1
|
Nasrine A, Mohanto S, Narayana S, Ahmed MG. Enhanced pharmacokinetic approach for anastrozole via macromolecule-based silk fibroin nanoparticles incorporated in situ injectables for oestrogen-positive breast cancer therapy. J Drug Target 2025; 33:793-803. [PMID: 39754343 DOI: 10.1080/1061186x.2024.2449486] [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: 10/05/2024] [Revised: 12/26/2024] [Accepted: 12/29/2024] [Indexed: 01/06/2025]
Abstract
Breast cancer (BC) is a substantial reason for cancer-related mortality among women across the globe. Anastrozole (ANS) is an effective orally administered hormonal therapy for oestrogen-positive (ER+) BC treatment. However, several side effects and pharmacokinetic limitations restricted the uses of ANS in BC therapy. Therefore, this investigation developed an in situ gelling injectable-loaded silk fibroin (SF)-ANS NPs, which offers sustained drug release and improved pharmacokinetic properties compared to conventional oral formulations at the targeted site. The optimised in situ gel (ISG) incorporated SF-ANS-NPs were developed, and the pharmacokinetic parameters were accessed in subcutaneous administration of NMU-induced Wistar albino rats. The results demonstrated that SF-ANS-NP-ISG exhibited a significantly higher Cmax, Tmax, and AUC compared to pure ANS suspension. In addition, tumour multiplicity (1.40 ± 0.66), tumour latency (75 ± 9.2 days), and incidence rate (90 ± 2.1%) were recorded, and post-treatment analysis reported a marked reduction in tumour volume and weight compared to positive control within 90 days of a single dose. The SF-ANS-NP-ISG treated group's histopathological assessment indicated a low-grade carcinoma, reduced epithelial hyperplasia, and haemorrhage in mammary tumour tissues compared to positive control. Thus, the SF-ANS-NPs-ISG investigated to overcome the pharmacokinetic limitations of ANS further exhibited targeted delivery and bioavailability compared to conventional dosage forms.
Collapse
Affiliation(s)
- Arfa Nasrine
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Soumya Narayana
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| |
Collapse
|
2
|
Shahrahmani F, Badamchizadeh S, Kaihani F, Alavi-Moghadam S, Keshtkari S, Rezaei-Tavirani M, Arjmand R, Larijani B, Arjmand B. Platinum-based chemotherapies-induced nephrotoxicity: mechanisms, potential treatments, and management. Int Urol Nephrol 2025; 57:1563-1583. [PMID: 39630371 DOI: 10.1007/s11255-024-04303-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 11/20/2024] [Indexed: 04/17/2025]
Abstract
Platinum-based chemotherapies are essential in the treatment of several malignancies. However, such medications can damage the kidneys, frequently leading to both acute and chronic kidney disease. Treatment becomes more difficult for such problems. Physicians may alter chemotherapy regimens and utilize kidney-protecting medications to lessen renal damage. New imaging techniques and biomarkers also aid in the early detection of renal issues. To effectively handle the mentioned situation, oncologists, nephrologists, and pharmacists must collaborate. However, additional study is still required to develop customized therapies, discover strategies to minimize kidney injury and produce new platinum medicines. Hereupon, the present review's authors are being sought to address the causes, prospective treatments, and management of nephrotoxicity caused by platinum-based chemotherapy.
Collapse
Affiliation(s)
- Fatemeh Shahrahmani
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sana Badamchizadeh
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Keshtkari
- Department of Internal Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Aghasizadeh M, Moghaddam T, Bahrami AR, Sadeghian H, Alavi SJ, Kazemi T, Matin MM. Evaluation of several farnesyloxycarbostyril derivatives as potential 15-LOX-1 inhibitors for prostate cancer treatment. Toxicol Appl Pharmacol 2025; 498:117293. [PMID: 40057000 DOI: 10.1016/j.taap.2025.117293] [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: 12/12/2024] [Revised: 02/20/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025]
Abstract
The impact of 15-lipoxygenase-1 (15-LOX-1) in the progression of prostate cancer (PCa) is noteworthy, as it correlates with the Gleason score of the disease. Thus, development of specific 15-LOX-1 inhibitors would be desirable for targeted therapy of PCa. This study focused on evaluating the anti-prostate cancer potency of three farnesyloxycarbostyril derivatives, 6-, 7- and 8-farnesyloxycarbostyril (6-, 7- and 8-FQ), as potential inhibitors of 15-LOX-1 on PCa cells. To this end, the enzymatic activity of 15-LOX was first assessed in PCa and human dermal fibroblast (HDF) cells. Subsequently, the cytotoxic effects and apoptosis-inducing capabilities of the compounds were assessed through MTT assay and FITC-annexin V/PI staining, respectively. Among the compounds, 8-FQ was selected for further assessment in a mouse model bearing xenograft human PCa tumor. The results demonstrated that the most effective compound, 8-FQ, caused an 84-fold and 15.7-fold reduction in 15-LOX activity in PC-3 cells at 30 and 14 μM concentrations, respectively. The MTT assay revealed a dose- and time-dependent toxicity of the compounds on PCa cells, and flow cytometry results indicated that apoptosis served as the dominant mechanism of cell death. Given the upregulation of 15-LOX-1 in human PCa cells, the study concludes that the heightened sensitivity to 8-FQ is likely associated with elevated levels of 15-LOX-1. In vivo experiments using immunosuppressed C57BL/6 mice bearing human PC-3 tumors revealed that 8-FQ, at a dosage of 10 mg/kg, exhibited strong antitumor effects with minimal side effects, indicating its potential as a promising therapeutic agent for PCa following further optimization.
Collapse
Affiliation(s)
- Mehrdad Aghasizadeh
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Tayebe Moghaddam
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Ahmad Reza Bahrami
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Industrial Biotechnology Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hamid Sadeghian
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Jamal Alavi
- Department of Laboratory Sciences, School of Paramedical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tahmineh Kazemi
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran; Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran.
| |
Collapse
|
4
|
Wang X, Tian S, Qu Z, Meng R, Ni G, Liu M, Cao H. Investigating the protective effect of hydroxylated fullerenes on cognitive function in rats with temporal lobe epilepsy. Sci Rep 2025; 15:14142. [PMID: 40269130 DOI: 10.1038/s41598-025-99259-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025] Open
Abstract
The objective of this study was to explore the protective effects of hydroxy fullerenes (HFs) on cognitive function in rats with temporal lobe epilepsy (TLE) and to elucidate the underlying mechanisms. Eighteen Sprague-Dawley (SD) rats were randomly selected and administered pilocarpine (50 mg/kg) intraperitoneally to establish a TLE model, and were then randomly assigned to the TLE group and the TLE + HFs group. An additional nine SD rats were served as a normal control group (CON group). The Morris water maze (MWM) test was utilized to assess the spatial learning and memory capabilities of the rats. Nissl staining was employed to observe the survival neurons in the CA1 and CA3 regions. In addition, the ultrastructure of synapses in the CA1 region was examined using transmission electron microscopy (TEM). The expressions of postsynaptic densitin-95 (PSD-95) and synaptophysin (SYP) in the hippocampus were detected via western blotting. The findings revealed that compared to the CON group, the TLE group exhibited significantly prolonged escape latency, reduced platform crossing frequency, and shortened time spent in the target quadrant. The number of surviving neurons in the CA1 and CA3 regions and the expression of PSD95 and SYP protein were significantly decreased (P < 0.05 or P < 0.001). However, these alterations were reversed in the TLE + HFs group. It is suggested that HFs may enhance the spatial learning and memory ability of TLE rats by preserving the integrity of hippocampal neurons, up-regulating the expression of SYP and PSD95 in hippocampus.
Collapse
Affiliation(s)
- Xiaoqing Wang
- The Department of Rehabilitation, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Shuang Tian
- The Department of Neurology, Shijiazhuang People's Hospital, Shijiazhuang, Hebei, People's Republic of China
| | - Zhenzhen Qu
- The Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Ran Meng
- The Department of Rehabilitation, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Guangxiao Ni
- The Department of Rehabilitation, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Min Liu
- The Department of Rehabilitation, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China
| | - Huifang Cao
- The Department of Rehabilitation, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China.
| |
Collapse
|
5
|
Cinar I, Yayla M, Bayram P, Binnetoğlu D. Comparison of the neuroprotective effects of gossypin on cisplatin-induced neurotoxicity in vitro and in vivo. Mol Biol Rep 2025; 52:412. [PMID: 40261449 DOI: 10.1007/s11033-025-10501-7] [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: 12/24/2024] [Accepted: 04/08/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Cisplatin (CIS), a widely used chemotherapeutic agent, is known for its severe neurotoxic side effects. This study investigates the neuroprotective effects of gossypin (GOS), a bioflavonoid with antioxidant and anti-inflammatory properties, against CIS-induced neurotoxicity. METHODS AND RESULTS In-vitro and in-vivo experiments were conducted.50 male Mus Musculus mice were divided into five groups (n:10). Gossypin was administered at varying doses to (in-vitro: 50, 75, 100 µM; in vivo: 5, 10, 20 mg/kg/day) evaluate its protective effects. Results showed gossypin significantly improved cell viability dose-dependently, normalized oxidative stress markers Superoxide dismutase (SOD) activity and glutathione (GSH) and malondialdehyde (MDA) levels, reduced pro-apoptotic and inflammatory genes (CASP-3, CASP-9, TNF-α, NF-kB, iNOS), and increased anti-apoptotic markers (BCL2/BAX ratio, nNOS). Histopathological analysis revealed gossypin mitigated CIS-induced brain tissue damage. CONCLUSIONS Gossypin exhibited significant neuroprotective effects against CIS-induced neurotoxicity via antioxidant (TNF-α), anti-inflammatory (NF-kB), and anti-apoptotic (BCL2/BAX ratio, nNOS) mechanisms by regulating various key regulatory genes, suggesting that it may be a promising adjuvant therapy to protect against the neurotoxic side effects of cisplatin in cancer treatment.
Collapse
Affiliation(s)
- Irfan Cinar
- Faculty of Medicine, Department of Pharmacology, Kastamonu University, Kastamonu, 3700, Turkey.
| | - Muhammed Yayla
- Faculty of Medicine, Department of Pharmacology, Selçuk University, Konya, Turkey
| | - Pınar Bayram
- Faculty of Medicine, Department of Histology and Embriology, Kafkas University, Kars, Turkey
| | - Damla Binnetoğlu
- Faculty of Medicine, Department of Pharmacology, Kafkas University, Kars, Turkey
| |
Collapse
|
6
|
Barnes DA, Janssen MJ, Yang H, Redegeld FA, Masereeuw R. An Adverse Outcome Pathway for DNA adduct formation leading to kidney failure. Toxicology 2025; 515:154162. [PMID: 40268266 DOI: 10.1016/j.tox.2025.154162] [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: 02/12/2025] [Revised: 04/11/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
An Adverse Outcome Pathway (AOP) is a conceptual framework in toxicology and risk assessment that outlines the series of events from a chemical's molecular interaction to the resulting adverse health effect. This framework offers a structured approach to organizing biological knowledge, making it especially useful for understanding the mechanisms through which chemicals cause harm. Following a comprehensive analysis of the literature, an AOP was elucidated for key events linking DNA adduct formation, caused by compounds such as platinum anticancer drugs, to tubular necrosis, resulting in kidney failure. Currently, cisplatin, carboplatin and oxaliplatin are the three most utilised Pt-based drugs used globally for the treatment of cancer. The hydrolysis of platinum anticancer agents post-cellular uptake yields electrophilic intermediates that covalently bind to nucleophilic sites on DNA to form adducts that represent the molecular initiating event. When DNA repair mechanisms become unbalanced, the nephrotoxic response following the formation of DNA adducts leads to DNA damage and mitochondrial dysfunction. These events promote the generation and release of reaction oxygen species (ROS) to induce oxidative stress, causing cell death and inflammation. Upon detachment from the basement membrane, these compromised cells are subsequently deposited in the tubular lumen. Tubular obstruction and inflammatory responses to proximal tubule insult can lead to secondary toxicity and tubular necrosis, further exacerbating kidney injury and precipitating a progressive decline of renal function, finally resulting in kidney failure.
Collapse
Affiliation(s)
- D A Barnes
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, Netherlands
| | - M J Janssen
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, Netherlands
| | | | - F A Redegeld
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, Netherlands
| | - R Masereeuw
- Utrecht University, Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, Netherlands.
| |
Collapse
|
7
|
Tao X, Zhang W, Chen L, Lu S, Li Z, Gao Y, Fan Q, Li J, Wu J, Zhao C. The DHCR7 is the key target of lipotoxic liver injury caused by matrine through abnormal activation of the cholesterol synthesis pathway. Toxicon 2025; 260:108366. [PMID: 40250732 DOI: 10.1016/j.toxicon.2025.108366] [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: 01/21/2025] [Revised: 04/01/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND Matrine, the main active ingredient in Sophora flavescens and Sophorae tonkinensis radix et rhizome, is a highly effective insecticide. However, its hepatotoxicity to some extent affects its application value. This study aimed to explore the mechanism underlying matrine-induced liver injury. METHODS The zebrafish (Danio rerio) and L02 cell model were utilized to investigate the toxic dose of matrine and its effects on liver tissue damage, liver cell morphology and activity, and expression levels of ALT and AST. Zebrafish and L02 cell samples were then collected for transcriptomic testing to further explore the possible mechanism by which matrine induced liver injury. Finally, integrated bioinformatics methods and experiments were used to elucidate the possible mechanisms behind matrine-induced liver injury. RESULTS The result presented solid in vivo evidence of matrine-induced hepatotoxicity, supported by abnormal changes of liver morphological, disturbed liver cell structure, obvious apoptosis, as well as elevated levels of ALT and AST in zebrafish. In addition, in vitro L02 cell experiments also showed that matrine can produce significant liver cell damage effects. The integrated bioinformatics analysis results revealed that differentially expressed genes (DEGs) were substantially enriched in multiple pathways related to lipid regulation. Among which, the steroid biosynthesis was the most key signaling pathway, evidenced by the enhanced expression of eight genes, including DHCR7, SQLE, CYP51, CYP24A1, SC5D, LSS, MSMO1 and SOAT1. Furthermore, AY9944, the targeted inhibitor of DHCR7, could offset the toxic effect, as reflected by diminished liver phenotype damage, steatosis, and cholesterol accumulation caused by matrine. CONCLUSIONS Matrine can upregulate the expression of key genes in steroid biosynthesis pathway, resulting in cholesterol accumulation and then inducing hepatotoxicity. Among them, targeted inhibition of DHCR7 gene expression can alleviate matrine-induced liver injury.
Collapse
Affiliation(s)
- Xiaoyu Tao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Wenting Zhang
- NMPA Key Laboratory for Quality Evaluation of Traditional Chinese Medicine (Traditional Chinese Patent Medicine), Beijing Institute for Drug Control, Beijing, China; Beijing Key Laboratory of Analysis and Evaluation on Chinese Medicine, Beijing Institute for Drug Control, Beijing, China.
| | - Linzhen Chen
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Shan Lu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Zhiqi Li
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yifei Gao
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Qiqi Fan
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jiaqi Li
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China; Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Jiarui Wu
- Department of Clinical Chinese Pharmacy, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China.
| | - Chongjun Zhao
- Beijing Key Laboratory for Quality Evaluation of Chinese Materia Medica, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
8
|
Jiang H, Zhou F, Guo L, Gao Y, Kong N, Xu M, Zhang F. Implications of hippocampal excitatory amino acid transporter 2 in modulating anxiety and visceral pain in a mouse model of inflammatory bowel disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167832. [PMID: 40203955 DOI: 10.1016/j.bbadis.2025.167832] [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: 12/08/2024] [Revised: 03/22/2025] [Accepted: 04/05/2025] [Indexed: 04/11/2025]
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic inflammation and significantly impairs quality of life through anxiety-like behaviors and visceral pain. Early evaluation of the risk of anxiety-like behaviors and visceral pain in IBD patients, along with targeted treatment, may benefit disease management. Visceral pain and anxiety-like behavior are often accompanied by neurological damage. Previous studies have shown that abnormal accumulation of glutamate can cause excitatory neurotoxic effects, leading to central nervous system (CNS) damage. Excitatory amino acid transporters (EAATs), particularly EAAT2, are known to regulate glutamate levels. The impact of hippocampal EAAT2 modulation on these clinical features in IBD is yet to be evaluated. Therefore, we designed this experiment to test this hypothesis. This study aimed to investigate the impact of altered levels of hippocampal EAAT2 on anxiety-like behaviors and visceral pain in mice with IBD. We observed reduced EAAT2 expression, increased glutamate levels, elevated N-methyl-d-aspartate receptors (NMDAR) expression, and obvious glutamate toxicity in the hippocampus of dextran sulfate sodium (DSS) induced IBD model mice. These mice exhibited significant visceral pain and anxiety-like behaviors. In summary, the reduced expression of EAAT2 in the hippocampus of individuals with IBD leads to elevated glutamate levels, resulting in neuronal damage and ultimately contributing to visceral pain and anxiety-like behaviors. These findings suggest that EAAT2 could serve as a therapeutic target for neurologically derived IBD symptoms.
Collapse
Affiliation(s)
- Hao Jiang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310024, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Feini Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Lingnan Guo
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310024, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Yiyuan Gao
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Ning Kong
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China.
| | - Fan Zhang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou 310006, China; Key Laboratory of Digestive Pathophysiology of Zhejiang Province, Hangzhou 310006, China.
| |
Collapse
|
9
|
Sharma P, Garg A, Nidhi, Sharma V. Amelioration of Ulcerative Colitis in BALB/c Mice by Probiotic-Fermented Aegle marmelos Juice. INTERNATIONAL JOURNAL OF FOOD SCIENCE 2025; 2025:5288406. [PMID: 40206440 PMCID: PMC11981707 DOI: 10.1155/ijfo/5288406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/15/2025] [Indexed: 04/11/2025]
Abstract
Aegle marmelos has been used traditionally in folk medicine for the treatment of gastrointestinal (GI) disorders. Fermentation using probiotics is well established to positively modulate the GI system. This study investigated the therapeutic potential of probiotic-fermented Aegle marmelos juice for ulcerative colitis (UC), using a mouse model. UC was induced in mice with dextran sulphate sodium (DSS), leading to weight loss, increased disease activity, and lowered antioxidant defenses. In contrast, mice treated with probiotic-fermented Aegle marmelos juice showed improved body weight, reduced disease activity index, and mitigated colon damage. Inflammatory biomarkers were decreased, while antioxidant activity increased. High-performance liquid chromatography analysis of the fresh and probiotic-fermented Aegle marmelos juice revealed an increase in potential bioactive compounds compared to its unfermented counterpart. These findings suggest that probiotic-fermented Aegle marmelos juice could be a promising therapeutic option for UC, countering inflammation and displaying antioxidant properties.
Collapse
Affiliation(s)
- Pritika Sharma
- Department of Food Technology, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi, India
| | - Aakriti Garg
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Nidhi
- Centre for Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India
| | - Vasudha Sharma
- Department of Food Technology, School of Interdisciplinary Sciences and Technology, Jamia Hamdard, New Delhi, India
| |
Collapse
|
10
|
Zaaba NE, Al-Salam S, Beegam S, Elzaki O, Aldhaheri F, Nemmar A, Ali BH, Nemmar A. Attenuation of cisplatin-induced acute kidney injury by sanguinarine: modulation of oxidative stress, inflammation, and cellular damage. Front Pharmacol 2025; 16:1567888. [PMID: 40242453 PMCID: PMC11999955 DOI: 10.3389/fphar.2025.1567888] [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: 01/28/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
Introduction Cisplatin (CP)-induced acute kidney injury (AKI) is a significant side effect of CP chemotherapy, driven by oxidative stress and inflammation. Sanguinarine (SANG), an alkaloid from the rhizomes of Sanguinaria canadensis and poppy-fumaria species, exhibits antioxidant and anti-inflammatory properties. This study examined SANG's effect on CP-induced AKI in mice and its underlying mechanisms. Methods Mice were orally administered 5 mg/kg SANG for 10 days. On the seventh day, they received a single intraperitoneal CP injection (20 mg/kg) and were sacrificed on the 11th day. Results SANG significantly improved CP-induced decreases in body weight, water intake, urine volume, relative kidney weight, creatinine clearance, albumin-to-creatinine ratio, and plasma urea and creatinine levels. It also reduced elevated plasma neutrophil gelatinase-associated lipocalin, kidney injury molecule-1, cystatin C, and adiponectin levels, as well as renal markers of inflammation and oxidative stress induced by CP administration. SANG normalized kidney mitochondrial dysfunction, DNA damage, and apoptosis caused by CP. It also inhibited the CP-induced increase in the expression of phosphorylated nuclear factor-κB and autophagy markers in the kidney. Histological analysis showed that SANG reduced acute tubular necrosis and intraluminal protein accumulation due to CP. Discussion In conclusion, SANG mitigated CP-induced AKI by reducing inflammation, oxidative stress, DNA damage, apoptosis, and autophagy. Pending more comprehensive pharmacological and toxicological assessments, SANG may be regarded as a potential therapeutic agent for mitigating CP-induced AKI.
Collapse
Affiliation(s)
- Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Fatima Aldhaheri
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Anas Nemmar
- College of Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Badreldin H. Ali
- Emeritus Professor, Department of Pharmacology and Clinical Pharmacy, College of Medicine and Health Science, Sultan Qaboos University, Muscat, Oman
| | - Abderrahim Nemmar
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
11
|
Terabayashi T, Sasaki T, Ishizaki T, Tomo T, Nishida Y, Hanada K. Analysis of accumulation of DNA double-strand breaks in mouse tissues by pulsed-field gel electrophoresis. Toxicol Appl Pharmacol 2025; 497:117278. [PMID: 39986485 DOI: 10.1016/j.taap.2025.117278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/14/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025]
Abstract
Double-strand DNA breaks (DSBs) are among the most cytotoxic DNA lesions, which can lead to chromosomal instability and eventually cell death. The substances that can potentially induce DSB formation are thus regarded as genotoxic. To date, many genotoxicity tests for detecting DNA breaks have been designed. However, there are limited options available for measuring the accumulation of DSBs in vivo. In this study, we aimed to evaluate a method of detecting the DSBs formed by the direct action of genotoxic substances using pulsed-field gel electrophoresis (PFGE). This approach has the advantage of making it easier to distinguish between DSBs and single-strand DNA breaks (SSBs) induced by the direct action of genotoxic substances. To confirm the detection of DSBs using PFGE, we investigated their accumulation after treatment with cis-diamminedichloroplatinum(II) (cisplatin) or γ-rays in mouse organs. The results revealed the successful detection of cisplatin-induced DSB formation in mouse kidney and thymus and γ-ray-induced DSB formation in all organs. We also discuss the advantages of PFGE-based detection of DSBs in vivo.
Collapse
Affiliation(s)
| | - Takako Sasaki
- Departiment of Pharmacology, Faculty of Medicine, Oita University, Japan
| | - Toshimasa Ishizaki
- Departiment of Pharmacology, Faculty of Medicine, Oita University, Japan
| | - Tadashi Tomo
- Advanced Medical Sciences, Faculty of Medicine, Oita University, Japan; Clinical Engineering Research Center, Faculty of Medicine, Oita University, Japan
| | - Yoshihiro Nishida
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Yufu, Oita 879-5593, Japan.
| | - Katsuhiro Hanada
- Advanced Medical Sciences, Faculty of Medicine, Oita University, Japan; Clinical Engineering Research Center, Faculty of Medicine, Oita University, Japan.
| |
Collapse
|
12
|
Pereira IM, Pereira M, Lopes JA, Gameiro J. The gut microbiome as a potential therapeutic target in IgA nephropathy. Nephrol Dial Transplant 2025; 40:641-650. [PMID: 39663206 DOI: 10.1093/ndt/gfae274] [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: 10/18/2024] [Indexed: 12/13/2024] Open
Abstract
Immunoglobulin A nephropathy (IgAN) is the most common primary glomerulonephritis and a leading cause of kidney failure, with limited treatment options available. The pathophysiology of IgAN remains unclear; however, recent studies suggest that genetic, epigenetic and environmental factors play significant roles. There is also strong evidence linking the gut microbiome to the development of IgAN. In this review, we will examine the relationship between the microbiome and the pathogenesis of IgAN, as well as its potential as a target for future therapeutic interventions.
Collapse
Affiliation(s)
- Inês Miguel Pereira
- Clínica Universitária de Nefrologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
| | - Marta Pereira
- Serviço de Nefrologia e Transplantação Renal, ULS Santa Maria, Lisboa, Portugal
| | - José António Lopes
- Clínica Universitária de Nefrologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Serviço de Nefrologia e Transplantação Renal, ULS Santa Maria, Lisboa, Portugal
| | - Joana Gameiro
- Clínica Universitária de Nefrologia, Faculdade de Medicina da Universidade de Lisboa, Lisboa, Portugal
- Serviço de Nefrologia e Transplantação Renal, ULS Santa Maria, Lisboa, Portugal
| |
Collapse
|
13
|
Kaden T, Alonso‐Román R, Stallhofer J, Gresnigt MS, Hube B, Mosig AS. Leveraging Organ-on-Chip Models to Investigate Host-Microbiota Dynamics and Targeted Therapies for Inflammatory Bowel Disease. Adv Healthc Mater 2025; 14:e2402756. [PMID: 39491534 PMCID: PMC12004439 DOI: 10.1002/adhm.202402756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Indexed: 11/05/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic gastrointestinal disease with drastically increasing incidence rates. Due to its multifactorial etiology, a precise investigation of the pathogenesis is extremely difficult. Although reductionist cell culture models and more complex disease models in animals have clarified the understanding of individual disease mechanisms and contributing factors of IBD in the past, it remains challenging to bridge research and clinical practice. Conventional 2D cell culture models cannot replicate complex host-microbiota interactions and stable long-term microbial culture. Further, extrapolating data from animal models to patients remains challenging due to genetic and environmental diversity leading to differences in immune responses. Human intestine organ-on-chip (OoC) models have emerged as an alternative in vitro model approach to investigate IBD. OoC models not only recapitulate the human intestinal microenvironment more accurately than 2D cultures yet may also be advantageous for the identification of important disease-driving factors and pharmacological interventions targets due to the possibility of emulating different complexities. The predispositions and biological hallmarks of IBD focusing on host-microbiota interactions at the intestinal mucosal barrier are elucidated here. Additionally, the potential of OoCs to explore microbiota-related therapies and personalized medicine for IBD treatment is discussed.
Collapse
Affiliation(s)
- Tim Kaden
- Dynamic42 GmbH07745JenaGermany
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
| | - Raquel Alonso‐Román
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | | | - Mark S. Gresnigt
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Junior Research Group Adaptive Pathogenicity StrategiesLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
| | - Bernhard Hube
- Department of Microbial Pathogenicity MechanismsLeibniz Institute for Natural Product Research and Infection Biology – Hans‐Knöll‐Institute07745JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
- Institute of MicrobiologyFaculty of Biological SciencesFriedrich Schiller University07743JenaGermany
| | - Alexander S. Mosig
- Institute of Biochemistry IICenter for Sepsis Control and CareJena University Hospital07747JenaGermany
- Cluster of Excellence Balance of the MicroverseFriedrich Schiller University Jena07745JenaGermany
| |
Collapse
|
14
|
Jash R, Maji HS, Chowdhury A, Maparu K, Seksaria S, Gupta P, Paria A, Nandi A, Das A, Chattaraj B, Prasanth DNK. Oxymatrine ameliorates epithelial mesenchymal transition in IgA nephropathy induced rats. Tissue Cell 2025; 93:102671. [PMID: 39705868 DOI: 10.1016/j.tice.2024.102671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
In this study, we investigated the efficacy of oxymatrine, a phytochemical alkaloid, in reducing inflammation and fibrosis in a rat model of IgA nephropathy (IgAN) through modulation of the TGF-β/SMAD signaling pathway. Thirty Sprague Dawley rats were randomized into control, IgAN, and treatment groups, the latter receiving oxymatrine postinduction of IgAN. Induced by bovine serum albumin, carbon tetrachloride, and lipopolysaccharides, the disease model was validated by immunofluorescence and histopathological analyses, confirming significant renal deposition of IgA and increased fibrosis markers (IL-6, TGF-β, SMAD 3, and α-SMA). Oxymatrine treatment led to a notable decrease in urine protein levels and red blood cells at 10 weeks, suggesting reduced kidney damage. There was no significant impact on the SGOT or SGPT levels, while it reduced the BUN, serum ALB and creatinine levels, indicating minimal hepatotoxicity and renoprotective effects. Histopathology demonstrated preservation of the glomerular diameter in the treatment group. Immunofluorescence and ELISA revealed a reduction in the levels of proinflammatory (IL-6,TNF-α) and profibrotic (TGF-β, SMAD 3, and α-SMA) markers in treated rats, suggesting that oxymatrine has renoprotective effects on the inhibition of pathological EMT processes and fibrosis in IgAN. Our results suggest that oxymatrine is a potential therapeutic agent for IgAN that attenuates disease progression by targeting the TGF-β/SMAD pathways involved in EMT and fibrosis. Further research is warranted to establish long-term efficacy and safety profiles.
Collapse
Affiliation(s)
- Rajiv Jash
- Department of Pharmaceutical Sciences, JIS University, Kolkata 700109, India; Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India.
| | | | - Arnab Chowdhury
- Department of Pathology, Shri Ramkrishna Institute of Medical Sciences and Sanaka Hospital, Malandighi, Durgapur, West Bengal 713212, India
| | - Kousik Maparu
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Sanket Seksaria
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Priyanka Gupta
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Arghya Paria
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Arijit Nandi
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Anwesha Das
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Bornika Chattaraj
- Department of Pharmacy, Sanaka Educational Trust's Group of Institutions, Malandighi, Durgapur, West Bengal 713212, India
| | - Ds Nb K Prasanth
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Polepally SEZ, TSIIC, Jadcherla, Mahbubnagar, Hyderabad 509301, India
| |
Collapse
|
15
|
Gorodetsky R, Lazmi Hailu A, Volinsky E, Adani B, Pappo O, Israeli E. The Use of Potent Populations of Expanded Fetal Human Placental Stromal Cells for the Treatment of Dextran Sodium Sulfate-Induced Colitis in a Mouse Model. Int J Mol Sci 2025; 26:3222. [PMID: 40244025 PMCID: PMC11989926 DOI: 10.3390/ijms26073222] [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: 12/26/2024] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Inflammatory Bowel Disease (IBD) is a multifactorial gastrointestinal condition encompassing two major forms of intestinal inflammation: Crohn's disease (CD) and ulcerative colitis (UC). Both conditions are linked to auto-inflammatory reactions and genetic predispositions. Various drug therapies and biological treatments proposed to reduce IBD-associated inflammation. We induced IBD in a mouse model by stimulating bowel inflammation with an oral dextran sodium sulfate (DSS) beverage. Our novel cell therapy approach for IBD involves intramuscular (IM) and intraperitoneal (IP) delivery of non-matched, expanded, potent xenogeneic fetal human mesenchymal stromal cells (f-hPSCs) in 2 × 106 cell injections. This cell therapy has already been shown previously to induce pro-regenerative and anti-inflammatory effects in different systemic and local disorders, where the injected f-hPSCs were shown to respond to the stress of the host and secrete the adequate secretome in response to this stress. In the current study, the IP-injected f-hPSCs treatment of the DSS-induced IBD enhanced the regenerative processes of the damaged bowel and reduced the inflammatory process. This was associated with rapid regain of the mice's weight and a decrease in inflammation-associated parameters, such as colon edema, bowel shortening, and a threefold increase in bowel mass, as estimated by increased colon weight and reduced length. This ratio best emphasized the induced inflammatory response associated with the decrease in the inflamed colon length with an increase in its mass. Although IM f-hPSCs delivery was somehow effective by a few parameters, the IP delivery produced a superior response. The IP f-hPSCs treated mice lost only ~15% of their weight at the peak of the IBD effect, compared to ~25% in untreated mice. A reduction in the inflammatory response of the gut was also indicated by a decrease in neutrophil infiltration, as assayed by a myeloperoxidase (MPO) assay. Additionally, a significant improvement in the histological score of the gut and faster recovery to 90% of its original size was observed. These findings suggest that f-hPSC treatments could serve as an effective and safe anti-inflammatory and pro-regenerative treatment for IBD.
Collapse
Affiliation(s)
- Raphael Gorodetsky
- Laboratory of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (A.L.H.); (E.V.)
| | - Astar Lazmi Hailu
- Laboratory of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (A.L.H.); (E.V.)
| | - Evgenia Volinsky
- Laboratory of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (A.L.H.); (E.V.)
| | - Boaz Adani
- Laboratory of Biotechnology and Radiobiology, Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel; (A.L.H.); (E.V.)
| | - Orit Pappo
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel;
| | - Eran Israeli
- Department of Gastroenterology, Hadassah-Hebrew University Medical Center, Jerusalem 9112001, Israel;
| |
Collapse
|
16
|
Díaz-Catalán D, Capó J, Vega-Beyhart A, Rodrigo-Calvo MT, Mora M, Vidal O, Squarcia M, Enseñat J, Casals G, Hanzu F. Sex-dependent effects of FGF21 on HPA axis regulation and adrenal regeneration after Cushing syndrome in mice. Mol Metab 2025; 96:102122. [PMID: 40154841 PMCID: PMC12013334 DOI: 10.1016/j.molmet.2025.102122] [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: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Cushing's syndrome (CS) results from prolonged exposure to excessive glucocorticoids (GCs), leading to metabolic disturbances and adrenal insufficiency (AI). Fibroblast growth factor 21 (FGF21) has shown promise as a potential therapeutic target for metabolic disorders. This study explores the effects of FGF21 on adrenal gland function in a mouse model of AI following chronic hypercortisolism and investigates sex-dependent differences in the hypothalamic-pituitary-adrenal (HPA) axis response. METHODS We employed a mouse model of AI after chronic corticosterone (CORT) treatment. The effects of recombinant human FGF21 (hFGF21) administration on adrenal function were evaluated in AI mice. Male and female wild-type (WT) and FGF21-overexpressing transgenic (Tg) mice were subjected to 5 weeks of CORT treatment, reaching CS phenotype, followed by immediate analysis or a 10-week recovery period. Metabolic parameters, HPA axis function, and adrenal gland morphology and gene expression were assessed. RESULTS Prolonged CORT exposure resulted in metabolic disturbances and HPA axis dysregulation. hFGF21 treatment increased CORT and ACTH secretion in AI mice. FGF21 overexpression influenced glucose homeostasis and insulin regulation during CORT treatment and recovery, with sex-specific effects. Tissue-specific regulation of Klb expression was observed across the HPA axis, with distinct patterns between males and females. Tg mice displayed altered adrenal progenitor cell activation and steroidogenic gene expression. Sex-specific differences were observed in adrenal capsule remodeling and gene expression patterns during recovery. CONCLUSIONS This study reveals the complex interplay between FGF21 signaling and GC-induced metabolic and endocrine changes, suggesting a potential sex-specific role of FGF21 in metabolic regulation and HPA axis recovery following after CS.
Collapse
Affiliation(s)
- Daniela Díaz-Catalán
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain.
| | - Júlia Capó
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain
| | - Arturo Vega-Beyhart
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain
| | | | - Mireia Mora
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; Endocrinology Department, Hospital Clínic, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
| | - Oscar Vidal
- Endocrine Surgery Department, Hospital Clínic Barcelona, University of Barcelona, Spain
| | - Mattia Squarcia
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; The Biomedical Diagnostic Center (CBD), Barcelona, Spain
| | - Joaquim Enseñat
- Neurosurgery Department, Hospital Clinic Barcelona, University of Barcelona, Spain
| | - Gregori Casals
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; Biochemistry and Molecular Genetics Service, Hospital Clínic, Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Spain
| | - Felicia Hanzu
- Endocrine Disorders Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)/ Hospital Clínic, Barcelona, Spain; Endocrinology Department, Hospital Clínic, Barcelona, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain.
| |
Collapse
|
17
|
Lu L, Hu N, Chen H, Wang S, Deng Y, Lin Z, Wang Z, Zhu X, Liu X, Liu L, Jiang L. Cisplatin-induced acute kidney injury increased brain 5-hydroxytryptamine levels partly due to the hippuric acid-induced upregulation of CYP2D4 expression and function in the brain of rats. Drug Metab Dispos 2025; 53:100068. [PMID: 40245581 DOI: 10.1016/j.dmd.2025.100068] [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: 12/18/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 04/19/2025] Open
Abstract
Patients with acute kidney injury (AKI) are often associated with uremic encephalopathy, but its underlying mechanisms remain unclear. This study aimed to investigate how AKI induced neuropsychiatric disorders through cerebral 5-hydroxytryptamine (5-HT) dysregulation in cisplatin-induced AKI rats. Our findings demonstrated that AKI induced anxiety-like behaviors and increased cerebral 5-HT levels, which may be attributed to the upregulated CYP2D4 expression and activity. The intraventricular injection of quinine (CYP2D4 inhibitor) attenuated the elevated cortical 5-HT levels in AKI rats. Intraperitoneal administration of 5-methoxytryptamine (CYP2D4 substrate) also provoked anxiety-like behaviors and cerebral 5-HT accumulation, which were reversed by cotreatment with quinine. Hippuric acid (HA), as a classical uremic toxin, was severely accumulated in both the plasma and brain of AKI rats. In vitro experiments demonstrated that HA-induced reactive oxygen species (ROS) upregulated expression of CYP2D6 (over 70% homology with rat CYP2D4) via suppressing Nrf2/HO-1 pathway in SH-SY5Y cells. These effects were reversed by ROS scavenger N-acetylcysteine, Nrf2 activator sulforaphane, and HO-1 activator cobalt-protoporphyrin IX. Similarly, either Nrf2 inhibitor ML385 or HO-1 inhibitor zinc-protoporphyrin IX exerted up-regulatory effects on CYP2D6 expression. In vivo studies confirmed that HA treatment induced AKI-like behavioral abnormalities in rats, accompanied by increased cerebral 5-HT levels and CYP2D4 expression as well as induced production of ROS, decreased Nrf2 and HO-1 protein levels. Our findings elaborate a novel mechanism between kidney failure and neuropsychiatric complications. Specifically, cisplatin-induced AKI upregulates CYP2D4 expression via HA-mediated ROS release, subsequently promoting generation of cerebral 5-HT by CYP2D4 and revealing material basis of AKI-associated uremic encephalopathy. SIGNIFICANCE STATEMENT: This study revealed that the psychiatric disorders of cisplatin-induced acute kidney injury rats are partly attributed to the increased 5-hydroxytryptamine levels induced by brain CYP2D. The induction of CYP2D4 is mainly due to brain accumulation of hippuric acid via inactivation of Nrf2/HO-1 pathway by reactive oxygen species.
Collapse
Affiliation(s)
- Lingjue Lu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Nan Hu
- Department of Pharmacy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Haoran Chen
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Siqian Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ying Deng
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zijin Lin
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhongyan Wang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xinyue Zhu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaodong Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Li Liu
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Ling Jiang
- Center of Drug Metabolism and Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
18
|
Xu J, Dong L, Xie X, Geng BD, Lu J, Dong Y, Hu Y, Liu C, Mao Y, Ge G, Ren Z. Human umbilical cord-derived mesenchymal stem cells improve thymus and spleen functions in D-galactose-induced aged mice. Sci Rep 2025; 15:9470. [PMID: 40108399 PMCID: PMC11923087 DOI: 10.1038/s41598-025-94364-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
As aging progresses, the structures and functions of immune organs such as the thymus and spleen deteriorate, leading to impaired immune function and immune senescence. This study investigated the potential of umbilical cord mesenchymal stem cells (UC-MSCs) to mitigate D-galactose-induced immune senescence by enhancing the structural and functional integrity of aging immune organs and regulating the gut microbiota. The findings show that UC-MSCs treatment significantly delayed thymus and spleen atrophy and reduced the number of senescence-associated β-galactosidase (SA-β-gal) positive cells. At the molecular level, UC-MSCs treatment downregulated the expression of aging-related genes, including p16, p53, p21, and RB. It also boosted antioxidant enzyme activity, increasing the levels of catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px), while decreasing serum malondialdehyde (MDA) levels by activating the Nrf2/HO-1 pathway. Additionally, UC-MSCs treatment restored the balance of the gut microbiota. These results demonstrate that UC-MSCs significantly improve the structural and functional integrity of immune organs and enhance the composition of the gut microbiome, offering a potential strategy for delaying immune senescence.
Collapse
Affiliation(s)
- Jianwei Xu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, China
- Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Li Dong
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Xiaofen Xie
- Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
| | - Bill D Geng
- School of Natural Science, University of Texas at Austin, Austin, USA
| | - Junhou Lu
- Translational Medicine Research Center, Guizhou Medical University, Guiyang, China
| | - Yongxi Dong
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yang Hu
- Children's Medical Center, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Can Liu
- Department of Pharmacology, School of Basic Medicine, Guizhou Medical University, Guiyang, China
- Guizhong Biotechnology Co., Ltd., Guiyang, China
| | - Yuanhu Mao
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Guo Ge
- Key Laboratory of Medical Biology, Guizhou Medical University, Ankang Avenue, Gui'an New District, China.
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, Guiyang, China.
| | - Zhenkui Ren
- Clinical Laboratory, Second People's Hospital of Guizhou Province, 206 South Section of Xintian Avenue, Guiyang City, China.
| |
Collapse
|
19
|
Buyukyildirim T, Senol Deniz FS, Tugay O, Salmas RE, Ulutas OK, Aysal IA, Orhan IE. Chromatographic Analysis and Enzyme Inhibition Potential of Reynoutria japonica Houtt.: Computational Docking, ADME, Pharmacokinetic, and Toxicokinetic Analyses of the Major Compounds. Pharmaceuticals (Basel) 2025; 18:408. [PMID: 40143185 PMCID: PMC11944761 DOI: 10.3390/ph18030408] [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: 02/17/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background:Reynoutria japonica Houtt. has been used for inflammatory diseases, skin burns, and high cholesterol in traditional Chinese medicine, and the roots and rhizomes of the plant were registered in the Chinese Pharmacopoeia. This study evaluated the enzyme inhibitory activities of R. japonica extracts from Türkiye. Its major phytochemical content was elucidated, molecular interaction studies of the main compounds were conducted, and toxicokinetic predictions and absorption, distribution, metabolism, and elimination studies were performed with in silico methods. Methods: R. japonica extracts were tested for their enzyme inhibitory activities using an ELISA microplate reader. The phytochemical profile was elucidated by LC-MS QTOF. Docking and other in silico studies evaluated interactions of its main components with cholinesterase, collagenase, and elastase. Results: R. japonica exhibited significant cholinesterase inhibitory effectiveness, while the stem and root extracts showed moderate tyrosinase inhibition. R. japonica leaf (IC50 = 117.20 ± 4.84 g/mL) and flower extracts (IC50 = 111.40 ± 1.45 µg/mL) exhibited considerable elastase activity. R. japonica leaf (IC50 = 171.00 ± 6.76 g/mL) and root (IC50 = 160.00 ± 6.81 g/mL) extracts displayed similar and potent collagenase inhibition. In the LC-MS QTOF analysis, procyanidin dimer, catechin, piceid, torachrysone, and its glucoside isomers were identified as the major components and resveratrol as the minor component. Galloylglucose showed the strongest binding at cholinesterase via key hydrogen bonds, while emodin-6-glucoside and emodin formed stable interactions with elastase. Piceid displayed significant polar and water-mediated contacts with collagenase. These findings underscore the potential of these ligands as protein inhibitors. In silico predictions reveal that emodin possessed the most favorable drug-like properties but posed potential interaction risks. Conclusions: This research represents the first investigation of the bioactivity and phytochemistry of R. japonica grown and documented in 2020 in Türkiye. Our findings point out that R. japonica could be used for cosmetic purposes, and further studies on neurological disorders could be performed.
Collapse
Affiliation(s)
- Tugsen Buyukyildirim
- Department of Pharmacognosy, Faculty of Pharmacy, Selcuk University, 42071 Konya, Türkiye;
| | | | - Osman Tugay
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Selcuk University, 42071 Konya, Türkiye;
| | | | - Onur Kenan Ulutas
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye;
| | - Ibrahim Ayhan Aysal
- Department of Analytical Chemistry, Faculty of Pharmacy, Gazi University, 06330 Ankara, Türkiye;
| | - Ilkay Erdogan Orhan
- Department of Pharmacognosy, Faculty of Pharmacy, Lokman Hekim University, 06510 Ankara, Türkiye
- Principal Member of Turkish Academy of Sciences (TÜBA), Vedat Dalokay Street, No. 112, 06670 Ankara, Türkiye
| |
Collapse
|
20
|
Jeong ES, Jung HK, Choi E, Yun K, Lee A, Kim YS. Characterization of post-inflammatory irritable bowel syndrome animal model following acute colitis recovery. Sci Rep 2025; 15:8512. [PMID: 40075091 PMCID: PMC11904205 DOI: 10.1038/s41598-025-88981-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 02/03/2025] [Indexed: 03/14/2025] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent disorder with an unclear pathophysiology. This study aimed to investigate the features of dextran sulfate sodium (DSS)-induced low-grade inflammation using murine models of acute severe colitis (acute model) and chronic mild repeated colitis (chronic model), with potential implications for IBS research. The acute model was induced with 3% DSS for 5 days, followed by a 12-week recovery period. The chronic model involved administration of 0.5% DSS for 5 days, followed by a 5-day resting period, repeated thrice. We conducted comparative analyses to assess inflammation severity, intestinal motility, permeability, visceral hypersensitivity, and microbiome composition. In the acute model, mild leukocyte infiltration was observed, colonic transit time shortened at 12 weeks (P < 0.001), occludin expression decreased (P = 0.041), inflammatory cytokines, and transient receptor potential vanilloid 1 was upregulated in colonic mucosa (P < 0.050). In the chronic model, only mild inflammatory changes were noted. Microbiota analysis in the acute model revealed differences in microbial abundance and compositions (P = 0.001). The acute model demonstrated low-grade inflammation that caused gut dysmotility, altered permeability, and increased visceral hypersensitivity with notable microbial composition changes, potentially relevant to IBS phenotypes.
Collapse
Affiliation(s)
- Eui Sun Jeong
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea
| | - Hye-Kyung Jung
- Department of Internal Medicine, College of Medicine, Ewha Womans University, Seoul, Korea.
| | - Euno Choi
- Department of Pathology, College of Medicine, Ewha Womans University, Seoul, Korea
| | | | - Ayoung Lee
- Department of Internal Medicine, College of Medicine, Korea University, Ansan, Gyeonggi-do, Korea
| | - Yong Sung Kim
- Digestive Disease Research Institute, College of Medicine, Wonkwang University, Iksan, Jeonlabuk-do, Korea
| |
Collapse
|
21
|
Tian C, Yan M, Guo J, Zhou Y, Du B, Cheng G. Yeast Cell Wall-Mediated Ileal Targeted Delivery System for IgA Nepharopathy Therapy. ACS Biomater Sci Eng 2025; 11:1498-1509. [PMID: 39957538 DOI: 10.1021/acsbiomaterials.4c01941] [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: 02/18/2025]
Abstract
IgA nephropathy (IgAN) is a primary glomerulonephritis mediated by autoimmunity, characterized by an abnormal increase and the deposition of IgA in the glomeruli. In recent years, most studies have emphasized the crucial role of the gut-kidney axis in the pathogenesis of IgA nephropathy, and the ileal Peyer patches in the intestinal mucosal immune system are the main site for IgA production. Therefore, in this study, hydroxychloroquine (HCQ) and dexamethasone (DXM) were used as model drugs, and yeast cell wall (YCW)-coated oleic acid-grafted chitosan (CSO) was used as a carrier to construct a yeast cell wall oral drug delivery system HCQ/DXM@CSO@YCW. This delivery system achieves ileal targeted delivery through the yeast cell wall (YCW), reduces IgA production, and synergistically regulates the inflammatory pathological environment. The delivery system had good gastrointestinal stability and biocompatibility. In vitro cell experiments had shown the targeted uptake ability of dendritic cells and macrophages, and in vitro intestinal experiments showed that the YCW has ileal targeting properties. In vivo pharmacodynamic experiments showed that the HCQ/DXM@CSO@YCW delivery system could significantly reduce the serum IgA levels and IgA deposition in the renal tissue of IgAN mice, as well as the levels of IL-6, TNF-α, and TGF-β in the renal tissue, improving the pathological morphology of the renal tissue. Therefore, the DXM/HCQ@CSO@YCW oral administration system provided a new intestinal targeted delivery platform for intestinal mucosal immunotherapy in IgA nephropathy.
Collapse
Affiliation(s)
- Chaoying Tian
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou, Henan 450001, China
| | - Mei Yan
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou, Henan 450001, China
| | - Jialing Guo
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou, Henan 450001, China
| | - Yingying Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou, Henan 450001, China
| | - Bin Du
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Science Road, Zhengzhou, Henan 450001, China
- Henan Key Laboratory of Nanomedicine for Targeting Diagnosis and Treatment, Zhengzhou, Henan 450001, China
| | - Genyang Cheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
22
|
Seke M, Stankovic A, Zivkovic M. Capacity of fullerenols to modulate neurodegeneration induced by ferroptosis: Focus on multiple sclerosis. Mult Scler Relat Disord 2025; 97:106378. [PMID: 40088719 DOI: 10.1016/j.msard.2025.106378] [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: 12/27/2023] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
Multiple sclerosis is an inflammatory disease of the central nervous system (CNS), characterized by oligodendrocyte loss and demyelination of axons leading to neurodegeneration and severe neurological disability. Despite the existing drugs that have immunomodulatory effects an adequate therapy that slow down or stop neuronal death has not yet been found. Oxidative stress accompanied by excessive release of iron into the extracellular space, mitochondrial damage and lipid peroxidation are important factors in the controlled cell death named ferroptosis, latterly recognized in MS. As the fullerenols exhibit potent antioxidant activity, recent results imply that they could have protective effects by suppressing ferroptosis. Based on the current knowledge we addressed the main mechanisms of the protective effects of fullerenols in the CNS in relation to ferroptosis. Inhibition of inflammation, iron overload and lipid peroxidation through the signal transduction mechanism of Nuclear Factor Erythroid 2-Related Factor 2 (NRF2), chelation of heavy metals and free radical scavenging using fullerenols are proposed as benefitial strategy preventing MS progression. Current review connects ferroptosis molecular targets and important factors of MS progression, with biomedical properties and mechanisms of fullerenols' actions, to propose new treatment strategies that could be addaptobale in other neurodegenerative diseases.
Collapse
Affiliation(s)
- Mariana Seke
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, ˮVinčaˮ Institute of Nuclear Sciences -National Institute of The Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Belgrade 11 000, Serbia.
| |
Collapse
|
23
|
de Freitas KS, Squarisi IS, de Souza LTM, Ozelin SD, de Souza Oliveira LT, Ribeiro VP, Bastos JK, Tavares DC. Evaluation of safety and efficacy of Brazilian brown propolis from Araucaria sp. in preventing colon cancer. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:196-208. [PMID: 39609034 DOI: 10.1080/15287394.2024.2431921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Brazilian propolis produced by honeybees have been widely studied, but few data exist regarding the safety and pharmacological potential of this natural product. The aim of the present study was to examine the toxicity, genotoxicity, and chemoprevention effects attributed to exposure to the brown propolis hydroalcoholic extract (BPHE) of Araucaria sp. Acute oral toxicity test was conducted using Wistar Hannover rats, demonstrating that the highest dose tested (2,000 mg/kg b.w.) produced no apparent adverse effects or lethality. The micronucleus (MN) genotoxicity test was conducted using peripheral blood from Swiss mice, which also noted that BPHE did not induce significant chromosomal damage. It is of interest that BPHE at a dose of 12 mg/kg b.w. exhibited antigenotoxic effects against the doxorubicin (DXR)-induced damage. However, BPHE did not influence the depletion of reduced glutathione induced by DXR in mice. It is noteworthy that BPHE exerted chemopreventive effects at doses 6, 12, and 24 mg/kg b.w. The determination of this effect of BPHE on colon carcinogenesis was examined using aberrant crypt foci (ACF) as evidenced by histological analysis. The colons of animals treated with BPHE (12 mg/kg b.w.) exhibited a significant reduction in staining for proliferating cell nuclear antigen (PCNA) and cyclooxygenase-2 (COX-2) protein following 1,2-dimethylhydrazine (DMH)-and BPHE combined treatments. Hence, it is conceivable that the anti-inflammatory activity of the chemical constituents of BPHE are involved in its chemopreventive action against colon carcinogenesis as evidenced from ACF assay. Therefore, BHPE was found to be a safe product, without any apparent significant acute adverse risk. Further, the extract exhibited antigenotoxic and anticarcinogenic activities which may be considered for beneficial uses in colon carcinogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | - Victor Pena Ribeiro
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jairo Kenupp Bastos
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | |
Collapse
|
24
|
Li J, Wu M, He L. Immunomodulatory effects of mesenchymal stem cell therapy in chronic kidney disease: a literature review. BMC Nephrol 2025; 26:107. [PMID: 40033224 DOI: 10.1186/s12882-025-04029-y] [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: 12/09/2024] [Accepted: 02/19/2025] [Indexed: 03/05/2025] Open
Abstract
Chronic kidney disease (CKD) has been a growing public medical concern in recent years which calls for effective interventions. Mesenchymal stem cells (MSCs) have garnered increased interest in past decades due to their potential to repair and regenerate damaged tissues. Many clinical trials have highlighted the safety and effectiveness of kidney disease with this novel cell therapy. MSC infusion can improve renal function indices such as glomerular filtration rate, urine protein, serum creatinine, and blood urea nitrogen, while inhibiting immune response by increasing regulatory T cells. The therapeutic mechanisms may be primarily attributed to a function combined with immunomodulation, anti-inflammation, anti-fibrosis, promoting angiogenesis, anti-oxidation, anti-apoptosis, or tissue healing produced by cell secretsome. However, CKD is a broad concept due to many pathological etiologies including diabetes, hypertension, heart disease, immunological damage, a family history of renal failure, and so on. Furthermore, the therapeutic efficacy of MSCs may be influenced by different cell sources, injection methods, medication dosage, or homing proportion. As a result, it is timely and essential to access recent advancements in the MSC application on CKD.
Collapse
Affiliation(s)
- Jipeng Li
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, Shaan Xi, China
| | - Mengting Wu
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, Shaan Xi, China
| | - Lijie He
- Department of Nephrology, Xijing Hospital, Air Force Medical University, Xi'an, Shaan Xi, China.
| |
Collapse
|
25
|
Blicharz-Futera K, Kamiński M, Grychowska K, Canale V, Zajdel P. Current development in sulfonamide derivatives to enable CNS-drug discovery. Bioorg Chem 2025; 156:108076. [PMID: 39889550 DOI: 10.1016/j.bioorg.2024.108076] [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: 10/06/2024] [Revised: 12/08/2024] [Accepted: 12/15/2024] [Indexed: 02/03/2025]
Abstract
The encouraging therapeutic potential of sulfonamide-based derivatives has been unraveled by breakthrough discovery of Paul Ehrlich, who pointed out the possibility of fighting microbes with chemicals. Over the decades, the utility of sulfonamides has expanded beyond antimicrobial agents, revealing their usefulness in many areas of pharmacotherapy, including the treatment of central nervous system (CNS) diseases. Through a detailed analysis of preclinical and clinical data, we identify key sulfonamide-based compounds that have demonstrated significant CNS activity. We also discuss the challenges in the development of sulfonamide derivatives as enzyme/ion channel inhibitors or receptor ligands for CNS applications, describing their mode of action and therapeutic significance. This is followed by the characteristics of pharmacological targets, structure-activity relationships, ADMET properties, efficacy in experimental animal models, and outcomes from clinical trials. Overall, the versatile nature of arylsulfonamides makes them a valuable motif in drug discovery, offering diverse opportunities for the development of novel agents for treating CNS disorders.
Collapse
Affiliation(s)
- Klaudia Blicharz-Futera
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Michał Kamiński
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland; Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, 16 Łazarza Street, 31-530 Krakow, Poland
| | - Katarzyna Grychowska
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Vittorio Canale
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland
| | - Paweł Zajdel
- Department of Organic Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Krakow, Poland.
| |
Collapse
|
26
|
Li W, Luo Y, Zhu S, Wang M, Zhao X, Ning Z. Integrated metabolome and transcriptome analysis reveals key genes and pathways associated with egg yolk percentage in chicken. Poult Sci 2025; 104:104815. [PMID: 39914020 PMCID: PMC11848451 DOI: 10.1016/j.psj.2025.104815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/10/2025] [Accepted: 01/11/2025] [Indexed: 02/27/2025] Open
Abstract
Yolk percentage is a critical index in the egg product industry, reflecting both nutritional value and economic benefits. To elucidate the underlying mechanisms that contribute to variations in egg yolk percentage, we performed integrated transcriptome and metabolome analyses on the liver, ovary, and magnum tissues of Rhode Island Red chickens with high and low yolk percentages. A total of 322 differentially expressed genes (DEGs) and 128 significantly differential metabolites (SDMs) (VIP>1, P < 0.05) were identified in the liver, whereas 419 DEGs and 215 SDMs were detected in the ovary, and 238 DEGs along with 47 SDMs were found in the magnum. In the liver, genes such as HMGCR, DHCR7, MSMO1, and CYP7A1 were linked to cholesterol metabolism, essential for steroid hormone synthesis and yolk formation, while ACACB, ACSL1, ACSL4, LPL, and SGPP2 were involved in fatty acid biosynthesis, a key process for supplying energy and structural components of the yolk. In the ovary, COL6A6, COMP, CHAD, ITGA7, THBS2, and TNC contributed to extracellular matrix-receptor interactions, which are fundamental for follicle development and oocyte maturation. In the magnum, UGT1A1, MAOB, and ALDH3B2 participated in drug metabolism-cytochrome P450 and amino acid metabolism, ensuring a proper environment for egg white formation and potentially influencing nutrient allocation to the yolk. Metabolic pathway enrichment revealed that steroid hormone biosynthesis, glycerophospholipid metabolism, and betaine metabolism were predominant in the liver; pyruvate, taurine, and hypotaurine metabolism in the ovary; and phenylalanine metabolism in the magnum. Moreover, integrated analysis highlighted key metabolites and genes potentially regulating yolk deposition, including 7,8-dihydroneopterin and Pg 38:4 in the liver (related to immune modulation and lipid metabolism, respectively), thalsimine in the ovary, as well as DL-glutamine in the magnum, all of which may be crucial for maintaining metabolic homeostasis and supporting egg formation. Collectively, these findings deepen our understanding of how distinct molecular and metabolic pathways in the liver, ovary, and magnum orchestrate yolk proportion and deposition. Such insights may advance future strategies to improve egg quality and productivity in poultry breeding programs.
Collapse
Affiliation(s)
- Wen Li
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuxing Luo
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Shoujia Zhu
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Mengyuan Wang
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xuli Zhao
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhonghua Ning
- National Engineering Laboratory for Animal Breeding and Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and Rural Affairs, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
27
|
Fan D, Jiao S, Fu Y, Zhang J, Zhuang Y, Huang J, Bi Y, Zhang J, Zhang N. Bacillus pumilus 315 improves intestinal microbiota and barrier function to alleviate diarrhea of neonatal goats. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:366-375. [PMID: 40034463 PMCID: PMC11872662 DOI: 10.1016/j.aninu.2024.10.008] [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: 05/17/2024] [Revised: 10/06/2024] [Accepted: 10/29/2024] [Indexed: 03/05/2025]
Abstract
Diarrhea is the leading cause of mortality in postnatal goat kids, seriously impacting breeding efficiency. This study aimed to explore the effects of Bacillus pumilus 315 (B. pumilus) on goat kids' diarrhea and its regulatory mechanism. Thirty-six 1-day-old goat kids were assigned into four treatments, the control (CON) group and low-, medium- and high-dose groups supplemented with B. pumilus at 1 × 108 (BP1), 5 × 108 (BP5), and 1 × 109 CFU/d (BP10). Each group consisted of 9 replicates with 1 goat kid per replicate. The results showed that the incidence of diarrhea and fecal scores decreased significantly (P < 0.05). A dose of 5 × 108 CFU/d B. pumilus reduced pro-inflammatory factors (including tumor necrosis factor-α [TNF-α], interleukin-1β [IL-1β], interleukin-6 [IL-6], P < 0.05), increased the expression levels of anti-inflammatory factors (including transforming growth factor-β [TGF-β], peroxisome proliferate-activated receptor-gamma [PPAR-γ], interleukin-10 [IL-10], P < 0.05), immune indicators (including immunoglobulin G [IgG], immunoglobulin A [IgA], immunoglobulin M [IgM], secretory immunoglobulin A [sIgA], P < 0.05) and antioxidant indicators (including total antoxidative capacity [T-AOC], superoxide dismutase [SOD], glutathione peroxidase [GSH-Px], catalase [CAT], P < 0.05) in both jejunum and colon, and ultimately improved the barrier function of the jejunum and colon mucosa. The enhanced gut immunity and barrier function were associated with increased abundance of Enterococcus and Lactobacillus (P < 0.05) and decreased abundance of Campylobacter and Escherichia-Shigella (P < 0.05). In conclusion, dietary addition of B. pumilus may improve gut health by modulating the composition and function of the flora, ultimately alleviating diarrhea in goat kids.
Collapse
Affiliation(s)
- Dingkun Fan
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Shuai Jiao
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China
| | - Yuze Fu
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jixian Zhang
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yimin Zhuang
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Juan Huang
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yanliang Bi
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jianxin Zhang
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China
| | - Naifeng Zhang
- Key Laboratory of Feed Biotechnology, The Ministry of Agriculture and Rural Affairs, Institute of Feed Research of Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
28
|
Sinha S, Cruz F, Cuenca EM, Przydacz M, Kheir GB, Kanai AJ, Van Huele A, Gajewski JB, Tarcan T, Lazar JM, Weiss JP, Tyagi P, Abrams P, Wein A. Is It Possible to Regenerate the Underactive Detrusor? Part 1. Molecular and Stem Cell Therapies Targeting the Urinary Bladder and Neural Axis - ICI-RS 2024. Neurourol Urodyn 2025; 44:577-584. [PMID: 39370871 DOI: 10.1002/nau.25597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
INTRODUCTION Detrusor muscle weakness is commonly noted on urodynamics in patients with refractory voiding difficulty. No approved therapies have been proven to augment the strength of a detrusor voiding contraction. METHODS This subject was discussed by a think-tank at the International Consultation on Incontinence- Research Society (ICI-RS) meeting held in Bristol, June 2024. The discussions of the think-tank are being published in two parts. This first part discusses molecular and stem cell therapies targeting the urinary bladder and the neural axis. RESULTS Senescence of the urothelium and extracellular ATP acting through P2X3 receptors might be important in detrusor underactivity. Several molecules such as parasympathomimetics, acotiamide, ASP8302, neurokinin-2 agonists have been explored but none has shown unequivocal clinical benefit. Different stem cell therapy approaches have been used, chiefly in neurogenic dysfunction, with some studies showing benefit. Molecular targets for the neural axis have included TRPV-4, Bombesin, and serotoninergic receptors and TAC-302 which induces neurite growth. CONCLUSIONS Several options are currently being pursued in the search for an elusive molecular or stem cell option for enhancing the power of the detrusor muscle. These encompass a wide range of approaches that target each aspect of the contraction mechanism including the urothelium of bladder and urethra, myocyte, and neural pathways. While none of these have shown unequivocal clinical utility, some appear promising. Lessons from other fields of medicine might prove instructive. CLINICAL TRIAL REGISTRATION Not necessary. Not a clinical trial.
Collapse
Affiliation(s)
- Sanjay Sinha
- Department of Urology, Apollo Hospital, Hyderabad, India
| | - Francisco Cruz
- Departamento de Urologia, Hospital de S. João, Faculdade de Medicina do Porto, Porto, Portugal
| | | | | | | | - Anthony J Kanai
- Department of Medicine and Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Jerzy B Gajewski
- Department of Urology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Tufan Tarcan
- Departments of Urology, Marmara University School of Medicine and Koç University School of Medicine, Istanbul, Turkey
| | - Jason M Lazar
- Department of Cardiology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Jeffrey P Weiss
- Department of Urology, State University of New York Downstate Health Sciences University, Brooklyn, New York, USA
| | - Pradeep Tyagi
- Department of Urology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Paul Abrams
- Department of Urology, University of Bristol, Bristol, UK
| | - Alan Wein
- Desai-Seth Institute of Urology, University of Miami, Miami, Florida, USA
| |
Collapse
|
29
|
Makled MN, Makled NN, Abdel-Rahman AM, Sharawy MH. Inhibition of p75 NTR/p53 axis by ambrisentan suppresses apoptosis and oxidative stress-mediated renal damage in a cisplatin AKI model. Chem Biol Interact 2025; 408:111408. [PMID: 39892498 DOI: 10.1016/j.cbi.2025.111408] [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: 11/21/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
Cisplatin (CP) is a potent antineoplastic agent that triggers nephrotoxicity as a major adverse effect which can cause treatment interruptions and limitations to its clinical use. Nephrotoxicity associated with CP involves inflammation, oxidative stress, and apoptosis in kidney tubules. The objective of this work was to assess the effect of the blockade of endothelin-1 (ET-1) receptor with ambrisentan on altered renal function induced by CP. Swiss albino mice were assigned into control, CP, CP/Amb-5, and CP/Amb-10 groups. Ambrisentan improved kidney function (serum creatinine and BUN) and histopathological changes in comparison to CP-treated group. Ambrisentan significantly reduced protein expression of p75NTR and protein level of JNK influencing renal apoptosis as evidenced by reducing p53, caspase-3, and Bax levels and elevating Bcl-2 level (p < 0.05 vs CP group). Moreover, vasodilatory effect of ambrisentan was indicated by significant increase in level of vascular endothelial growth factor (VEGF) and endothelial nitric oxide synthase (eNOS) (p < 0.05 vs CP group). Ambrisentan also significantly restored oxidative balance in renal tissues as evidenced by reduced malondialdehyde and increased total antioxidant capacity and superoxide dismutase activity, in addition to decreasing nitric oxide levels (p < 0.05 vs CP group). This protective effect of ambrisentan might be mediated through the downregulation of death receptor, P75NTR that in turn restores renal blood flow and oxidative balance and regulates p53, VEGF/eNOS, NF-κB, and Bcl-2/Bax/caspase-3 signaling.
Collapse
Affiliation(s)
- Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Noran N Makled
- Department of Urology, Faculty of Medicine, Damietta University, Damietta, Egypt; Department of Urology, Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | - Ahmed M Abdel-Rahman
- Department of Nephrology, Urology and Nephrology Center, Mansoura University, Mansoura, 35516, Egypt
| | - Maha H Sharawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
30
|
Yousef DA, Abdalla MS, Elshopakey GE, Al-Olayan E, Abdel Moneim AE, Ramadan SS. Diosmin-loaded chitosan nanoparticles mitigate doxorubicin-evoked cardiotoxicity in rats by featuring oxidative imbalance mechanism, NF-κB, and Bcl-2/Bax pathways. Int J Biol Macromol 2025; 305:140991. [PMID: 39952491 DOI: 10.1016/j.ijbiomac.2025.140991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 01/12/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
Cardiotoxicity is doxorubicin's primary side effect. Its cardiac toxicity has been attributed to the generation of free radicals. The present work was designed to understand the potential underlying pathways behind the cardioprotective action of diosmin (Dio) and Dio-loaded chitosan nanoparticles (DCNPs) against doxorubicin (Dox)-mediated cardiotoxicity. Male rats were allocated into five groups: control, Dio (100 mg/kg), Dox (12 mg/kg), Dio + Dox (100 mg/kg + 12 mg/kg), and DCNPs+Dox (100 mg/kg DCNPs/orally+12 mg/kg Dox/IP). Notably, in response to Dox, a significant increase of cardiac biomarkers with a decrease in Na+/K+-ATPase activity was detected. The cardiac inflammatory and pro-apoptotic protein levels were elevated with decreased cardiac interleukin-10 and Bcl-2 levels when the rats were subjected to Dox. Also, the cardiac expression of the fibrotic marker MMP-9 was increased. Moreover, Dox raised malondialdehyde and nitric oxide levels, accompanied by minimizing antioxidant status. Also, Dox-treated rats showed cardiac histopathological impairment compared to the control. The oral administration of Dio or DCNPs enhanced the activity of antioxidant enzymes and diminished inflammatory cytokines and apoptotic markers in the Dox-exposed rats. In summary, these findings indicate that DCNPs exhibit significant cardioprotective effectiveness against Dox-mediated toxicity by suppressing various mechanisms, such as redox status, the NF-κB pathway, and apoptosis.
Collapse
Affiliation(s)
- Doaa A Yousef
- Molecular Biotechnology Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Mohga S Abdalla
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| | - Gehad E Elshopakey
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Mansoura University, 35516, Egypt; Department of Veterinary Diseases, Faculty of Veterinary Medicine, Delta University for Science and Technology, 35712 Gamasa, Egypt
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ahmed E Abdel Moneim
- Zoology and Entomology Department, Faculty of Science, Helwan University, Cairo 11795, Egypt; Al-Ayen Scientific Research Center, Al-Ayen Iraqi University, AUIQ, P.O. Box: 64004, An Nasiriyah, Thi Qar, Iraq.
| | - Shimaa S Ramadan
- Biochemistry Sector, Chemistry Department, Faculty of Science, Helwan University, Cairo 11795, Egypt
| |
Collapse
|
31
|
Chiba T, Oda A, Zhang Y, Pfister K, Bons J, Bharathi SS, Kinoshita A, Zhang BB, Richert A, Schilling B, Goetzman E, Sims-Lucas S. Loss of long-chain acyl-CoA dehydrogenase protects against acute kidney injury. JCI Insight 2025; 10:e186073. [PMID: 39932791 PMCID: PMC11949023 DOI: 10.1172/jci.insight.186073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
The renal tubular epithelial cells (RTECs) are particularly vulnerable to acute kidney injury (AKI). While fatty acids are the preferred energy source for RTECs via fatty acid oxidation (FAO), FAO-mediated H2O2 production in mitochondria has been shown to be a major source of oxidative stress. We have previously shown that a mitochondrial flavoprotein, long-chain acyl-CoA dehydrogenase (LCAD), which catalyzes a key step in mitochondrial FAO, directly produces H2O2 in vitro. Furthermore, we showed that renal LCAD becomes hyposuccinylated during AKI. Here, we demonstrated that succinylation of recombinant LCAD protein suppresses the production of H2O2. Following 2 distinct models of AKI, cisplatin treatment or renal ischemia/reperfusion injury (IRI), LCAD-/- mice demonstrated renoprotection. Specifically, LCAD-/- kidneys displayed mitigated renal tubular injury, decreased oxidative stress, preserved mitochondrial function, enhanced peroxisomal FAO, and decreased ferroptotic cell death. LCAD deficiency confers protection against 2 distinct models of AKI. This suggests a therapeutically attractive mechanism whereby preserved mitochondrial respiration as well as enhanced peroxisomal FAO by loss of LCAD mediates renoprotection against AKI.
Collapse
Affiliation(s)
- Takuto Chiba
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Akira Oda
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yuxun Zhang
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Katherine Pfister
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, California, USA
| | - Sivakama S. Bharathi
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ayako Kinoshita
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Bob B. Zhang
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam Richert
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Eric Goetzman
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sunder Sims-Lucas
- Department of Pediatrics, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh (UPMC CHP), University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
32
|
Xin J, He L, Li Y, Pu Q, Du X, Ban F, Han D. Sanguinarine chloride hydrate mitigates colitis symptoms in mice through the regulation of the intestinal microbiome and metabolism of short-chain fatty acids. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167579. [PMID: 39561858 DOI: 10.1016/j.bbadis.2024.167579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/21/2024] [Accepted: 11/13/2024] [Indexed: 11/21/2024]
Abstract
Sanguinarine constitutes the main components of Macleaya cordata, and exhibits diverse biological and pharmacological activities. This study investigated the effects of sanguinarine chloride hydrate (SGCH) on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) mice. Five groups were designed to investigate the effects of SGCH on the pathological symptoms, the mRNA expression levels of inflammatory cytokines, colonic mucosal barrier damage, microbiota composition, and SCFAs metabolism in UC mice. The administration of SGCH in DSS-induced UC mice resulted in the amelioration of pathological symptoms, as evidenced by an increase in body weight, a decrease in disease activity index score, elongation of colon length, reduction in spleen index, and improvement in colon injury. SGCH can regulate the expression of inflammatory cytokines (IL-6, TNF-α, IL-1β and IL-10) and tight junction proteins (ZO-1 and Occludin) associated with UC. SGCH exhibited a significant decrease in NF-κB P65 mRNA expression levels, accompanied by a significantly reduced protein level of NF-κB P-P65/P65. Further studies revealed SGCH effectively reversed the decrease in intestinal microbiota diversity induced by UC, thereby promoting the growth of beneficial bacteria such as Akkermansia, Alistipes, and norank_o_Clostridia_UCG-014. Correlation analysis demonstrated a positive association between butanoic acid, propanoic acid, isobutyric acid, isovaleric acid, valeric acid, hexanoic acid with Colidextribacter, while Coriobacteriaceae_UCG-002 exhibited a negative correlation with butanoic acid, acetic acid and propanoic acid. In conclusion, the administration of SGCH can ameliorate clinical symptoms in UC mice, regulate the expression of inflammatory cytokines and tight junction proteins, modulate intestinal microbiota metabolism and SCFAs production.
Collapse
Affiliation(s)
- Jige Xin
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Lin He
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Yanlin Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Qiqi Pu
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Xuan Du
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Fuze Ban
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming 650201, China
| | - Diangang Han
- Technology Center of Kunming Customs, Kunming 650200, China.
| |
Collapse
|
33
|
Karunakar KK, Edwin ER, Gopalakrishnan M, Cheriyan BV, Ramaiyan V, Karthikha VS, Justin JP. Advances in nephroprotection: the therapeutic role of selenium, silver, and gold nanoparticles in renal health. Int Urol Nephrol 2025; 57:479-510. [PMID: 39312019 DOI: 10.1007/s11255-024-04212-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 09/18/2024] [Indexed: 01/29/2025]
Abstract
Renal toxicity is a disorder that causes considerable issues in healthcare systems world, highlighting the critical importance of creating alternative treatments. Metallic nanoparticles have recently emerged as promising therapeutic agents for nephroprotection because of their remarkable properties. Numerous disciplines, including medicine, biotechnology, and the food industry, are currently investigating and exploring metallic nanoparticles, such as selenium, silver, and gold, with promising outcomes. In this overview, we provide the most current findings on cutting-edge nephroprotection through metallic nanoparticles, especially selenium, silver, and gold nanoparticles. While outlining the benefits, we outline possible methods for developing metallic nanoparticles, characterization techniques, and nephroprotection therapies. Selenium nanoparticles (SeNPs) minimize oxidative stress, a primary cause of nephrotoxicity through cell regeneration which protects kidneys. Silver nanoparticles (AgNPs) have anti-inflammatory capabilities that help alleviate kidney damage and nephrotoxicity. Gold nanoparticles (AuNPs), which are biocompatible and immune-modifying, reduce inflammation and promote renal cell regeneration, indicating nephroprotective advantages. Renal protection via the use of metallic nanoparticles represents a promising new frontier in the fight against kidney disease and other renal disorders. Metallic nanoparticles of selenium, silver, and gold can protect the kidneys by lowering oxidative stress, reducing inflammation, and improving cell repair. Through their mechanisms, these nanoparticles effectively safeguard and repair kidney function, making them suitable for treating renal diseases. The potential applications of selenium, silver, and gold nanoparticles, as well as their complex modes of action and renal penetration, provide fresh hope for improving renal health and quality of life in patients with kidney disease. The current study highlights therapeutic ability, stability, nephroprotection, and toxicity profiles, as well as the importance of continuous research in this dynamic and evolving field.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Elizabeth Rani Edwin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Meenaloshini Gopalakrishnan
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India.
| | - Velmurugan Ramaiyan
- Department of Pharmacology, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - V S Karthikha
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| | - Jerry Peliks Justin
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, TN, 602105, India
| |
Collapse
|
34
|
Lim JJ, Klaassen CD, Cui JY. Deciphering the cell type-specific and zonal distribution of drug-metabolizing enzymes, transporters, and transcription factors in livers of mice using single-cell transcriptomics. Drug Metab Dispos 2025; 53:100029. [PMID: 39919554 DOI: 10.1016/j.dmd.2024.100029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
The liver contains multiple cell types, including resident cell types and immune cells. The liver is also categorized into 3 zones: periportal (zone 1), midzonal (zone 2), and centrilobular (zone 3). The goal of this study was to characterize the distribution of drug-processing genes (DPGs) in mouse liver using published single-cell and nuclei transcriptomic datasets, which were subjected to zonal deconvolution. Filtering, normalization, clustering, and differential expression analyses were performed using Seurat V5 in R. Hepatocytes were assigned to 3 zones based on known zonal markers and validated with published spatial transcriptomics data. Among the 195 DPGs profiled, most were expressed highest in hepatocytes (61.3%). Interestingly, certain DPGs were expressed most highly in nonparenchymal cells, such as in cholangiocytes (11.2%, eg, carboxylesterase [Ces] 2e, Ces2g), endothelial cells (7.2%, eg, aldo-keto reductase [Akr] 1c19, Akr1e1), Kupffer cells (5.3%, eg, Akr1a1, Akr1b10), stellate cells (5.1%, eg, retinoic acid receptor [Rar] α, Rarβ), myofibroblasts (2.9%, RAR-related orphan receptor [Rar] α), and a few were expressed in immune cell types. In hepatocytes, 72.4% of phase-I enzymes were enriched in zone 3. Phase-II conjugation enzymes such as UDP-glucuronosyltransferases (75%) were enriched in zone 3, whereas sulfotransferases (40%) were enriched in zone 1. Hepatic xenobiotic transporters were enriched in zone 3. The xenobiotic biotransformation-regulating transcription factors were enriched in zone 3 hepatocytes. The enrichment of DPGs in liver cell types, including non-parenchymal cells and zone 1 hepatocytes, may serve as an additional repertoire for xenobiotic biotransformation. SIGNIFICANCE STATEMENT: Our study is among the first to systematically characterize the baseline mRNA enrichment of important drug-processing genes in different cell types and zones in the liver. This finding will aid in further understanding the mechanisms of chemical-induced liver injury with improved resolution and precision.
Collapse
Affiliation(s)
- Joe Jongpyo Lim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington; Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington
| | - Curtis Dean Klaassen
- Department of Pharmacology, Toxicology, and Therapeutics, School of Medicine, University of Kansas, Kansas City, Kanas.
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, Washington; Environmental Health and Microbiome Research Center (EHMBRACE), Seattle, Washington.
| |
Collapse
|
35
|
Al‐Naggar IM, Antony M, Baker D, Wang L, Godoy LDC, Kuo C, Fraser MO, Smith PP, Xu M, Kuchel GA. Polyploid superficial uroepithelial bladder barrier cells express features of cellular senescence across the lifespan and are insensitive to senolytics. Aging Cell 2025; 24:e14399. [PMID: 39644167 PMCID: PMC11822673 DOI: 10.1111/acel.14399] [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: 11/17/2023] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 12/09/2024] Open
Abstract
Lower urinary tract dysfunction (LUTD) increases with aging. Ensuing symptoms including incontinence greatly impact quality of life, isolation, depression, and nursing home admission. The aging bladder is hypothesized to be central to this decline, however, it remains difficult to pinpoint a singular strong driver of aging-related bladder dysfunction. Many molecular and cellular changes occur with aging, contributing to decreased resilience to internal and external stressors, affecting urinary control and exacerbating LUTD. In this study, we examined whether cellular senescence, a cell fate involved in the etiology of most aging diseases, contributes to LUTD. We found that umbrella cells (UCs), luminal barrier uroepithelial cells in the bladder, show senescence features over the mouse lifespan. These polyploid UCs exhibit high cyclin D1 staining, previously reported to mediate tetraploidy-induced senescence in vitro. These senescent UCs were not eliminated by the senolytic combination of Dasatinib and Quercetin. We also tested the effect of a high-fat diet (HFD) and senescent cell transplantation on bladder function and showed that both models induce cystometric changes similar to natural aging in mice, with no effect of senolytics on HFD-induced changes. These findings illustrate the heterogeneity of cellular senescence in varied tissues, while also providing potential insights into the origin of urothelial cancer. We conclude that senescence of bladder uroepithelial cells plays a role in normal physiology, namely in their role as barrier cells, helping promote uroepithelial integrity and impermeability and maintaining the urine-blood barrier.
Collapse
Affiliation(s)
- Iman M. Al‐Naggar
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- Department of Cell BiologyUniversity of Connecticut HealthFarmingtonConnecticutUSA
- Department of SurgeryUniversity of Connecticut HealthFarmingtonConnecticutUSA
| | - Maria Antony
- The University of Connecticut School of MedicineFarmingtonConnecticutUSA
| | - Dylan Baker
- Department of Genetics & Genome SciencesUniversity of Connecticut HealthFarmingtonConnecticutUSA
- The Jackson Laboratory for Genomic MedicineFarmingtonConnecticutUSA
| | - Lichao Wang
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
| | - Lucas Da Cunha Godoy
- The Cato T. Laurencin Institute for Regenerative EngineeringFarmingtonConnecticutUSA
| | - Chia‐Ling Kuo
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- The Cato T. Laurencin Institute for Regenerative EngineeringFarmingtonConnecticutUSA
| | - Matthew O. Fraser
- Department of Research & DevelopmentDurham Veterans Affairs Medical CentersDurhamNorth CarolinaUSA
| | - Phillip P. Smith
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- Department of SurgeryUniversity of Connecticut HealthFarmingtonConnecticutUSA
- Connecticut Institute for Brain and Cognitive ScienceUniversity of ConnecticutStorrsConnecticutUSA
| | - Ming Xu
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
- Department of Genetics & Genome SciencesUniversity of Connecticut HealthFarmingtonConnecticutUSA
| | - George A. Kuchel
- Center on AgingUniversity of ConnecticutFarmingtonConnecticutUSA
| |
Collapse
|
36
|
Nettlefold C, Chakraborty P, Al Shaheen A, Denham N, Kakarla J, Burg MR, Hayashi T, Ahmed I, Nanthakumar K. A Primer on the Evolving Subspecialty of Onco-Electrophysiology. Can J Cardiol 2025; 41:181-194. [PMID: 39521053 DOI: 10.1016/j.cjca.2024.11.005] [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: 09/02/2024] [Revised: 10/15/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Cardio-oncology has become a well-established subspecialty because of the growing burden of cardiovascular diseases in oncology patients, resulting from the cardiac toxicities of cancer therapies and the coexistence of both conditions in the same population. As with other cardiovascular conditions, cardiac arrhythmias have emerged as an important concern in patients with cancer. However, the management of arrhythmias is more complicated in these patients because of complex interactions between oncotherapeutics and arrhythmia-treatment strategies. Similarly, patients with cardiac implantable electronic devices (CIEDs) require cancer treatment strategies that involve radiation therapy require specific management strategies. Thus, there is a need for a specific mechanistic understanding of electrophysiological abnormalities, arrhythmia, and device management in oncology patients, especially given the expanding range of oncologic therapies and radiation strategies. This increasingly prevalent clinical challenge requires new expertise that expands on a yearly basis. This narrative review deals with this recent expansion and addresses key areas of onco-electrophysiology, including the mechanistic basis of common electrocardiographic changes, diagnosis, and management of arrhythmias attributable to oncotherapeutics and the care of patients with arrhythmias who require oncologic therapies, especially patients with devices and drug interactions leading to arrhythmias as seen by cardiac physicians dealing with oncology patients. In addition, it reviews evolving management strategies and protocols for patients with implantable devices, especially if urgent radiation is needed. This review aims to bridge the recent knowledge growth in arrhythmia care for patients with cancer and highlight the evolution of onco-electrophysiology as a subspeciality.
Collapse
Affiliation(s)
- Chloe Nettlefold
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Praloy Chakraborty
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Abdullah Al Shaheen
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Nathan Denham
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Jayant Kakarla
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Melanie R Burg
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Takahiro Hayashi
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Intisar Ahmed
- Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
37
|
Luo JX, Gao XT, Rong Z, Zhang LH, Sun YF, Qi ZL, Yu Q, Waiho K, Zhao WX, Xu YH, Zhao CL, Wu CB. Transcriptome Sequencing Reveals Effects of Artificial Feed Domestication on Intestinal Performance and Gene Expression of Carnivorous Mandarin Fish (Siniperca chuatsi) and Related Mechanisms. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2025; 27:41. [PMID: 39891779 DOI: 10.1007/s10126-025-10420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
Mandarin fish (Siniperca chuatsi) is a voracious carnivorous species, usually consuming only live bait fish, but dietary acclimation enables it to accept artificial feed. However, the effects of dietary acclimation on intestinal performance and gene expression in mandarin fish and related mechanisms remain largely unknown. Therefore, this study investigated the effects of artificial feed on intestinal physicochemical and biochemical performance and gene expression in mandarin fish. Mandarin fish were sampled on day 10 after feeding with live dace (LD), at day 40 after subsequent feeding with dead dace plus artificial feed (DD + AF) from day 11 to day 40, and at day 90 after continuous feeding with artificial feed (AF) alone from day 41 to day 90 for transcriptome sequencing. The biochemical analysis results indicated that artificial feed significantly increased the activity of antioxidant enzymes glutathione peroxidase and superoxide dismutase in the intestine, liver, and stomach. Histological analysis demonstrated intestinal damage in mandarin fish fed with artificial feed. The GO and KEGG enrichment analyses indicated that the DEGs in AF vs. DD + AF were significantly enriched in the pentose phosphate pathway, and the DEGs in AF vs. LD were mainly significantly enriched in glycolysis/gluconeogenesis and PPAR signaling pathways. Nineteen feed acclimation-related key genes such as gene pfkfb4a and scd were identified in the intestine and found to exhibit upregulated expressions. These results revealed that artificial feed domestication enhanced the antioxidant capacity of the mandarin fish intestine and reduced hepatic lipid deposition by upregulating the related gene expression of mandarin fish and that the regulation of carbon metabolisms, including sugar, lipid, and steroid metabolisms, might be fundamental mechanisms for mandarin fish to acclimatize to dietary changes. These findings provide novel insights into the feed acclimation mechanism of mandarin fish, holding implications for promoting large-scale artificial feed aquaculture of mandarin fish and improving economic efficiency.
Collapse
Affiliation(s)
- Jia-Xing Luo
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China
| | - Xiao-Tian Gao
- Hebei Academy of Ocean and Fishery Sciences, Qinhuangdao, 066200, Hebei, China
| | - Zhen Rong
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China
| | - Li-Han Zhang
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China
- Hebei Key Laboratory of Nutritional Regulation and Disease Control for Aquaculture, Qinhuangdao, 066003, Hebei, China
| | - Yan-Feng Sun
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China
- Hebei Key Laboratory of Nutritional Regulation and Disease Control for Aquaculture, Qinhuangdao, 066003, Hebei, China
| | - Zun-Li Qi
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China
- Hebei Key Laboratory of Nutritional Regulation and Disease Control for Aquaculture, Qinhuangdao, 066003, Hebei, China
| | - Qi Yu
- Hebei Academy of Ocean and Fishery Sciences, Qinhuangdao, 066200, Hebei, China
| | - Khor Waiho
- Higher Institution Center of Excellence (HICoE), Institute of Tropical Aquaculture and Fisheries, University Malaysia Terengganu, 20000, Kuala Terengganu, Terengganu, Malaysia
| | - Wei-Xu Zhao
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China
| | - Yi-Huan Xu
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China.
- Hebei Key Laboratory of Nutritional Regulation and Disease Control for Aquaculture, Qinhuangdao, 066003, Hebei, China.
| | - Chun-Long Zhao
- Hebei Academy of Ocean and Fishery Sciences, Qinhuangdao, 066200, Hebei, China.
| | - Cheng-Bin Wu
- Ocean College, Hebei Agricultural University, Qinhuangdao, 066003, Hebei, China.
- Hebei Key Laboratory of Nutritional Regulation and Disease Control for Aquaculture, Qinhuangdao, 066003, Hebei, China.
| |
Collapse
|
38
|
Kim YH, Jeong S, Cho KA, Woo SY, Han SH, Ryu KH. Reduction of Low-Density Lipoprotein Cholesterol by Mesenchymal Stem Cells in a Mouse Model of Exogenous Cushing's Syndrome. Tissue Eng Regen Med 2025; 22:237-248. [PMID: 39873947 PMCID: PMC11794754 DOI: 10.1007/s13770-024-00697-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/11/2024] [Accepted: 12/22/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Exogenous Cushing's syndrome, which results from prolonged glucocorticoid treatment, is associated with metabolic abnormalities. Previously, we reported the inhibitory effect of tonsil-derived mesenchymal stem cell conditioned medium (T-MSC CM) on glucocorticoid signal transduction. In this study, we investigated the therapeutic efficacy of T-MSCs in a mouse model of exogenous Cushing's syndrome. METHODS Exogenous Cushing's syndrome model mice was generated by corticosterone administration in the drinking water for 5 weeks, and T-MSCs were injected intraperitoneally twice during the third week. Serum lipid profiles were measured using a chemistry analyzer. HepG2 cells were treated with dexamethasone and co-cultured with T-MSCs. Expression levels of genes involved in cholesterol metabolism were examined using real-time PCR. Low-density lipoprotein receptor (LDLR) protein levels were determined using western blotting and immunohistochemistry. Liver RNA extracted from the CORT and CORT + MSC mouse groups was used for transcriptome sequencing analysis and protein-protein interaction analysis. RESULTS Weight reduction and improvements in dyslipidemia by T-MSC administration were observed only in female mice. T-MSCs reduce circulating LDL cholesterol levels by downregulating liver X receptor α (LXRα) and inducible degrader of LDLR (IDOL) expression, thereby stabilizing LDLRs in the liver. Transcriptome analysis of liver tissue revealed pathways that are regulated by T-MSCs administration. CONCLUSION Administration of MSCs to female mice receiving chronic corticosterone treatment reduced the circulating LDL cholesterol level by downregulating the LXRα-IDOL axis in hepatocytes. These results suggest that T-MSCs may offer a novel therapeutic strategy for managing exogenous Cushing's syndrome by regulating cholesterol metabolism.
Collapse
Affiliation(s)
- Yu-Hee Kim
- Advance Biomedical Research Institute, Ewha Womans University Seoul Hospital, Seoul, South Korea
| | - Seonghee Jeong
- Ewha Medical Academy, Ewha Womans University Medical Center, Seoul, South Korea
| | - Kyung-Ah Cho
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - So-Youn Woo
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Seung-Ho Han
- Ewha Medical Academy, Ewha Womans University Medical Center, Seoul, South Korea
- Department of Anatomy, College of Medicine, Ewha Womans University, Seoul, South Korea
| | - Kyung-Ha Ryu
- Department of Pediatrics, College of Medicine, Ewha Womans University, Seoul, 07804, South Korea.
| |
Collapse
|
39
|
Salah TM, Rabie MA, El Sayed NS. Renoprotective effect of berberine in cisplatin-induced acute kidney injury: Role of Klotho and the AMPK/mtor/ULK1/Beclin-1 pathway. Food Chem Toxicol 2025; 196:115179. [PMID: 39645019 DOI: 10.1016/j.fct.2024.115179] [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: 10/13/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Cisplatin (Cisp) is a potent cancer drug, but its use is limited by acute kidney injury (AKI). Autophagy, a process that removes damaged proteins and maintains cellular homeostasis, has been shown to alleviate Cisp-induced AKI. The balance between autophagy and apoptosis is crucial to kidney protection. Treatment with Berberine, known for its antioxidant and anti-inflammatory effects in nephrotoxicity models, was studied for its potential to enhance autophagy in Cisp-induced AKI. Treatment with Berberine (Berb) upregulated Klotho gene expression, enhancing autophagy as indicated by elevated protein levels of pS486-AMPK, pS638-ULK1, and Beclin-1, accompanied by a decrease in pS248-mTOR protein expression. Also, Berb mitigated oxidative stress by reducing elevated MDA levels and boosting SOD activity, which in turn suppressed inflammation by down-regulating HMGB1 and RAGE gene expression, as well as reducing pS536-NF-κB and IL-6 protein contents. Additionally, Berb reduced apoptosis by increasing Bcl-2 and decreasing Bax. This coordinated action preserved kidney function, evidenced by reductions in early injury markers (cystatin C, KIM-1, NGAL) and late markers (creatinine, BUN), along with attenuation of histopathological alterations. The use 3-MA, autophagy inhibitor, nullified these protective effects, highlighting Berb's role in promoting autophagy, reducing oxidative stress, inflammation, and apoptosis, and preserving renal health in Cisp-induced AKI.
Collapse
Affiliation(s)
- Tasneem M Salah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt.
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| |
Collapse
|
40
|
Cheng P, Zeng Z, Liu J, Liew SS, Hu Y, Xu M, Pu K. Urinary bioorthogonal reporters for the monitoring of the efficacy of chemotherapy for lung cancer and of associated kidney injury. Nat Biomed Eng 2025:10.1038/s41551-024-01340-1. [PMID: 39880894 DOI: 10.1038/s41551-024-01340-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/16/2024] [Indexed: 01/31/2025]
Abstract
The utility of urinary tests for the monitoring of the treatment efficacy and adverse events of anticancer therapies is constrained by the low concentration of relevant urinary biomarkers. Here we report, using mice with lung cancer and treated with chemotherapy, of a urinary fluorescence test for the concurrent monitoring of the levels of a tumour biomarker (cathepsin B) and of a biomarker of chemotherapy-induced kidney injury (N-acetyl-β-D-glucosaminidase). The test involves two intratracheally administered urinary reporters leveraging caged bioorthogonal click handles for the biomarker-dependent activation of 'clickability' and renal clearance, and the bioorthogonal click reaction of each renally cleared reporter with paired fluorescence indicators in the collected urine. In mouse models of chemotherapy-treated orthotopic lung cancer and of cisplatin-induced kidney injury, lower urinary fluorescence signals (which can be measured by a smartphone camera) for tumour and kidney injury levels positively correlated with animal weight gain and survival time. Biomarker-activated bioorthogonal click reactivity and renal clearance combined with bioorthogonally triggered fluorescence in vitro may enable specific, sensitive and rapid urinary assays for the monitoring of other physiopathological processes.
Collapse
Affiliation(s)
- Penghui Cheng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Ziling Zeng
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Jing Liu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Si Si Liew
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Yuxuan Hu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Mengke Xu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
41
|
Beauchemin ET, Hunter C, Maurice CF. Dextran sodium sulfate-induced colitis alters the proportion and composition of replicating gut bacteria. mSphere 2025; 10:e0082524. [PMID: 39723822 PMCID: PMC11774032 DOI: 10.1128/msphere.00825-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/15/2024] [Indexed: 12/28/2024] Open
Abstract
The bacteria living in the human gut are essential for host health. Though the composition and metabolism of these bacteria are well described in both healthy hosts and those with intestinal disease, less is known about the metabolic activity of the gut bacteria prior to, and during, disease development-especially regarding gut bacterial replication. Here, we use a recently developed single-cell technique alongside existing metagenomics-based tools to identify, track, and quantify replicating gut bacteria both ex vivo and in situ in the dextran sodium sulfate (DSS) mouse model of colitis. We show that the proportion of replicating gut bacteria decreases when mice have the highest levels of inflammation and returns to baseline levels as mice begin recovering. In addition, we report significant alterations in the composition of the replicating gut bacterial community ex vivo during colitis development. On the taxa level, we observe significant changes in the abundance of taxa such as the mucus-degrading Akkermansia and the poorly described Erysipelatoclostridium genus. We further demonstrate that many taxa exhibit variable replication rates in situ during colitis, including Akkermansia muciniphila. Lastly, we show that colitis development is positively correlated with increases in the presence and abundance of bacteria in situ which are predicted to be fast replicators. This could suggest that taxa with the potential to replicate quickly may have an advantage during intestinal inflammation. These data support the need for additional research using activity-based approaches to further characterize the gut bacterial response to intestinal inflammation and its consequences for both the host and the gut microbial community.IMPORTANCEIt is well known that the bacteria living inside the gut are important for human health. Indeed, the type of bacteria that are present and their metabolism are different in healthy people versus those with intestinal disease. However, less is known about how these gut bacteria are replicating, especially as someone begins to develop intestinal disease. This is particularly important as it is thought that metabolically active gut bacteria may be more relevant to health. Here, we begin to address this gap using several complementary approaches to characterize the replicating gut bacteria in a mouse model of intestinal inflammation. We reveal which gut bacteria are replicating, and how quickly, as mice develop and recover from inflammation. This work can serve as a model for future research to identify how actively growing gut bacteria may be impacting health, or why these particular bacteria tend to thrive during intestinal inflammation.
Collapse
Affiliation(s)
- Eve T. Beauchemin
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Claire Hunter
- Department of Public Health and Primary Care, School of Clinical Medicine, University of Cambridge, Cambridge, England, United Kingdom
| | - Corinne F. Maurice
- Department of Microbiology & Immunology, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- McGill Centre for Microbiome Research, Montreal, Quebec, Canada
| |
Collapse
|
42
|
Alipanah-Moghadam R, Aghamohammadi V, Seifi S, Esmaeili H, Noroozzadeh S, Jeddi F, Salimnejad R, Nemati A. Protective effect of Panax ginseng extract on cisplatin-induced AKI via downregulating cell death associated genes. Sci Rep 2025; 15:3233. [PMID: 39863673 PMCID: PMC11763254 DOI: 10.1038/s41598-025-87447-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
This study is designed to assess the effect of root extract of P. ginseng on kidney tissue injury attributed to cisplatin and its molecular mechanism involved in this process in the AKI rat model. Twenty-four male Wistar rats were randomly allocated into 4 experimental groups including: the control group, the cisplatin group, the extract 100 mg/kg group, and the extract 200 mg/kg group. The duration of the investigation was 7 days, and all rats except the control group received a single dose of 10 mg/kg cisplatin on the 4th day. Our findings exhibited a significant reduction in blood concentration of creatinine in extract groups compared to the cisplatin group. In the cisplatin group, severe renal histopathological alterations were observed compared to the control group. In extract groups, significantly less tissue damage was observed than in the cisplatin group. Ginseng extract 200 showed minimal tissue damage as compared to extract 100. The expression of p21, p27, p53, TIMP2, IGFBP7, and NF-κB decreased significantly in extract groups compared to the cisplatin group. Our findings displayed amelioration of cisplatin-induced AKI and dose-dependent decrease of the NF-κB gene expression and cell death-inducing genes by administration of P. ginseng extract.
Collapse
Affiliation(s)
- Reza Alipanah-Moghadam
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | | | - Sina Seifi
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Hedieh Esmaeili
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Somaieh Noroozzadeh
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ramin Salimnejad
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Ali Nemati
- Department of Clinical Biochemistry, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| |
Collapse
|
43
|
Kobayashi T, Kessoku T, Iwaki M, Nogami A, Yoneda M, Saito S, Yamana Y, Nishitani Y, Kuwahara H, Nakajima A. Lactiplantibacillus plantarum 22 A-3 ameliorates leaky gut in mice through its anti-inflammatory effects. Sci Rep 2025; 15:3264. [PMID: 39863665 PMCID: PMC11762275 DOI: 10.1038/s41598-025-87428-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
There are limited studies on the improvement of leaky gut with minor inflammation associated with various diseases. To explore the therapeutic potential of Lactiplantibacillus plantarum 22 A-3, a member of the Lactobacillus species, in addressing a leaky gut. Lactiplantibacillus plantarum 22 A-3 was administered to a leaky gut mice model with low dextran sulfate sodium concentrations. The Lactiplantibacillus plantarum 22 A-3-treated group exhibited amelioration of increased intestinal permeability, as indicated by lower blood fluorescein isothiocyanate-dextran levels compared with that of the control group. Furthermore, the messenger RNA expression of interleukin-10, an anti-inflammatory cytokine, was upregulated in the small intestine of Lactiplantibacillus plantarum 22 A-3-treated mice. Moreover, forkhead box P3 was upregulated in the small intestine and colon following Lactiplantibacillus plantarum 22 A-3 administration. Flow cytometry showed that forkhead box P3-positive regulatory T cells tended to increase in the small intestine and colon; however, this was not significant. Messenger RNA levels for the pro-inflammatory cytokines, interleukin-1 beta, and tumor necrosis factor-alpha showed no significant changes in the small intestine; however, their expressions significantly decreased in the colon. Blood fluorescein isothiocyanate-dextran levels showed that intestinal permeability also decreased in Lactiplantibacillus plantarum 22 A-3-dead bacteria. The bacterial component of Lactiplantibacillus plantarum 22 A-3 ameliorates increased intestinal permeability through its anti-inflammatory effect in the intestinal tract and may be a novel treatment for leaky gut.
Collapse
Affiliation(s)
- Takashi Kobayashi
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takaomi Kessoku
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Palliative Medicine, International University of Health and Welfare Narita Hospital, Chiba, Japan
- Department of Gastroenterology, International University of Health and Welfare Graduate School of Medicine, Chiba, Japan
| | - Michihiro Iwaki
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Asako Nogami
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masato Yoneda
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan.
| | - Satoru Saito
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
- Department of Gastroenterology, Sanno Hospital, Tokyo, Japan
| | - Yoshie Yamana
- Research Center, Maruzen Pharmaceuticals Co., Ltd., Hiroshima, Japan
| | - Yosuke Nishitani
- Research Center, Maruzen Pharmaceuticals Co., Ltd., Hiroshima, Japan
| | | | - Atsushi Nakajima
- Department of Gastroenterology and Hepatology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
44
|
Przybyciński J, Czerewaty M, Kwiatkowska E, Dziedziejko V, Safranow K, Domański L, Pawlik A. MicroRNAs miR-148a-3p, miR-425-3p, and miR-20a-5p in Patients with IgA Nephropathy. Genes (Basel) 2025; 16:125. [PMID: 40004454 PMCID: PMC11854660 DOI: 10.3390/genes16020125] [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: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES IgA nephropathy (IgAN) is one of the most common forms of glomerulonephritis leading to renal failure. MicroRNAs have been shown to play an important role in the pathogenesis and clinical course of IgA nephropathy; therefore, they offer the possibility of noninvasive diagnosis of this disease and have some value in predicting disease prognosis. This study aimed to evaluate the plasma levels of miR-148a-3p, miR-425-3p, and miR-20a-5p in patients with IgA nephropathy and their correlation with selected clinical parameters. METHODS This study included 44 patients with IgA nephropathy and 46 control subjects. RESULTS The results of our study indicated that in patients with IgA nephropathy, the increased plasma levels of miR-148a-3p and miR-425-3p correlated negatively with eGFR values. According to the Haas classification, plasma levels of miR-20a-5p were statistically significantly increased in patients with histopathological changes classified as Stages 3, 4, and 5 compared with patients with histopathological changes classified as Stages 1 and 2. CONCLUSIONS The results of our study suggest the possible involvement of miR-148a-3p, miR-425-3p, and miR-20a-5p in the pathogenesis of IgA nephropathy.
Collapse
Affiliation(s)
- Jarosław Przybyciński
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.P.); (E.K.); (L.D.)
| | - Michał Czerewaty
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Ewa Kwiatkowska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.P.); (E.K.); (L.D.)
| | - Violetta Dziedziejko
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (V.D.); (K.S.)
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, 70-111 Szczecin, Poland; (V.D.); (K.S.)
| | - Leszek Domański
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, 70-111 Szczecin, Poland; (J.P.); (E.K.); (L.D.)
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| |
Collapse
|
45
|
Smerdu V, Ugwoke CK, Šink Ž. Co-expression of MyHC-15 with other known isoforms in rat muscle spindles. Eur J Histochem 2025; 69:4192. [PMID: 40126372 PMCID: PMC11983093 DOI: 10.4081/ejh.2025.4192] [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: 02/03/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
Muscle spindles are skeletal muscle sensory receptors composed of intrafusal fibres, partially encapsulated by connective tissue capsule. This capsule encloses the central A and B regions while leaving the distal C region extracapsular. Several past studies in rat have shown that muscle spindles typically contain a single bag1 fibre, a single bag2 fibre, and two smaller chain fibres. Intrafusal fibres co-express multiple myosin heavy chain (MyHC) isoforms: -slow or -1, -slow-tonic, -α, -2a, -2b, -embryonic, and -neonatal. While MyHC-2x was previously thought absent, the recently discovered MyHC-15 isoform has been identified in the C region of rat bag fibres. Using antibodies specific for nine MyHC isoforms and analyzing four different rat skeletal muscles-soleus, extensor digitorum longus, and the lateral and medial heads of gastrocnemius-we aimed to further characterize the co-expression pattern of MyHC-15 with other known isoforms and to determine whether MyHC-2x is expressed in rat intrafusal fibres. While rodents are widely used as animal models in skeletal muscle research, notable species-specific differences in MyHC isoform expression exist. Our findings revealed that MyHC-15 expression in rat intrafusal fibres is less abundant than in human fibres. MyHC-15 was primarily observed in bag fibres but was not detected in the C region, contrary to previous reports in both rat and human. We confirmed the absence of MyHC-2x in rat intrafusal fibres. Similarly, MyHC-embryonic and -neonatal were not detected in the analyzed spindles, suggesting that previously used antibodies may have cross-reacted with MyHC-2a and -2b. While our results partially corroborate previous extensive studies, discrepancies suggest that MyHC expression in intrafusal fibres varies not only along the fibre length but also across muscles.
Collapse
Affiliation(s)
- Vika Smerdu
- Institute of Anatomy, Faculty of Medicine, University of Ljubljana, Slovenia
| | | | | |
Collapse
|
46
|
Khalil A, Al Toufaily S, Shebaby W, Hage ME, Mroue D, Faour W, Mroueh M. Lebanese Cannabis sativa L. extract protects from cisplatin-induced nephrotoxicity in mice by inhibiting podocytes apoptosis. J Cannabis Res 2025; 7:3. [PMID: 39819647 PMCID: PMC11737194 DOI: 10.1186/s42238-025-00260-4] [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: 11/05/2023] [Accepted: 12/19/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Cisplatin is an anti-cancer drug used to treat a plethora of solid tumors. However, it is associated with dose dependent nephrotoxicity limiting its use as anticancer agent. OBJECTIVE The current study aimed to investigate the nephroprotective effect of native Lebanese Cannabis sativa in both in vitro and in vivo mice model of cisplatin-induced nephrotoxicity. METHODS Podocytes cell viability was assessed using MTS assay with cisplatin (30µM) in presence or absence of Cannabis oil extract (COE) at 0.5, 1 and 2µg/ml for 24h. Acute renal injury was established in adult female C57BL/6 mice with 20mg/kg, i.p. single dose cisplatin. Mice were divided into control group (vehicle), COE group, cisplatin group and cisplatin plus COE (2.5, 5 and 20mg/kg, i.p.). Animal body weight, serum creatinine, blood urea nitrogen (BUN), and proteinuria were measured. RESULTS Cell viability assay and western blot analysis revealed that COE prevented apoptosis induced by cisplatin in cultured immortalized rat podocytes. In addition, in vitro scratch assay demonstrated the ability of COE to promote and restore the migratory capacity of podocytes in cisplatin-treated cells. Interestingly, COE treatment improved urinary and serum parameters characterized by a significant decrease in serum creatinine, urea, and proteinuria at various COE doses. Western blot analysis showed that COE inhibited COX-2 protein induction as well as apoptosis marker production (Bax/Bcl2 ratio) in cisplatin-treated mice when compared to mice treated with cisplatin alone. CONCLUSION Collectively, the aforementioned findings indicate that COE could be a promising approach to protect against cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Alia Khalil
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box36, Byblos, Lebanon
| | - Sahar Al Toufaily
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| | - Wassim Shebaby
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| | - Marissa El Hage
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
- Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, Brazil
| | - Dima Mroue
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos, Lebanon
| | - Wissam Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box36, Byblos, Lebanon.
| | - Mohamad Mroueh
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
47
|
Giammona A, Galuzzi BG, Imperia E, Gervasoni C, Remedia S, Restaneo L, Nespoli M, De Gara L, Tani F, Cicala M, Guarino MPL, Porro D, Cerasa A, Lo Dico A, Altomare A, Bertoli G. Chronic Gastrointestinal Disorders and miRNA-Associated Disease: An Up-to-Date. Int J Mol Sci 2025; 26:413. [PMID: 39796266 PMCID: PMC11720538 DOI: 10.3390/ijms26010413] [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: 12/05/2024] [Revised: 12/27/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Chronic gastrointestinal disorders such as inflammatory bowel diseases (IBDs) and irritable bowel syndrome (IBS) impose significant health burdens globally. IBDs, encompassing Crohn's disease and ulcerative colitis, are multifactorial disorders characterized by chronic inflammation of the gastrointestinal tract. On the other hand, IBS is one of the principal gastrointestinal tract functional disorders and is characterized by abdominal pain and altered bowel habits. Although the precise etiopathogenesis of these disorders remains unclear, mounting evidence suggests that non-coding RNA molecules play crucial roles in regulating gene expression associated with inflammation, apoptosis, oxidative stress, and tissue permeability, thus influencing disease progression. miRNAs have emerged as possible reliable biomarkers, as they can be analyzed in the biological fluids of patients at a low cost. This review explores the roles of miRNAs in IBDs and IBS, focusing on their involvement in the control of disease hallmarks. By an extensive literature review and employing bioinformatics tools, we identified the miRNAs frequently studied concerning these diseases. Ultimately, specific miRNAs could be proposed as diagnostic biomarkers for IBDs and IBS. Their ability to be secreted into biofluids makes them promising candidates for non-invasive diagnostic tools. Therefore, understanding molecular mechanisms through the ways in which they regulate gastrointestinal inflammation and immune responses could provide new insights into the pathogenesis of IBDs and IBS and open avenues for miRNA-based therapeutic interventions.
Collapse
Affiliation(s)
- Alessandro Giammona
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Bruno Giovanni Galuzzi
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Elena Imperia
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
| | - Clarissa Gervasoni
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Sofia Remedia
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Dipartimento di Scienze della Terra e del Mare (DISTEM), Università di Palermo, Via Archirafi, 22, 90123 Palermo, Italy
| | - Laura Restaneo
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
| | - Martina Nespoli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Laura De Gara
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
| | - Flaminia Tani
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Michele Cicala
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.C.); (M.P.L.G.)
- Unit of Gastroenterology, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Michele Pier Luca Guarino
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.C.); (M.P.L.G.)
- Unit of Gastroenterology, Fondazione Policlinico Campus Bio-Medico di Roma, Via Alvaro del Portillo 200, 00128 Rome, Italy
| | - Danilo Porro
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano Bicocca, 20126 Milan, Italy
| | - Antonio Cerasa
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Alessia Lo Dico
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Annamaria Altomare
- Department of Sciences and Technologies for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (E.I.); (L.R.); (L.D.G.); (A.A.)
- Research Unit of Gastroenterology, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo 21, 00128 Rome, Italy; (M.C.); (M.P.L.G.)
| | - Gloria Bertoli
- Istituto di Bioimmagini e Sistemi Biologici Complessi (IBSBC), National Research Council (CNR), Segrate, 20054 Milan, Italy; (A.G.); (B.G.G.); (C.G.); (S.R.); (M.N.); (F.T.); (D.P.); (A.C.)
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
48
|
Wang X, Zhou XJ, Qiao X, Falchi M, Liu J, Zhang H. The evolving understanding of systemic mechanisms in organ-specific IgA nephropathy: a focus on gut-kidney crosstalk. Theranostics 2025; 15:656-681. [PMID: 39744688 PMCID: PMC11671385 DOI: 10.7150/thno.104631] [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: 10/04/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
The interplay between multiple organs, known as inter-organ crosstalk, represents a complex and essential research domain in understanding the mechanisms and therapies for kidney diseases. The kidneys not only interact pathologically with many other organs but also communicate with other systems through various signaling pathways. It is of paramount importance to comprehend these mechanisms for the development of more efficient therapeutic strategies. Despite extensive research in IgA nephropathy (IgAN), the most common kidney disease, the elaboration mechanism of IgAN remains challenging. Numerous studies suggest that alterations in the intestinal microbiome and its metabolites are pivotal in the progression of IgAN, opening new avenues for understanding its mechanisms. Interestingly, certain presumed probiotics, such as Akkermansia muciniphila, have been implicated in the onset of IgAN, making the exploration of gut microbiota in the context of IgAN pathogenesis even more intriguing. In this review, we summarize the status of gut microbiology studies of IgAN and explore the possible mechanisms and intervention prospects. Future research and treatment directions may increasingly emphasize systemic, multi-organ combined interventions to decelerate the advancement of kidney disease and enhance the overall prognosis of patients.
Collapse
Affiliation(s)
- Xin Wang
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xu-Jie Zhou
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xue Qiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jing Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Science, Beijing, 100190, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Beijing, China
- Peking University Institute of Nephrology, Beijing, China
- Key Laboratory of Renal Disease, Ministry of Health of China, Beijing, China
- Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
49
|
Li R, Wang M, Li J, Zhu L, Xie X, Wang H, Zhang X, Tian W, Zhang Y, Dong Y, Zan J, Li H, Zhang Y, Zhou X, Shi S, Shu C, Liu L, Jin J, Lv J, Zhang H. Lactobacillus casei Cell Wall Extract and Production of Galactose-Deficient IgA1 in a Humanized IGHA1 Mouse Model. J Am Soc Nephrol 2025; 36:60-72. [PMID: 39172518 PMCID: PMC11706567 DOI: 10.1681/asn.0000000000000465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 08/16/2024] [Indexed: 08/24/2024] Open
Abstract
Key Points We generated a transgenic mouse model expressing the human IgA1 heavy chain, which has a hinge region with rich O -linked glycosylation. After inflammatory stimulation, the mouse model showed elevated galactose-deficient IgA1 levels in the serum. Coupled with complement H factor mutant, the mice model exhibited glomerular lesions, associated with hematuria and albuminuria like IgA nephropathy. Background IgA nephropathy is the most common primary glomerulonephritis worldwide, and there is emerging evidence linking galactose-deficient IgA1 (Gd-IgA1) to the pathogenesis of the disease. However, mouse models that can be used to study Gd-IgA1's origin of production, biochemical characteristics, and immune reactivity are lacking. Methods We generated a humanized IgA1 mouse model with transgenic expression of the human IGHA1 gene from the mouse chromosomal locus of IgA heavy chain. The IGHA1 +/+ mice were crossed with complement factor H heterozygous mutant (FHW/R) to generate IGHA1 +/+FHW/R mice. IGHA1 +/+ mice were exposed to different levels of environmental pathogens in the first 4 months, as housed in germ-free, specific pathogen–free, or conventional environments. In addition, wild-type C57BL/6J mice, IGHA1 +/+ mice, and IGHA1 +/+FHW/R mice were inoculated with Lactobacillus casei cell wall extract (LCWE) mixed with complete Freund's adjuvant (CFA) at 2 months of age to develop a mouse model of IgA nephropathy. Results Elevated levels of human IgA1 in blood circulation and mucosal sites were observed in IGHA1 +/+ mice from exposure to pathogens. Compared with buffer-treated control mice, LCWE plus CFA-treated mice had moderately elevated levels of circulating human IgA1 (by one-fold) and human IgA1 immune complexes (by two-fold). Serum Gd-IgA1 levels increased four-fold after LCWE treatments. Analyses of the O -glycopeptides of the IgA1 hinge region confirmed hypogalactosylation of IgA1, with the variety of the glycoforms matching those seen in clinical samples. Furthermore, LCWE induced persistent IgA1 and C3 deposition in the glomerular mesangial areas in association with mesangial expansion and hypercellularity, which are frequently observed in IgA nephropathy biopsies. The IGHA1+/+FHW/R mice stimulated with LCWE and CFA developed albuminuria and hematuria. Conclusions We observed elevated plasma Gd-IgA1 levels with kidney deposition of IgA1 in the IGHA1 +/+ mice after LCWE and CFA. In conjunction with factor H mutation, the mice exhibited severe glomerular alterations, associated with hematuria and albuminuria in resemblance of clinical IgA nephropathy.
Collapse
Affiliation(s)
- Run Li
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Manliu Wang
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jingyi Li
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Li Zhu
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xinfang Xie
- Department of Nephrology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Wang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Xu Zhang
- Laboratory of Electron Microscopy, Pathological Center, Peking University First Hospital, Beijing, China
| | - Wenmin Tian
- Department of Biochemistry and Biophysics, Center for Precision Medicine Multi-Omics Research, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yong Zhang
- Department of Nephrology, Institutes for Systems Genetics, West China Hospital, Sichuan University, Chengdu, China
| | - Yaping Dong
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Jincan Zan
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Hongyu Li
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
| | - Yuemiao Zhang
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Xujie Zhou
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Sufang Shi
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Chutian Shu
- Shanghai Alezyme Pharmaceuticals Ltd., Shanghai, China
| | - Lijun Liu
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Jing Jin
- Division of Nephrology and Hypertension, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Jicheng Lv
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| | - Hong Zhang
- Renal Division, Key Laboratory of Renal Disease, Ministry of Health of China, Peking University Institute of Nephrology, Peking University First Hospital, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediate Kidney Disease, Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
| |
Collapse
|
50
|
Zhang Y, Cao P, Qin D, Zhao Y, Chen X, Ma P. Anti-inflammatory, anti-colitis, and antioxidant effects of columbianadin against DSS-induced ulcerative colitis in rats via alteration of HO-1/Nrf2 and TLR4-NF-κB signaling pathway. Inflammopharmacology 2025; 33:341-352. [PMID: 39757276 DOI: 10.1007/s10787-024-01630-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a significant inflammatory bowel disease (IBD) that typically arises from chronic inflammation of the intestinal tract. Report suggest that anti-inflammatory drug plays a crucial role in the protection of UC. The recent study demonstrated that columbianadin has a protective effect against UC induced by dextran sulfate sodium (DSS) in rats through the modulation of HO-1/Nrf2 and TLR4-NF-κB signaling pathways. MATERIAL AND METHODS In this study, Swiss Wistar rats were utilized, and UC was induced using 2% DSS. The treatment regimen included oral administration of columbianadin (5, 10 and 15 mg/kg) and sulfasalazine to the rats. The body weight, spleen index, disease activity index (DAI), colon length, food and water intake were estimated. Moreover, antioxidant, cytokines, inflammatory and apoptosis parameters were determined. mRNA expression levels were also quantitatively analyzed. RESULTS Columbianadin treatment significantly (P < 0.001) boosted the body weight and suppressed the DAI. Columbianadin significantly (P < 0.001) enhanced the colon length and repressed the spleen index along with enhanced food and water intake. Columbianadin significantly (P < 0.001) suppressed the level of lactate dehydrogenase (LDH), myeloperoxidase (MPO) and altered the level of oxidative stress parameters such as catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GR), glutathione peroxidase (GPx), malonaldehyde (MDA), nitric oxide (NO), SA; cytokines level such as interleukin (IL)-1, 1β, 6, 10, 17, 18, TNF-α; inflammatory parameters viz., cyclooxygenase-2 (COX-2), prostaglandin (PGE2), inducible nitric oxide synthetase (iNOS), nuclear factor kappa B (NF-κB), transforming growth factor (TGF-β); apoptosis parameters include Bax, Bcl-2, Bcl-2/Bax ratio, caspase-1 and A-caspase-3 activity, respectively. Columbianadin significantly altered the mRNA expression of IFN-γ, IL-6, IL-1β, IL-8, TNF-α, NF-κB, TLR4, Bcl-2, caspase-9, Bax, p38, ASC, MCP-1, ZO-1, and Ocln. While this study focused on COX-2 modulation as a marker of inflammatory response, no direct measurements or inferences were made regarding leukotriene activity, which involves a separate lipoxygenase pathway. CONCLUSION Columbianadin exhibited the protective effect against DSS-induced UC via alteration of HO-1/Nrf2 and TLR4-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yanping Zhang
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, No.99 Longcheng street, Xiao dian District, Taiyuan, 030032, China
- Faculty of Graduate Studies, Shanxi Medical University, NO.56 Road, NO. Xinjiannan Yingze District, Taiyuan, 030000, China
| | - Ping Cao
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, No.99 Longcheng street, Xiao dian District, Taiyuan, 030032, China
| | - Dongyuan Qin
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, No.99 Longcheng street, Xiao dian District, Taiyuan, 030032, China
| | - Ying Zhao
- Department of Gastroenterology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, No.99 Longcheng street, Xiao dian District, Taiyuan, 030032, China
| | - Xing Chen
- Early Gastrointestinal Cancer Diagnosis and Treatment Research Center, First Hospital of Shanxi Medical University, NO.85 Xinjian nan Road, Yingze District, Taiyuan, 030000, China.
- Department of Gastroenterology, First Hospital of Shanxi Medical University, NO.56 Jiefangnan Road, Taiyuan, 030000, China.
| | - Peng Ma
- General Surgery Department, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|