1
|
Hassan FE, El-Mosallamy AEMK, Khalifa MM, Aljuaydi SH, Ali ME, Hosny SA, Bastawy N. Evaluating the potential impact of sodium-glucose cotransporter-2 inhibitor "canagliflozin" on the hepatic damage triggered by hypertension in rats. Microsc Res Tech 2024; 87:2929-2942. [PMID: 39051105 DOI: 10.1002/jemt.24665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/06/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Hypertension (HTN) is a prevalent chronic disease. HTN and liver disease association is extensively noted. Thus, finding a medication that can alleviate HTN and its accompanying liver insult would be promising. This study investigated the potential impacts of canagliflozin "sodium-glucose cotransporter-2 inhibitor" on the liver of the Nω-nitro-L-arginine methyl ester (L-NAME)-induced HTN rat model. Twenty-four adult male rats were divided into four groups; negative control group, canagliflozin group, L-NAME group: 50 mg/kg of L-NAME was injected daily for 5 weeks and L-NAME + canagliflozin group: 1 week after L-NAME injection both L-NAME + canagliflozin (40 mg/kg) were given concomitantly daily for further 4 weeks. Liver functions, serum lipid profile, hepatic oxidative/nitrative stress biomarkers, gene expression of lipogenic enzymes, B-cell lymphoma 2 (Bcl2), and DNA fragmentation, were measured. Besides, hepatic histology and immunohistochemistry of nuclear factor kappa B (NF-κB) and endothelial nitric oxide synthase (eNOS) were assessed. Canagliflozin improved hepatic lipogenesis via the downregulation of fatty acid synthase (FAS) and transcriptional regulatory element binding protein 1c (SREBP1c) genes leading to an improved serum lipid profile. Further, canagliflozin modified the eNOS/inducible nitric oxide synthase (iNOS) pathway and decreased the NF-κB immunoreactivity besides restoring the oxidants-antioxidants balance; increased reduced glutathione concomitant with declined malondialdehyde. This improvement of the liver was mirrored by the significant restoration of liver architecture and confirmed by the preserved liver DNA content and upregulation of the antiapoptotic Bcl2 mRNA level and attenuation of the alanine transaminase, aspartate aminotransferase. In conclusion, canagliflozin is a promising anti-hypertensive and hepatic-supportive medication. RESEARCH HIGHLIGHTS: Canagliflozin's antioxidant, anti-inflammatory, anti-lipogenic, and antiapoptotic characteristics mitigate remote liver compromise caused by hypertension. Canagliflozin can be exploited as a hepatoprotective and antihypertensive medication.
Collapse
Affiliation(s)
- Fatma E Hassan
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
- General Medicine Practice Program, Department of Physiology, Batterjee Medical College, Jeddah, Saudi Arabia
| | - Aliaa E M K El-Mosallamy
- Department of Pharmacology, Medical Research and Clinical Studies Institute, National Research Center, Giza, Egypt
| | - Mohamed Mansour Khalifa
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
- Department of Human Physiology, College of Medicine, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Samira H Aljuaydi
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Merhan E Ali
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sara Adel Hosny
- Histology Department, Faculty of Medicine, Cairo University, Giza, Egypt
| | - Nermeen Bastawy
- Medical Physiology Department, Kasr Alainy, Faculty of Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
2
|
Prasad MK, Victor PS, Ganesh GV, Juttada U, Kumpatla S, Viswanathan V, Ramkumar KM. Sodium-Glucose Cotransporter-2 Inhibitor Suppresses Endoplasmic Reticulum Stress and Oxidative Stress in Diabetic Nephropathy Through Nrf2 Signaling: A Clinical and Experimental Study. J Clin Pharmacol 2024; 64:1193-1203. [PMID: 38831713 DOI: 10.1002/jcph.2465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 05/08/2024] [Indexed: 06/05/2024]
Abstract
Diabetic nephropathy (DN), a severe complication of type 2 diabetes mellitus (T2DM), is marked by heightened endoplasmic reticulum stress (ERS) and oxidative stress (OS) due to protein misfolding and free radical generation. We investigated the sodium-glucose co-transporter-2 inhibitor (SGLT2i), canagliflozin (Cana), in alleviating ERS and OS in DN patients and THP-1 cells under hyperglycemic condition. A total of 120 subjects were divided into four groups, with 30 subjects in each group: healthy controls, T2DM individuals, DN patients receiving standard treatment, and those treated with Cana. The control group had no history of diabetes, cardiovascular or renal diseases, or other comorbidities. Cana was administered at doses of either 100 or 300 mg per day based on the estimated glomerular filtration rate (eGFR) value of DN individuals, with a mean follow-up of 6 months. Additionally, THP-1 monocytes were exposed to HGM (33.3 mM glucose with a cytokine cocktail of TNF-α and IFN-γ at 50 ng/mL each) to evaluate the relative levels of ERS, OS markers, and nuclear factor erythroid 2-related factor 2 (Nrf2), the transcription factor regulating cellular redox, which is downregulated in diabetes. Our results revealed that ERS markers GRP78 and PERK, as well as OS markers TXNIP and p22phox, were elevated in both DN patients and HGM-treated THP-1 monocytes and were reduced by Cana intervention. Furthermore, Cana regulated the phosphorylation of Nrf2, Akt, and EIF2α in HGM-treated monocytes. In conclusion, our findings highlight the role of Cana in activating Nrf2, thereby attenuating ERS and OS to mitigate DN progression.
Collapse
Affiliation(s)
- Murali Krishna Prasad
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Paul S Victor
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Goutham V Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| | - Udayama Juttada
- Department of Biochemistry and Molecular Genetics, Prof. M. Viswanathan's Diabetes Research Center, M.V. Hospital for Diabetes, Royapuram Chennai, Tamilnadu, India
| | - Satyavani Kumpatla
- Department of Biochemistry and Molecular Genetics, Prof. M. Viswanathan's Diabetes Research Center, M.V. Hospital for Diabetes, Royapuram Chennai, Tamilnadu, India
| | - Vijay Viswanathan
- Department of Biochemistry and Molecular Genetics, Prof. M. Viswanathan's Diabetes Research Center, M.V. Hospital for Diabetes, Royapuram Chennai, Tamilnadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, India
| |
Collapse
|
3
|
Liang X, Hou X, Bouhamdan M, Sun Y, Song Z, Rajagopalan C, Jiang H, Wei HG, Song J, Yang D, Guo Y, Zhang Y, Mou H, Zhang J, Chen YE, Sun F, Jin JP, Zhang K, Xu J. Sotagliflozin attenuates liver-associated disorders in cystic fibrosis rabbits. JCI Insight 2024; 9:e165826. [PMID: 38358827 PMCID: PMC10972622 DOI: 10.1172/jci.insight.165826] [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: 10/03/2022] [Accepted: 02/09/2024] [Indexed: 02/17/2024] Open
Abstract
Mutations in the cystic fibrosis (CF) transmembrane conductance regulator (CFTR) gene lead to CF, a life-threating autosomal recessive genetic disease. While recently approved Trikafta dramatically ameliorates CF lung diseases, there is still a lack of effective medicine to treat CF-associated liver disease (CFLD). To address this medical need, we used a recently established CF rabbit model to test whether sotagliflozin, a sodium-glucose cotransporter 1 and 2 (SGLT1/2) inhibitor drug that is approved to treat diabetes, can be repurposed to treat CFLD. Sotagliflozin treatment led to systemic benefits to CF rabbits, evidenced by increased appetite and weight gain as well as prolonged lifespan. For CF liver-related phenotypes, the animals benefited from normalized blood chemistry and bile acid parameters. Furthermore, sotagliflozin alleviated nonalcoholic steatohepatitis-like phenotypes, including liver fibrosis. Intriguingly, sotagliflozin treatment markedly reduced the otherwise elevated endoplasmic reticulum stress responses in the liver and other affected organs of CF rabbits. In summary, our work demonstrates that sotagliflozin attenuates liver disorders in CF rabbits and suggests sotagliflozin as a potential drug to treat CFLD.
Collapse
Affiliation(s)
- Xiubin Liang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Xia Hou
- Department of Physiology, and
| | | | - Yifei Sun
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Zhenfeng Song
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | - Jun Song
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Dongshan Yang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yanhong Guo
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Yihan Zhang
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hongmei Mou
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jifeng Zhang
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Y. Eugene Chen
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| | - Fei Sun
- Department of Physiology, and
| | | | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jie Xu
- Center for Advanced Models for Translational Sciences and Therapeutics, University of Michigan Medical Center, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Yang G, Liu R, Rezaei S, Liu X, Wan YJY. Uncovering the Gut-Liver Axis Biomarkers for Predicting Metabolic Burden in Mice. Nutrients 2023; 15:3406. [PMID: 37571345 PMCID: PMC10421148 DOI: 10.3390/nu15153406] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Western diet (WD) intake, aging, and inactivation of farnesoid X receptor (FXR) are risk factors for metabolic and chronic inflammation-related health issues ranging from metabolic dysfunction-associated steatotic liver disease (MASLD) to dementia. The progression of MASLD can be escalated when those risks are combined. Inactivation of FXR, the receptor for bile acid (BA), is cancer prone in both humans and mice. The current study used multi-omics including hepatic transcripts, liver, serum, and urine metabolites, hepatic BAs, as well as gut microbiota from mouse models to classify those risks using machine learning. A linear support vector machine with K-fold cross-validation was used for classification and feature selection. We have identified that increased urine sucrose alone achieved 91% accuracy in predicting WD intake. Hepatic lithocholic acid and serum pyruvate had 100% and 95% accuracy, respectively, to classify age. Urine metabolites (decreased creatinine and taurine as well as increased succinate) or increased gut bacteria (Dorea, Dehalobacterium, and Oscillospira) could predict FXR deactivation with greater than 90% accuracy. Human disease relevance is partly revealed using the metabolite-disease interaction network. Transcriptomics data were also compared with the human liver disease datasets. WD-reduced hepatic Cyp39a1 (cytochrome P450 family 39 subfamily a member 1) and increased Gramd1b (GRAM domain containing 1B) were also changed in human liver cancer and metabolic liver disease, respectively. Together, our data contribute to the identification of noninvasive biomarkers within the gut-liver axis to predict metabolic status.
Collapse
Affiliation(s)
- Guiyan Yang
- Department of Medical Pathology, Laboratory Medicine in Sacramento, University of California, Davis, CA 95817, USA;
| | - Rex Liu
- Department of Computer Science, University of California, Davis, CA 95616, USA; (R.L.); (S.R.); (X.L.)
| | - Shahbaz Rezaei
- Department of Computer Science, University of California, Davis, CA 95616, USA; (R.L.); (S.R.); (X.L.)
| | - Xin Liu
- Department of Computer Science, University of California, Davis, CA 95616, USA; (R.L.); (S.R.); (X.L.)
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology, Laboratory Medicine in Sacramento, University of California, Davis, CA 95817, USA;
| |
Collapse
|
5
|
An Z, Sun Y, Shi C, Liu L. Metabonomic and transcriptomic analyses of Tripterygium glycosides tablet-induced hepatotoxicity in rats. Drug Chem Toxicol 2022:1-15. [PMID: 35603506 DOI: 10.1080/01480545.2022.2077360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We aimed to explore novel biomarkers involved in alterations of metabolism and gene expression related to the hepatotoxic effects of Tripterygium glycosides tablet (TGT) in rats. Rats were randomly divided into groups based on oral administration of TGTs for 6 weeks: control, low-dose (9.5 mg/kg), and high-dose (18.9 mg/kg). Serum samples and total liver RNA were subjected to metabonomic and transcriptomic analyses. Thirteen metabolites were significantly up-regulated by liver injury induced by Tripterygium glycosides. Five potential biomarkers were more sensitive than Alanine aminotransferase (ALT) for accurate and timely prediction of hepatic damage. The four metabolic pathways most obviously regulated by hepatotoxicity were D-glutamine and D-glutamate metabolism, alanine, aspartate and glutamate metabolism, ether lipid metabolism, and tryptophan metabolism. Transcriptomics revealed significant differences in 1792 mRNAs and 400 long non-coding (lnc) RNAs. Dysregulated lncRNAs in the TGT-induced hepatotoxicity group were associated with genes involved in amino acid metabolism using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis. Up-regulated expression of Ehhadh, Gpt, and Got1, and down-regulated expression of dopa decarboxylase (Ddc), Cyp1a2, Ido2, Aldh1b1, and asparagine synthetase (Asns) was validated by quantitative real-time PCR. This multiomics study has elucidated the relationship between amino metabolism and liver injury, revealing potential biomarkers.
Collapse
Affiliation(s)
- Zhuoling An
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yuan Sun
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Chen Shi
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Lihong Liu
- Pharmacy Department of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Du S, Shi H, Xiong L, Wang P, Shi Y. Canagliflozin mitigates ferroptosis and improves myocardial oxidative stress in mice with diabetic cardiomyopathy. Front Endocrinol (Lausanne) 2022; 13:1011669. [PMID: 36313744 PMCID: PMC9616119 DOI: 10.3389/fendo.2022.1011669] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 09/27/2022] [Indexed: 11/13/2022] Open
Abstract
Canagliflozin (Cana), an anti-diabetes drug belongs to sodium-glucose cotransporter 2 inhibitor, is gaining interest because of its extra cardiovascular benefits. Ferroptosis is a new mode of cell death, which can promote the occurrence of diabetic cardiomyopathy (DCM). Whether Cana can alleviate DCM by inhibiting ferroptosis is the focus of this study. Here, we induced DCM models in diabetic C57BL6 mice and treated with Cana. Meanwhile, in order to exclude its hypoglycemic effect, the high glucose model in H9C2 cells were established. In the in vivo study, we observed that Cana could effectively alleviate the damage of cardiac function in DCM mice, including the increasing of lactate dehydrogenase (LDH) and cardiac troponin I (cTnI), the alleviating of myocardial fiber breakage, inflammation, collagen fiber deposition and mitochondrial structural disorder. We evaluated reactive oxygen species (ROS) levels by DCFH-DA and BODIPY 581/591 C11, in vitro Cana reduced ROS and lipid ROS in H9C2 cells induced by high glucose. Meanwhile, JC-1 fluorochrome assay showed that the decreased mitochondrial membrane potential (MMP) was increased by Cana. Furthermore, the inhibitory effects of Cana on myocardial oxidative stress and ferroptosis were verified in vivo and in vitro by protein carbonyl (PCO), malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione (GSH). As a key inducer of ferroptosis, the deposition of total iron and Fe2+ can be inhibited by Cana both in vivo and in vitro. In addition, western blot results indicated that the expression of ferritin heavy-chain (FTN-H) was down-regulated, and cystine-glutamate antiporter (xCT) was up-regulated by Cana in DCM mice and cells, suggesting that Cana inhibit ferroptosis by balancing cardiac iron homeostasis and promoting the system Xc-/GSH/GPX4 axis in DCM. These findings underscore the fact that ferroptosis plays an important role in the development and progression of DCM and targeting ferroptosis may be a novel strategy for prevention and treatment. In conclusion, Cana may exert some of its cardiovascular benefits by attenuating ferroptosis.
Collapse
Affiliation(s)
- Shuqin Du
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
- School of Medicine, Jiaxing University, Jiaxing, China
| | - Hanqiang Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
| | - Lie Xiong
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
| | - Ping Wang
- School of Pharmacy, Zhejiang University of Technology, Hangzhou, China
| | - Yanbo Shi
- Central Laboratory of Molecular Medicine Research Center, Jiaxing Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Jiaxing, China
- Jiaxing Key Laboratory of Diabetic Angiopathy Research, Jiaxing, China
- *Correspondence: Yanbo Shi,
| |
Collapse
|