseous transmitter in the central nervous system, influencing synaptic plasticity, learning, and memory by modulating various molecular pathways. This review examines recent evidence regarding how H2S regulates NMDA receptor function and neurotransmitter release in neuronal circuits. By synthesizing findings from animal and cellular models, we investigate the impacts of enzymatic H2S production and exogenous H2S on excitatory synaptic currents, long-term potentiation, and intracellular calcium signaling. Data suggest that H2S interacts directly with NMDA receptor subunits, altering receptor function and modulating neuronal excitability. Simultaneously, H2S promotes the release of neurotransmitters such as glutamate and GABA, shaping synaptic dynamics and plasticity. Furthermore, reports indicate that disruptions in H2S metabolism contribute to cognitive impairments and neurodegenerative disorders, underscoring the potential therapeutic value of targeting H2S-mediated pathways. Although the precise mechanisms of H2S-induced changes in synaptic strength remain elusive, a growing body of evidence positions H2S as a significant regulator of memory formation processes. This review calls for more rigorous exploration into the molecular underpinnings of H2S in synaptic plasticity, paving the way for novel pharmacological interventions in cognitive dysfunction.
Collapse
Affiliation(s)
- Constantin Munteanu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
| | - Anca Irina Galaction
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Gelu Onose
- Neuromuscular Rehabilitation Clinic Division, Clinical Emergency Hospital “Bagdasar-Arseni”, 041915 Bucharest, Romania;
- Faculty of Medicine, University of Medicine and Pharmacy “Carol Davila”, 020022 Bucharest, Romania
| | - Marius Turnea
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| | - Mariana Rotariu
- Department of Biomedical Sciences, Faculty of Medical Bioengineering, University of Medicine and Pharmacy “Grigore T. Popa”, 700454 Iasi, Romania; (A.I.G.); (M.R.)
| |
Collapse
|
48
|
Kordana N, Johnson A, Quinn K, Obar JJ, Cramer RA. Recent developments in Aspergillus fumigatus research: diversity, drugs, and disease. Microbiol Mol Biol Rev 2025; 89:e0001123. [PMID: 39927770 PMCID: PMC11948498 DOI: 10.1128/mmbr.00011-23] [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] [Indexed: 02/11/2025] Open
Abstract
SUMMARYAdvances in modern medical therapies for many previously intractable human diseases have improved patient outcomes. However, successful disease treatment outcomes are often prevented due to invasive fungal infections caused by the environmental mold Aspergillus fumigatus. As contemporary antifungal therapies have not experienced the same robust advances as other medical therapies, defining mechanisms of A. fumigatus disease initiation and progression remains a critical research priority. To this end, the World Health Organization recently identified A. fumigatus as a research priority human fungal pathogen and the Centers for Disease Control has highlighted the emergence of triazole-resistant A. fumigatus isolates. The expansion in the diversity of host populations susceptible to aspergillosis and the complex and dynamic A. fumigatus genotypic and phenotypic diversity call for a reinvigorated assessment of aspergillosis pathobiological and drug-susceptibility mechanisms. Here, we summarize recent advancements in the field and discuss challenges in our understanding of A. fumigatus heterogeneity and its pathogenesis in diverse host populations.
Collapse
Affiliation(s)
- Nicole Kordana
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Angus Johnson
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Katherine Quinn
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Joshua J. Obar
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Robert A. Cramer
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| |
Collapse
|
49
|
de Oliveira THC, Gonçalves GKN. Liver ischemia reperfusion injury: Mechanisms, cellular pathways, and therapeutic approaches. Int Immunopharmacol 2025; 150:114299. [PMID: 39961215 DOI: 10.1016/j.intimp.2025.114299] [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: 12/16/2024] [Revised: 01/28/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Liver ischemia-reperfusion injury (LIRI) is a critical challenge in liver transplantation, resection, and trauma surgeries, leading to significant hepatic damage due to oxidative stress, inflammation, and mitochondrial dysfunction. This review explores the cellular and molecular mechanisms underlying LIRI, focusing on ATP depletion, mitochondrial dysfunction, and the involvement of reactive oxygen species (ROS). Inflammatory pathways, including the activation of nuclear factor-kappa B (NF-κB) and the NLRP3 inflammasome, as well as pro-inflammatory cytokines such as TNF-α and IL-1β, play a crucial role in exacerbating tissue damage. Various types of cell death, including necrosis, apoptosis, necroptosis, pyroptosis, ferroptosis and cuproptosis are also discussed. Therapeutic interventions targeting these mechanisms, such as antioxidants, anti-inflammatories, mitochondrial protectors, and signaling modulators, have shown promise in pre-clinical studies. However, translating these findings into clinical practice faces challenges due to the limitations of animal models and the complexity of human responses. Emerging therapies, such as RNA-based treatments, genetic editing, and stem cell therapies, offer potential breakthroughs in LIRI management. This review highlights the need for further research and the development of innovative therapeutic approaches to improve clinical outcomes.
Collapse
|
50
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|