1
|
Song JH, Kim SJ, Kwon S, Jeon SY, Park SE, Choi SJ, Oh SY, Jeon HB, Chang JW. Nervonic acid improves fat transplantation by promoting adipogenesis and angiogenesis. Int J Mol Med 2024; 54:108. [PMID: 39364738 PMCID: PMC11517738 DOI: 10.3892/ijmm.2024.5432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 08/19/2024] [Indexed: 10/05/2024] Open
Abstract
Adipose tissue engraftment has become a promising strategy in the field of regenerative surgery; however, there are notable challenges associated with it, such as resorption of 50‑90% of the transplanted fat or cyst formation due to fat necrosis after fat transplantation. Therefore, identifying novel materials or methods to improve the engraftment efficiency is crucial. The present study investigated the effects of nervonic acid (NA), a monounsaturated very long‑chain fatty acid, on adipogenesis and fat transplantation, as well as its underlying mechanisms. To assess this, NA was used to treat cells during adipogenesis in vitro, and the expression levels of markers, including PPARγ and CEBPα, and signaling molecules were detected through reverse transcription‑quantitative PCR and western blotting. In addition, NA was mixed with fat grafts in in vivo fat transplantation, followed by analysis through Oil Red O staining, hematoxylin & eosin staining and immunohistochemistry. It was demonstrated that NA treatment accelerated adipogenesis through activation of the Akt/mTOR pathway and inhibition of Wnt signaling. NA treatment enriched the expression of Akt/mTOR signaling‑related genes, and increased the expression of genes involved in angiogenesis and fat differentiation in human mesenchymal stem cells (MSCs). Additionally, NA effectively improved the outcome of adipose tissue engraftment in mice. Treatment of grafts with NA at transplantation reduced the resorption of transplanted fat and increased the proportion of perilipin‑1+ adipocytes with a lower portion of vacuoles in mice. Moreover, the NA‑treated group exhibited a reduced pro‑inflammatory response and had more CD31+ vessel structures, which were relatively evenly distributed among viable adipocytes, facilitating successful engraftment. In conclusion, the present study demonstrated that NA may not only stimulate adipogenesis by regulating signaling pathways in human MSCs, but could improve the outcome of fat transplantation by reducing inflammation and stimulating angiogenesis. It was thus hypothesized that NA could serve as an adjuvant strategy to enhance fat engraftment in regenerative surgery.
Collapse
Affiliation(s)
- Jae Hoon Song
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sun Jeong Kim
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Soojin Kwon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Su Yeon Jeon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Sang Eon Park
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Suk-Joo Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Soo-Young Oh
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Hong Bae Jeon
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
| | - Jong Wook Chang
- Cell and Gene Therapy Institute, ENCell Co., Ltd., Seoul 06072, Republic of Korea
- Cell and Gene Therapy Institute, Samsung Medical Center, Seoul 06351, Republic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology (SAIHST), Sungkyunkwan University, Seoul 06355, Republic of Korea
| |
Collapse
|
2
|
Xiao G, Yang M, Zeng Z, Tang R, Jiang J, Wu G, Xie C, Jia D, Bi X. Investigation into the anti-inflammatory mechanism of Pothos chinensis (Raf.) Merr. By regulating TLR4/MyD88/NF-κB pathway: Integrated network pharmacology, serum pharmacochemistry, and metabolomics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118520. [PMID: 38964626 DOI: 10.1016/j.jep.2024.118520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/06/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Inflammation is directly related to disease progression and contributes significantly to the global burden of disease. Pothos chinensis (Raf.) Merr. (PCM) is commonly used in Yao medicine in China to treat tumors, and orthopedic illnesses such as knee osteoarthritis, and rheumatic bone discomfort. PCM was found to have significant anti-inflammatory properties in previous studies. AIM OF THE STUDY To explore the active compounds of PCM and their anti-inflammatory pharmacological mechanisms through an integrated strategy of serum pharmacochemistry, network pharmacology, and serum metabolomics. MATERIALS AND METHODS The qualitative and quantitative analyses of the chemical components of PCM were performed using UPLC-QTOF-MS/MS and UPLC, respectively, and the prototype components of PCM absorbed into the blood were analyzed. Based on the characterized absorbed into blood components, potential targets and signaling pathways of PCM anti-inflammatory were found using network pharmacology. Furthermore, metabolomics studies using UPLC-QTOF-MS/MS identified biomarkers and metabolic pathways related to the anti-inflammatory effects of PCM. Finally, the hypothesized mechanisms were verified by in vivo and in vitro experiments. RESULTS Forty chemical components from PCM were identified for the first time, and seven of them were quantitatively analyzed, while five serum migratory prototype components were found. Network pharmacology KEGG enrichment analysis revealed that arachidonic acid metabolism, Tyrosine metabolism, TNF signaling pathway, NF-κB signaling pathway, and phenylalanine metabolism were the main signaling pathways of PCM anti-inflammatory. Pharmacodynamic results showed that PCM ameliorated liver injury and inflammatory cell infiltration and downregulated protein expression of IL-1β, NF-κB p65, and MyD88 in the liver. Metabolomics studies identified 53 different serum metabolites, mainly related to purine and pyrimidine metabolism, phenylalanine metabolism, primary bile acid biosynthesis, and glycerophospholipid metabolism. The comprehensive results demonstrated that the anti-inflammatory modulatory network of PCM was related to 5 metabolites, 3 metabolic pathways, 7 targets, and 4 active components of PCM. In addition, molecular docking identified the binding ability between the active ingredients and the core targets, and the anti-inflammatory efficacy of the active ingredients was verified by in vitro experiments. CONCLUSION Our study demonstrated the anti-inflammatory effect of PCM, and these findings provide new insights into the active ingredients and metabolic mechanisms of PCM in anti-inflammation.
Collapse
Affiliation(s)
- Guanlin Xiao
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine/Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China.
| | - Minjuan Yang
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhihao Zeng
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ruiyin Tang
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jieyi Jiang
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine/Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Guangyin Wu
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Canhui Xie
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dezheng Jia
- School of the Fifth Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaoli Bi
- Guangdong Provincial Engineering Technology Research Institute of Traditional Chinese Medicine/Guangdong Provincial Key Laboratory of Research and Development in Traditional Chinese Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
3
|
Zheng W, Xu G, Lue Z, Zhou X, Wang N, Ma Y, Yuan W, Yu L, Zhu D, Zhang X. Nervonic acid protects against oligodendrocytes injury following chronic cerebral hypoperfusion in mice. Eur J Pharmacol 2024; 982:176932. [PMID: 39182543 DOI: 10.1016/j.ejphar.2024.176932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) has been acknowledged as a potential contributor to cognitive dysfunction and brain injury, causing progressive demyelination of white matter, oligodendrocytes apoptosis and microglia activation. Nervonic acid (NA), a naturally occurring fatty acid with various pharmacological effects, has been found to alleviate neurodegeneration. Nonetheless, evidence is still lacking on whether NA can protect against neurological dysfunction resulting from CCH. To induce CCH in mice, we employed the right unilateral common carotid artery occlusion (rUCCAO) method, followed by oral administration of NA daily for 28 days after the onset of hypoperfusion. We found that NA ameliorated cognitive function, as evidenced by improved performance of NA-treated mice in both novel object recognition test and Morris water maze test. Moreover, NA mitigated demyelination and loss of oligodendrocytes in the corpus callosum and hippocampus of rUCCAO-treated mice, and prevented oligodendrocyte apoptosis. Furthermore, NA protected primary cultured murine oligodendrocytes against oxygen-glucose deprivation (OGD)-induced cell death in a concentration-dependent manner. These findings indicated that NA promotes oligodendrocyte maturation both in vivo and in vitro. Our findings suggest that NA offers protective effects against cerebral hypoperfusion, highlighting its potential as a promising treatment for CCH and related neurological disorders.
Collapse
Affiliation(s)
- Wanqing Zheng
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Genghua Xu
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Zhengwei Lue
- Jinhua Institute of Zhejiang University, 321299, Jinhua, China
| | - Xinyu Zhou
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
| | - Ning Wang
- Jinhua Institute of Zhejiang University, 321299, Jinhua, China
| | - Yun Ma
- Jinhua Institute of Zhejiang University, 321299, Jinhua, China
| | - Wenyue Yuan
- Jinhua Institute of Zhejiang University, 321299, Jinhua, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China; Jinhua Institute of Zhejiang University, 321299, Jinhua, China
| | - Danyan Zhu
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China.
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China; Institute of Drug Metabolism and Pharmaceutical Analysis, State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
4
|
Huang Y, Wu Q, Li S, Lin X, Yang S, Zhu R, Fu C, Zhang Z. Harnessing nature's pharmacy: investigating natural compounds as novel therapeutics for ulcerative colitis. Front Pharmacol 2024; 15:1394124. [PMID: 39206263 PMCID: PMC11349575 DOI: 10.3389/fphar.2024.1394124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024] Open
Abstract
Backgrounds Ulcerative colitis (UC) is a form of chronic inflammatory bowel disease, and UC diagnosis rates continue to rise throughout the globe. The research and development of new drugs for the treatment of UC are urgent, and natural compounds are an important source. However, there is a lack of systematic summarization of natural compounds and their mechanisms for the treatment of UC. Methods We reviewed the literature in the databases below from their inception until July 2023: Web of Science, PubMed, China National Knowledge Infrastructure, and Wanfang Data, to obtain information on the relationship between natural compounds and UC. Results The results showed that 279 natural compounds treat UC through four main mechanisms, including regulating gut microbiota and metabolites (Mechanism I), protecting the intestinal mucosal barrier (Mechanism II), regulating intestinal mucosal immune response (Mechanism III), as well as regulating other mechanisms (Mechanism Ⅳ) such as cellular autophagy modulation and ferroptosis inhibition. Of these, Mechanism III is regulated by all natural compounds. The 279 natural compounds, including 62 terpenoids, 57 alkaloids, 52 flavonoids, 26 phenols, 19 phenylpropanoids, 9 steroids, 9 saponins, 8 quinonoids, 6 vitamins, and 31 others, can effectively ameliorate UC. Of these, terpenoids, alkaloids, and flavonoids have the greatest potential for treating UC. It is noteworthy to highlight that a total of 54 natural compounds exhibit their therapeutic effects by modulating Mechanisms I, II, and III. Conclusion This review serves as a comprehensive resource for the pharmaceutical industry, researchers, and clinicians seeking novel therapeutic approaches to combat UC. Harnessing the therapeutic potential of these natural compounds may significantly contribute to the improvement of the quality of life of patients with UC and promotion of disease-modifying therapies in the future.
Collapse
Affiliation(s)
- You Huang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuhong Wu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sha Li
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xia Lin
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shasha Yang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Zhu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomei Fu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Zhang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
Haque M, Kaminsky L, Abdulqadir R, Engers J, Kovtunov E, Rawat M, Al-Sadi R, Ma TY. Lactobacillus acidophilus inhibits the TNF-α-induced increase in intestinal epithelial tight junction permeability via a TLR-2 and PI3K-dependent inhibition of NF-κB activation. Front Immunol 2024; 15:1348010. [PMID: 39081324 PMCID: PMC11286488 DOI: 10.3389/fimmu.2024.1348010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/25/2024] [Indexed: 08/02/2024] Open
Abstract
Background Defective intestinal epithelial tight junction (TJ), characterized by an increase in intestinal TJ permeability, has been shown to play a critical role in the pathogenesis of inflammatory bowel disease (IBD). Tumor necrosis factor-α (TNF-α) is a key pro-inflammatory cytokine involved in the immunopathology of IBD and has been shown to cause an increase in intestinal epithelial TJ permeability. Although TNF-α antibodies and other biologics have been advanced for use in IBD treatment, these therapies are associated with severe side effects and have limited efficacy, and there is an urgent need for therapies with benign profiles and high therapeutic efficacy. Probiotic bacteria have beneficial effects and are generally safe and represent an important class of potential therapeutic agents in IBD. Lactobacillus acidophilus (LA) is one of the most used probiotics for wide-ranging health benefits, including in gastrointestinal, metabolic, and inflammatory disorders. A specific strain of LA, LA1, was recently demonstrated to have protective and therapeutic effects on the intestinal epithelial TJ barrier. However, the mechanisms of actions of LA1 remain largely unknown. Methods The primary aim of this study was to investigate microbial-epithelial interactions and novel signaling pathways that regulate the effect of LA1 on TNF-α-induced increase in intestinal epithelial TJ permeability, using cell culture and animal model systems. Results and Conclusion Pre-treatment of filter-grown Caco-2 monolayers with LA1 prevented the TNF-α-induced increase in intestinal epithelial TJ permeability by inhibiting TNF-α-induced activation of NF-κB p50/p65 and myosin light chain kinase (MLCK) gene and kinase activity in a TLR-2-dependent manner. LA1 produced a TLR-2- and MyD88-dependent activation of NF-κB p50/p65 in immune cells; however, LA1, in intestinal cells, inhibited the NF-κB p50/p65 activation in a TLR-2-dependent but MyD88-independent manner. In addition, LA1 inhibition of NF-κB p50/p65 and MLCK gene was mediated by TLR-2 pathway activation of phosphatidylinositol 3-kinase (PI3K) and IKK-α phosphorylation. Our results demonstrated novel intracellular signaling pathways by which LA1/TLR-2 suppresses the TNF-α pathway activation of NF-κB p50/p65 in intestinal epithelial cells and protects against the TNF-α-induced increase in intestinal epithelial TJ permeability.
Collapse
Affiliation(s)
- Mohammad Haque
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Lauren Kaminsky
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Raz Abdulqadir
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Jessica Engers
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Evgeny Kovtunov
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Manmeet Rawat
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| | - Rana Al-Sadi
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Thomas Y. Ma
- Department of Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA, United States
| |
Collapse
|
6
|
Zhu J, Wu Y, Ge X, Chen X, Mei Q. Discovery and Validation of Ferroptosis-Associated Genes of Ulcerative Colitis. J Inflamm Res 2024; 17:4467-4482. [PMID: 39006497 PMCID: PMC11246036 DOI: 10.2147/jir.s463042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/03/2024] [Indexed: 07/16/2024] Open
Abstract
Background Ulcerative colitis (UC) is a long-lasting idiopathic condition, but its precise mechanisms remain unclear. Meanwhile, evidence has demonstrated that ferroptosis seems to interlock with the progress of UC. This research sought to identify hub genes of UC related to ferroptosis. Methods First, the relevant profiles for this article were obtained from GEO database. From the FerrDb, 479 genes linked to ferroptosis were retrieved. Using analysis of the difference and WGCNA on colonic samples from GSE73661, the remaining six hub genes linked to ferroptosis and UC were discovered. Through logistic regression analyses, the diagnostic model was constructed and was then evaluated by external validation using dataset GSE92415. Afterwards, the correlation between immune cell filtration in UC and hub genes was examined. Finally, a mice model of colitis was established, and the results were verified using qRT-PCR. Results We acquired six hub genes linked to ferroptosis and UC. In order to create a diagnostic model for UC, we used logistic regression analysis to screen three of the six ferroptosis related genes (HIF1A, SLC7A11, and LPIN1). The ROC curve showed that the three hub genes had outstanding potential for disease diagnosis (AUC = 0.976), which was subsequently validated in samples from GSE92415 (AUC = 0.962) and blood samples from GSE3365 (AUC = 0.847) and GSE94648 (AUC = 0.769). These genes might be crucial for UC immunity based upon the results on the immune system. Furthermore, mouse samples examined using qRT-PCR also verified our findings. Conclusion In conclusion, the findings have important implications for ferroptosis and UC, and these hub genes may also offer fresh perspectives on the aetiology and therapeutic approaches of UC.
Collapse
Affiliation(s)
- Jiejie Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Yumei Wu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Xiaoyuan Ge
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Xinwen Chen
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| | - Qiao Mei
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei City, Anhui Province, People's Republic of China
- Key Laboratory of Digestive Diseases of Anhui Province, Hefei, People's Republic of China
| |
Collapse
|
7
|
Giordano MV, Crisi PE, Gramenzi A, Cattaneo D, Corna L, Sung CH, Tolbert KM, Steiner JM, Suchodolski JS, Boari A. Fecal microbiota and concentrations of long-chain fatty acids, sterols, and unconjugated bile acids in cats with chronic enteropathy. Front Vet Sci 2024; 11:1401592. [PMID: 38933703 PMCID: PMC11199873 DOI: 10.3389/fvets.2024.1401592] [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: 03/15/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Feline chronic enteropathies (FCE) are common causes of chronic gastrointestinal signs in cats and include different diseases such as food-responsive enteropathy (FRE), inflammatory bowel diseases (IBD), and low-grade intestinal T-cell lymphoma (LGITL). Although changes in intestinal microbiota and fecal metabolites have been reported in dogs and humans with chronic enteropathy, research in cats has been limited. Therefore, this study aimed to evaluate the fecal microbiota and lipid-related fecal metabolites in cats with FCE to a clinically healthy comparison group (CG). A total of 34 cats with FCE (13 FRE, 15 IBD, and 6 LGITL) and 27 cats in the CG were enrolled in this study. The fecal microbiota was evaluated by the qPCR-based feline Dysbiosis Index (DI). The feline DI in cats with CE (median: 1.3, range: -2.4 to 3.8) was significantly higher (p < 0.0001) compared to CG (median: - 2.3, Range: -4.3 to 2.3), with no difference found among the FCE subgroups. The fecal abundances of Faecalibacterium (p < 0.0001), Bacteroides (p < 0.0001), Fusobacterium (p = 0.0398), Bifidobacterium (p = 0.0004), and total bacteria (p = 0.0337) significantly decreased in cats with FCE. Twenty-seven targeted metabolites were measured by gas chromatography-mass spectrometry, including long-chain fatty acids (LCFAs), sterols, and bile acids (BAs). Fecal concentrations of 5 of 12 LCFAs were significantly increased in cats with FCE compared to CG. Fecal concentrations of zoosterol (p = 0.0109), such as cholesterol (p < 0.001) were also significantly increased in cats with FCE, but those of phytosterols were significantly decreased in this group. No differences in fecal BAs were found between the groups. Although no differences were found between the four groups, the fecal metabolomic pattern of cats with FRE was more similar to that of the CG than to those with IBD or LGITL. This could be explained by the mild changes associated with FRE compared to IBD and LGITL. The study showed changes in intestinal microbiota and alteration of fecal metabolites in FCE cats compared to the CG. Changes in fecal lipids metabolites suggest a dysmetabolism of lipids, including LCFAs, sterols, and unconjugated BAs in cats with CE.
Collapse
Affiliation(s)
| | - Paolo Emidio Crisi
- Department of Veterinary Medicine, University of Teramo, Piano D’Accio, Teramo, Italy
| | - Alessandro Gramenzi
- Department of Veterinary Medicine, University of Teramo, Piano D’Accio, Teramo, Italy
| | | | - Luca Corna
- Endovet Professional Association, Rome, Italy
| | - Chi-Hsuan Sung
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Katherine M. Tolbert
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Joerg M. Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Andrea Boari
- Department of Veterinary Medicine, University of Teramo, Piano D’Accio, Teramo, Italy
| |
Collapse
|
8
|
Lee Y, Yoon B, Son S, Cho E, Kim KB, Choi EY, Kim DE. Inhibition of Immunoproteasome Attenuates NLRP3 Inflammasome Response by Regulating E3 Ubiquitin Ligase TRIM31. Cells 2024; 13:675. [PMID: 38667290 PMCID: PMC11048918 DOI: 10.3390/cells13080675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Excessive secretion of pro-inflammatory cytokines leads to the disruption of intestinal barrier in inflammatory bowel disease (IBD). The inflammatory cytokine tumor necrosis factor alpha (TNFα) induces the assembly of the NLRP3 inflammasome, resulting in the augmented secretion of inflammatory cytokines implicated in the pathogenesis of inflammatory bowel disease (IBD). TNFα has also been known to induce the formation of immunoproteasome (IP), which incorporates immunosubunits LMP2, LMP7, and MECL-1. Inhibition of IP activity using the IP subunit LMP2-specific inhibitor YU102, a peptide epoxyketone, decreased the protein levels of NLRP3 and increased the K48-linked polyubiquitination levels of NLRP3 in TNFα-stimulated intestinal epithelial cells. We observed that inhibition of IP activity caused an increase in the protein level of the ubiquitin E3 ligase, tripartite motif-containing protein 31 (TRIM31). TRIM31 facilitated K48-linked polyubiquitination and proteasomal degradation of NLRP3 with an enhanced interaction between NLRP3 and TRIM31 in intestinal epithelial cells. In addition, IP inhibition using YU102 ameliorated the symptoms of colitis in the model mice inflicted with dextran sodium sulfate (DSS). Administration of YU102 in the DSS-treated colitis model mice caused suppression of the NLRP3 protein levels and accompanied inflammatory cytokine release in the intestinal epithelium. Taken together, we demonstrated that inhibiting IP under inflammatory conditions induces E3 ligase TRIM31-mediated NLRP3 degradation, leading to attenuation of the NLRP3 inflammatory response that triggers disruption of intestinal barrier.
Collapse
Affiliation(s)
- Yubin Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Boran Yoon
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Sumin Son
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Eunbin Cho
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| | - Kyung Bo Kim
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Center for Translational Science at Port St. Lucie, Florida International University, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA;
| | - Eun Young Choi
- Department of Cellular & Molecular Medicine, Herbert Wertheim College of Medicine, Center for Translational Science at Port St. Lucie, Florida International University, 11350 SW Village Pkwy, Port St. Lucie, FL 34987, USA;
| | - Dong-Eun Kim
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; (Y.L.); (B.Y.); (S.S.); (E.C.)
| |
Collapse
|
9
|
Bai J, Wang Y, Li F, Wu Y, Chen J, Li M, Wang X, Lv B. Research advancements and perspectives of inflammatory bowel disease: A comprehensive review. Sci Prog 2024; 107:368504241253709. [PMID: 38778725 PMCID: PMC11113063 DOI: 10.1177/00368504241253709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease with increasing incidence, such as Crohn's disease and ulcerative colitis. The accurate etiology and pathogenesis of IBD remain unclear, and it is generally believed that it is related to genetic susceptibility, gut microbiota, environmental factors, immunological abnormalities, and potentially other factors. Currently, the mainstream therapeutic drugs are amino salicylic acid agents, corticosteroids, immunomodulators, and biological agents, but the remission rates do not surpass 30-60% of patients in a real-life setting. As a consequence, there are many studies focusing on emerging drugs and bioactive ingredients that have higher efficacy and long-term safety for achieving complete deep healing. This article begins with a review of the latest, systematic, and credible summaries of the pathogenesis of IBD. In addition, we provide a summary of the current treatments and drugs for IBD. Finally, we focus on the therapeutic effects of emerging drugs such as microRNAs and lncRNAs, nanoparticles-mediated drugs and natural products on IBD and their mechanisms of action.
Collapse
Affiliation(s)
- Junyi Bai
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Ying Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Fuhao Li
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yueyao Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Meng Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bin Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
10
|
Namiecinska M, Piatek P, Lewkowicz P. Nervonic Acid Synthesis Substrates as Essential Components in Profiled Lipid Supplementation for More Effective Central Nervous System Regeneration. Int J Mol Sci 2024; 25:3792. [PMID: 38612605 PMCID: PMC11011827 DOI: 10.3390/ijms25073792] [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: 02/27/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Central nervous system (CNS) damage leads to severe neurological dysfunction as a result of neuronal cell death and axonal degeneration. As, in the mature CNS, neurons have little ability to regenerate their axons and reconstruct neural loss, demyelination is one of the hallmarks of neurological disorders such as multiple sclerosis (MS). Unfortunately, remyelination, as a regenerative process, is often insufficient to prevent axonal loss and improve neurological deficits after demyelination. Currently, there are still no effective therapeutic tools to restore neurological function, but interestingly, emerging studies prove the beneficial effects of lipid supplementation in a wide variety of pathological processes in the human body. In the future, available lipids with a proven beneficial effect on CNS regeneration could be included in supportive therapy, but this topic still requires further studies. Based on our and others' research, we review the role of exogenous lipids, pointing to substrates that are crucial in the remyelination process but are omitted in available studies, justifying the properly profiled supply of lipids in the human diet as a supportive therapy during CNS regeneration.
Collapse
Affiliation(s)
- Magdalena Namiecinska
- Department of Immunogenetics, Medical University of Lodz, Pomorska 251/A4 Street, 92-213 Lodz, Poland; (P.P.); (P.L.)
| | | | | |
Collapse
|
11
|
Zhang S, Ding C, Liu X, Zhao Y, Li S, Ding Q, Zhao T, Ma S, Li W, Liu W. New resource food-arabinogalactan improves DSS-induced acute colitis through intestinal flora and NLRP3 signaling pathway. Int J Biol Macromol 2024; 258:129118. [PMID: 38163502 DOI: 10.1016/j.ijbiomac.2023.129118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Colitis can significantly impact daily life. This study utilized DSS to induce acute colitis in mice and examined the regulatory effect of arabinogalactan (AG). The findings demonstrated that AG intake effectively alleviated the phenotype of DSS-induced colitis in mice and protected against small intestine damage. Furthermore, AG suppressed the secretion of pro-inflammatory factors TNF-α and IL-1β, while promoting the secretion of anti-inflammatory factor IL-10. It also inhibited the secretion of LPS in serum and MPO in colon tissue. Additionally, AG regulated the NF-κB/MAPK/PPARγ signaling pathway and inhibited the NLRP3 inflammasome signaling pathway, thereby ameliorating DSS-induced colitis inflammation in mice. AG also influenced the metabolism of short-chain fatty acids, particularly butyrate, in the intestinal tract of mice. Moreover, AG modulated and enhanced the composition of intestinal flora in mice with colitis, increasing the diversity of dominant flora and promoting the growth of beneficial bacteria. These results highlight the protective effects of arabinogalactan against colitis and its potential applications in the food industry.
Collapse
Affiliation(s)
- Shuai Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Chuanbo Ding
- College of traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin 132101, China
| | - Xinglong Liu
- College of traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin 132101, China
| | - Yingchun Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Shanshan Li
- Institute of Biological and Pharmaceutical Engineering, Jilin Agricultural Science and Technology University, Jilin 132101, China
| | - Qiteng Ding
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Ting Zhao
- College of traditional Chinese Medicine, Jilin Agricultural Science and Technology University, Jilin 132101, China
| | - Shuang Ma
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, China; College of Life Sciences, Engineering Research Center of the Chinese Ministry of Education for Bioreactor and Pharmaceutical Development, Jilin Agricultural University, Changchun 130118, China.
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543003, China.
| |
Collapse
|
12
|
Liu Y, Jiang B, Li Y, Zhang X, Wang L, Yao Y, Zhu B, Shi H, Chai X, Hu X, Zhang B, Li H. Effect of traditional Chinese medicine in osteosarcoma: Cross-interference of signaling pathways and potential therapeutic targets. Medicine (Baltimore) 2024; 103:e36467. [PMID: 38241548 PMCID: PMC10798715 DOI: 10.1097/md.0000000000036467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/14/2023] [Indexed: 01/21/2024] Open
Abstract
Osteosarcoma (OS) has a high recurrence rate, disability rate, mortality and metastasis, it brings great economic burden and psychological pressure to patients, and then seriously affects the quality of life of patients. At present, the treatment methods of OS mainly include radiotherapy, chemotherapy, surgical therapy and neoadjuvant chemotherapy combined with limb salvage surgery. These treatment methods can relieve the clinical symptoms of patients to a certain extent, and also effectively reduce the disability rate, mortality and recurrence rate of OS patients. However, because metastasis of tumor cells leads to new complications, and OS cells become resistant with prolonged drug intervention, which reduces the sensitivity of OS cells to drugs, these treatments still have some limitations. More and more studies have shown that traditional Chinese medicine (TCM) has the characteristics of "multiple targets and multiple pathways," and can play an important role in the development of OS through several key signaling pathways, including PI3K/AKT, Wnt/β-catenin, tyrosine kinase/transcription factor 3 (JAK/STAT3), Notch, transforming growth factor-β (TGF-β)/Smad, nuclear transcription factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), nuclear factor E2-related factor 2 (Nrf2), Hippo/YAP, OPG/RANK/RANKL, Hedgehog and so on. In this paper, the signaling pathways of cross-interference between active ingredients of TCM and OS were reviewed, and the development status of novel OS treatment was analyzed. The active ingredients in TCM can provide therapeutic benefits to patients by targeting the activity of signaling pathways. In addition, potential strategies for targeted therapy of OS by using ferroptosis were discussed. We hope to provide a unique insight for the in-depth research and clinical application of TCM in the fields of OS growth, metastasis and chemotherapy resistance by understanding the signaling crosstalk between active ingredients in TCM and OS.
Collapse
Affiliation(s)
- Yuezhen Liu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yanqiang Li
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaoshou Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Lijun Wang
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yasai Yao
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Baohong Zhu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Hengwei Shi
- The Second Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiping Chai
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xingrong Hu
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bangneng Zhang
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Hongzhuan Li
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
13
|
Sung CH, Pilla R, Marsilio S, Chow B, Zornow KA, Slovak JE, Lidbury JA, Steiner JM, Hill SL, Suchodolski JS. Fecal Concentrations of Long-Chain Fatty Acids, Sterols, and Unconjugated Bile Acids in Cats with Chronic Enteropathy. Animals (Basel) 2023; 13:2753. [PMID: 37685017 PMCID: PMC10486672 DOI: 10.3390/ani13172753] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/17/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Chronic enteropathy (CE) in cats encompasses food-responsive enteropathy, chronic inflammatory enteropathy (or inflammatory bowel disease), and low-grade intestinal T-cell lymphoma. While alterations in the gut metabolome have been extensively studied in humans and dogs with gastrointestinal disorders, little is known about the specific metabolic profile of cats with CE. As lipids take part in energy storage, inflammation, and cellular structure, investigating the lipid profile in cats with CE is crucial. This study aimed to measure fecal concentrations of various fatty acids, sterols, and bile acids. Fecal samples from 56 cats with CE and 77 healthy control cats were analyzed using gas chromatography-mass spectrometry, targeting 12 fatty acids, 10 sterols, and 5 unconjugated bile acids. Fecal concentrations of nine targeted fatty acids and animal-derived sterols were significantly increased in cats with CE. However, fecal concentrations of plant-derived sterols were significantly decreased in cats with CE. Additionally, an increased percentage of primary bile acids was observed in a subset of cats with CE. These findings suggest the presence of lipid maldigestion, malabsorption, and inflammation in the gastrointestinal tract of cats with CE. Understanding the lipid alterations in cats with CE can provide insights into the disease mechanisms and potential future therapeutic strategies.
Collapse
Affiliation(s)
- Chi-Hsuan Sung
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (C.-H.S.)
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (C.-H.S.)
| | - Sina Marsilio
- UC Davis School of Veterinary Medicine, Department of Veterinary Medicine and Epidemiology, University of California, Davis, CA 95616, USA
| | - Betty Chow
- Veterinary Specialty Hospital, San Diego, CA 92121, USA
- VCA Animal Specialty and Emergency Center, Los Angeles, CA 90025, USA
| | | | | | - Jonathan A. Lidbury
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (C.-H.S.)
| | - Joerg M. Steiner
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (C.-H.S.)
| | - Steve L. Hill
- Veterinary Specialty Hospital, San Diego, CA 92121, USA
- Flagstaff Veterinary Internal Medicine Consulting, Flagstaff, AZ 86004, USA
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (C.-H.S.)
| |
Collapse
|
14
|
Wang X, Liang T, Mao Y, Li Z, Li X, Zhu X, Cao F, Zhang J. Nervonic acid improves liver inflammation in a mouse model of Parkinson's disease by inhibiting proinflammatory signaling pathways and regulating metabolic pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154911. [PMID: 37276724 DOI: 10.1016/j.phymed.2023.154911] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/25/2023] [Accepted: 05/29/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND Nervonic acid (NA) - a type of bioactive fatty acid that is found in natural sources - can inhibit inflammatory reactions and regulate immune system balance. Therefore, the use of NA for the treatment of neurodegenerative diseases has received considerable attention. Our previous study found that NA inhibited inflammatory responses in the brain of Parkinson's disease (PD) mouse models. In addition to the brain, PD is also associated with visceral organ dysfunction, especially impaired liver function. Thus, studying the role of NA in PD-mediated inflammation of the liver is particularly important. METHODS A combined transcriptome and metabolomic approach was utilized to investigate the anti-inflammatory effects of NA on the liver of PD mice. Inflammatory signaling molecules and metabolic pathway-related genes were examined in the liver using real-time PCR and western blotting. RESULTS Liver transcriptome analysis revealed that NA exerted anti-inflammatory effects by controlling several pro-inflammatory signaling pathways, such as the down-regulation of the tumor necrosis factor and nuclear factor kappa B signaling pathways, both of which were essential in the development of inflammatory disease. In addition, liver metabolomic results revealed that metabolites related to steroid hormone biosynthesis, arachidonic acid metabolism, and linoleic acid metabolism were up-regulated and those related to valine, leucine, and isoleucine degradation pathways were down-regulated in NA treatment groups compared with the PD model. The integration of metabolomic and transcriptomic results showed NA significantly exerted its anti-inflammatory function by regulating the transcription and metabolic pathways of multiple genes. Particularly, linoleic acid metabolism, arachidonic acid metabolism, and steroid hormone biosynthesis were the crucial pathways of the anti-inflammatory action of NA. Key genes in these metabolic pathways and key molecules in inflammatory signaling pathways were also verified, which were consistent with transcriptomic results. CONCLUSION These findings provide novel insights into the liver protective effects of NA against PD mice. This study also showed that NA could be a useful dietary element for improving and treating PD-induced liver inflammation.
Collapse
Affiliation(s)
- Xueqi Wang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Tingyu Liang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Ying Mao
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Zhengdou Li
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Xu Li
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Xinliang Zhu
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China; Bioactive Products Engineering Research Center for Gansu Distinctive Plants, Lanzhou 730070, China; Institute of Rural Development and Research, Northwest Normal University, Lanzhou, Gansu Province 730070, China
| | - Fuliang Cao
- Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China.
| | - Ji Zhang
- College of Life Science, Northwest Normal University, Lanzhou, Gansu Province 730070, China; Bioactive Products Engineering Research Center for Gansu Distinctive Plants, Lanzhou 730070, China; Institute of Rural Development and Research, Northwest Normal University, Lanzhou, Gansu Province 730070, China.
| |
Collapse
|