1
|
Yang X, Hong C, Guan T, Zhang C, Xiao P, Yang Y, Xiao H, He Z. Investigation of the effects of Periplaneta americana (L.) extract on ischemic stroke based on combined multi-omics of gut microbiota. Front Pharmacol 2024; 15:1429960. [PMID: 39679371 PMCID: PMC11638836 DOI: 10.3389/fphar.2024.1429960] [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: 05/09/2024] [Accepted: 11/13/2024] [Indexed: 12/17/2024] Open
Abstract
Ischemic stroke (IS) is a highly lethal type of cardiovascular and cerebrovascular disease. Improving survival rates and promoting recovery in patients with IS pose significant challenges, however, recent research has identified the gut-brain axis as a therapeutic target. In this study, we evaluated the regulatory effect of Periplaneta americana (L.) extract (PAS840), which has established anti-inflammatory, antioxidant, and neuroprotective effects, on the gut microbiota using a rat model of temporary middle cerebral artery occlusion (tMCAO). We evaluated the protective effects of PAS840 on brain damage in IS rats through TTC (triphenyltetrazolium chloride), Nissl staining, and pathological section analysis. Additionally, we investigated the impact of PAS840 on the gut microbiota and metabolites using 16S rRNA sequencing, untargeted metabolomics of gut contents, and transcriptomics analyses of brain tissues to explore its mechanism of action. PAS840 intervention resulted in significant changes in the gut microbiota, including an increase in the abundance of probiotic flora, decrease in the abundance of harmful flora, and significant changes in metabolite profiles. It also attenuated brain damage, decreased platelet activity, inhibited oxidative stress and genes related to inflammation, and improved neurological function in rats. These findings suggest that PAS840 has preventive and therapeutic effects against IS via the gut-brain axis by regulating the gut microbiota and related metabolites. Accordingly, PAS840 is a candidate therapeutic drug for further research.
Collapse
Affiliation(s)
- Xin Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Canhui Hong
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Tangfei Guan
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Sichuan, Chengdu, China
| | - ChengGui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Peiyun Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Yongshou Yang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Huai Xiao
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| | - Zhengchun He
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, College of Pharmacy, Dali University, Dali, Yunnan, China
- National-Local Joint Engineering Research Center of Entomoceutics, Dali, Yunnan, China
| |
Collapse
|
2
|
Wang Z, Ye R, Xu Z, Zhang S, Liu C, Zhu K, Wang P, Huang J. Protective Effect of IgY Embedded in W/O/W Emulsion on LPS Enteritis-Induced Colonic Injury in Mice. Nutrients 2024; 16:3361. [PMID: 39408328 PMCID: PMC11479051 DOI: 10.3390/nu16193361] [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: 08/30/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Chicken yolk immunoglobulin (IgY), an immunologically active component, is used as an alternative to antibiotics for the treatment of enteritis. In this study, IgY was embedded in a W/O/W emulsion to overcome the digestive barrier and to investigate the protective effect of IgY against LPS-induced enteritis in mice. Four different hydrophilic emulsifiers (T80, PC, SC, and WPI) were selected to prepare separate W/O/W emulsions for encapsulating IgY. The results showed that the IgY-embedded double emulsion in the WPI group was the most effective. IgY embedded in the W/O/W emulsion could reduce the damage of LPS to the mouse intestine and prevent LPS-induced intestinal mucosal damage in mice. It increased the number of cup cells, promoted the expression of Muc2, and increased the mRNA expression levels of KLF3, TFF3, Itln1, and Ang4 (p < 0.05). It also enhanced the antioxidant capacity of the colon tissue, reduced the level of inflammatory factors in the colon tissue, and protected the integrity of the colon tissue. Stable embedding of IgY could be achieved using the W/O/W emulsion. In addition, the IgY-embedded W/O/W emulsion can be used as a dietary supplement to protect against LPS-induced enteritis in mice.
Collapse
Affiliation(s)
- Zhaohui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100083, China; (Z.W.); (Z.X.); (S.Z.); (C.L.); (K.Z.)
| | - Ruihua Ye
- College of Veterinary Medicine, China Agricultural University, Beijing 100083, China;
| | - Zijian Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100083, China; (Z.W.); (Z.X.); (S.Z.); (C.L.); (K.Z.)
| | - Shidi Zhang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100083, China; (Z.W.); (Z.X.); (S.Z.); (C.L.); (K.Z.)
| | - Chuanming Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100083, China; (Z.W.); (Z.X.); (S.Z.); (C.L.); (K.Z.)
| | - Kongdi Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100083, China; (Z.W.); (Z.X.); (S.Z.); (C.L.); (K.Z.)
| | - Pengjie Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100083, China
| | - Jiaqiang Huang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, Ministry of Education, China Agricultural University, Beijing 100083, China; (Z.W.); (Z.X.); (S.Z.); (C.L.); (K.Z.)
| |
Collapse
|
3
|
Wu L, Hu Z, Luo X, Ge C, Lv Y, Zhan S, Huang W, Shen X, Yu D, Liu B. Itaconic Acid Alleviates Perfluorooctanoic Acid-Induced Oxidative Stress and Intestinal Damage by Regulating the Keap1/Nrf2/Ho-1 Pathway and Reshaping the Gut Microbiota. Int J Mol Sci 2024; 25:9826. [PMID: 39337313 PMCID: PMC11432532 DOI: 10.3390/ijms25189826] [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: 07/22/2024] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/30/2024] Open
Abstract
Itaconic acid (IA) is recognized for its potential application in treating intestinal diseases owing to the anti-inflammatory and antioxidant properties. Perfluorooctanoic acid (PFOA) can accumulate in animals and result in oxidative and inflammatory damages to multi-tissue and organ, particularly in the intestinal tract. This study aimed to explore whether IA could mitigate intestinal damage induced by PFOA exposure in laying hens and elucidate its potential underlying mechanisms. The results showed that IA improved the antioxidant capacity of laying hens and alleviated the oxidative damage induced by PFOA, as evidenced by the elevated activities of T-SOD, GSH-Px, and CAT, and the decreased MDA content in both the jejunum and serum. Furthermore, IA improved the intestinal morphological and structural integrity, notably attenuating PFOA-induced villus shedding, length reduction, and microvillus thinning. IA also upregulated the mRNA expression of ZO-1, Occludin, Claudin-1, and Mucin-2 in the jejunum, thereby restoring intestinal barrier function. Compared with the PF group, IA supplementation downregulated the gene expression of Keap1 and upregulated the HO-1, NQO1, SOD1, and GPX1 expression in the jejunum. Meanwhile, the PF + IA group exhibited lower expressions of inflammation-related genes (NF-κB, IL-1β, IFN-γ, TNF-α, and IL-6) compared to the PF group. Moreover, IA reversed the PFOA-induced imbalance in gut microbiota by reducing the harmful bacteria such as Escherichia-Shigella, Clostridium innocuum, and Ruminococcus torques, while increasing the abundance of beneficial bacteria like Lactobacillus. Correlation analysis further revealed a significant association between gut microbes, inflammatory factors, and the Keap1/Nrf2/HO-1 pathway expression. In conclusion, dietary IA supplementation could alleviate the oxidative and inflammatory damage caused by PFOA exposure in the intestinal tract by reshaping the intestinal microbiota, modulating the Keap1/Nrf2/HO-1 pathway and reducing oxidative stress and inflammatory response, thereby promoting intestinal homeostasis.
Collapse
Affiliation(s)
- Lianchi Wu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhaoying Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Luo
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chaoyue Ge
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yujie Lv
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Shenao Zhan
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weichen Huang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Shen
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Dongyou Yu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Shaoxing 312500, China
| | - Bing Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
- ZJU-Xinchang Joint Innovation Centre (TianMu Laboratory), Gaochuang Hi-Tech Park, Shaoxing 312500, China
| |
Collapse
|
4
|
Zhao H, Huang Y, Yang W, Huang C, Ou Z, He J, Yang M, Wu J, Yao H, Yang Y, Yi J, Kong L. Viola yedoensis Makino alleviates lipopolysaccharide-induced intestinal oxidative stress and inflammatory response by regulating the gut microbiota and NF-κB-NLRP3/ Nrf2-MAPK signaling pathway in broiler. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 282:116692. [PMID: 38971097 DOI: 10.1016/j.ecoenv.2024.116692] [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: 01/24/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Viola yedoensis Makino (Vy) is a well-known traditional Chinese medicine widely used to treat inflammatory diseases. However, the regulatory effects of dietary Vy supplementation on lipopolysaccharide (LPS)-induced intestinal damage in broilers and the underlying molecular mechanisms remain unclear. In this study, broilers were intraperitoneally injected with 1 mg/kg LPS on days 17, 19 and 21 to induce intestinal damage. Vy supplementation at 0.5, 1.5 and 4.5 % in the diet was administered separately for 21 days to investigate the potential protective effects of Vy supplementation against LPS-induced intestinal impairment in broilers. Vy supplementation improved intestinal morphology and restored growth performance. Vy supplementation attenuated intestinal inflammation by regulating the nuclear factor kappa B (NF-κB) / NLR family pyrin domain-containing 3 (NLRP3) signaling pathway and inhibited its downstream pro-inflammatory factor levels. In addition, Vy supplementation relieved intestinal oxidative impairment by regulating the nuclear factor erythroid-2 related factor 2 (Nrf2) / mitogen-activated protein kinase (MAPK) signaling pathway and downstream antioxidant enzyme activity. Vy supplementation reduced LPS-induced mitochondrial damage and apoptosis. Furthermore, Vy supplementation alleviated LPS-induced intestinal inflammation and oxidative damage in chickens by increasing the abundance of protective bacteria (Lactobacillus and Romboutsia) and reducing the number of pathogenic bacteria (unclassified_f_Ruminococcaceae, unclassified_f_Oscillospiraceae and norank_f_norank_o_Clostridia_vadinBB60_group). Overall, Vy supplementation effectively ameliorated LPS-induced intestinal damage by regulating the NF-κB-NLRP3/Nrf2-MAPK signaling pathway and maintaining intestinal microbiota balance. Vy supplementation can be used as a dietary supplement to protect broilers against intestinal inflammation and oxidative damage.
Collapse
Affiliation(s)
- Haoqiang Zhao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - You Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Wenjiang Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Chunlin Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zhaoping Ou
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jiayu He
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Mingqi Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jiao Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Huan Yao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yu Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
5
|
Shen X, Zhang X, Li K, Huang G, Li X, Hou Y, Ge X. Combined bacterial translocation and cholestasis aggravates liver injury by activation pyroptosis in obstructive jaundice. Heliyon 2024; 10:e35793. [PMID: 39220957 PMCID: PMC11363856 DOI: 10.1016/j.heliyon.2024.e35793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
This study explores the mechanism by which obstructive jaundice (OJ) induces liver damage through pyroptosis. We induced OJ in rats via bile duct ligation and assessed liver damage using serum biochemical markers and histological analysis of liver tissue. Pyroptosis was investigated through immunofluorescence, ELISA, Western blot, and quantitative RT-PCR techniques. Additionally, we examined intestinal function and fecal microbiota alterations in the rats using 16S rDNA sequencing. In vitro experiments involved co-culturing Kupffer cells and hepatocytes, which were then exposed to bile and lipopolysaccharide (LPS). Our findings indicated that OJ modified the gut microbiota, increasing LPS levels, which, in conjunction with bile, initiated a cycle of inflammation, fibrosis, and cell death in the liver. Mechanistically, OJ elevated necrotic markers such as ATP, which in turn activated pyroptotic pathways. Increased levels of pyroptosis-related molecules, including NLRP3, caspase-1, gasdermin D, and IL-18, were confirmed. In our co-cultured cell model, bile exposure resulted in cell death and ATP release, leading to the activation of the NLRP3 inflammasome and its downstream effectors, caspase-1 and IL-18. The combination of bile and LPS significantly intensified pyroptotic responses. This study is the first to demonstrate that LPS and bile synergistically exacerbate liver injury by promoting necrosis and pyroptosis, unveiling a novel mechanism of OJ-associated hepatic damage and suggesting avenues for potential preventive or therapeutic interventions.
Collapse
Affiliation(s)
- Xin Shen
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Zhang
- Luoyang Orthopedic-Traumatological Hospital of Henan Province (Henan Provincial Orthopedic Hospital), Luoyang, 471002, Henan, China
| | - Kaiyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Guangming Huang
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Xinyu Li
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| | - Yunlong Hou
- National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Shijiazhuang, 050000, Hebei, China
| | - Xin Ge
- Department of General Surgery, Heilongjiang Provincial Hospital, 82 Zhongshan Road, Harbin, 150036, Heilongjiang, China
| |
Collapse
|
6
|
Qu J, Pei H, Li XZ, Li Y, Chen JM, Zhang M, Lu ZQ. Erythrocyte membrane biomimetic EGCG nanoparticles attenuate renal injury induced by diquat through the NF-κB/NLRP3 inflammasome pathway. Front Pharmacol 2024; 15:1414918. [PMID: 39045044 PMCID: PMC11263105 DOI: 10.3389/fphar.2024.1414918] [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: 04/09/2024] [Accepted: 06/14/2024] [Indexed: 07/25/2024] Open
Abstract
Diquat (DQ) poisoning can cause multiple organ damage, and the kidney is considered to be the main target organ. Increasing evidence shows that alleviating oxidative stress and inflammatory response has promising application prospects. Epigallocatechin gallate (EGCG) has potent antioxidant and anti-inflammatory effects. In this study, red blood cell membrane (RBCm)-camouflaged polylactic-co-glycolic acid (PLGA) nanoparticles (NPs) were synthesized to deliver EGCG (EGCG-RBCm/NPs) for renal injury induced by DQ. Human renal tubular epithelial cells (HK-2 cells) were stimulated with 600 μM DQ for 12 h and mice were intraperitoneally injected with 50 mg/kg b.w. DQ, followed by 20 mg/kg b.w./day EGCG or EGCG-RBCM/NPs for 3 days. The assessment of cellular vitality was carried out using the CCK-8 assay, while the quantification of reactive oxygen species (ROS) was performed through ROS specific probes. Apoptosis analysis was conducted by both flow cytometry and TUNEL staining methods. Pathological changes in renal tissue were observed. The expressions of NLRP3, IL-1β, IL-18, NFκB and Caspase1 were detected by quantitative reverse transcription polymerase chain reaction (qRT-PCR), immunohistochemistry, immunofluorescence, and Western blot. The results showed that the DQ group had increased ROS expression, increased the level of oxidative stress, and increased apoptosis rate compared with the control group. Histopathological analysis of mice in the DQ group showed renal tubular injury and elevated levels of blood urea nitrogen (BUN), serum creatinine (SCr), kidney injury molecule-1 (KIM-1), and cystatin C (Cys C). Furthermore, the DQ group exhibited heightened expression of NLRP3, p-NFκB p65, Caspase1 p20, IL-1β, and IL-18. However, EGCG-RBCm/NPs treatment mitigated DQ-induced increases in ROS, apoptosis, and oxidative stress, as well as renal toxicity and decreases in renal biomarker levels. Meanwhile, the expression of the above proteins were significantly decreased, and the survival rate of mice was ultimately improved, with an effect better than that of the EGCG treatment group. In conclusion, EGCG-RBCm/NPs can improve oxidative stress, inflammation, and apoptosis induced by DQ. This effect is related to the NF-κB/NLRP3 inflammasome pathway. Overall, this study provides a new approach for treating renal injury induced by DQ.
Collapse
Affiliation(s)
- Jie Qu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Hui Pei
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Xin-Ze Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Yan Li
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Jian-Ming Chen
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Min Zhang
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| | - Zhong-Qiu Lu
- Emergency Department, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Wenzhou Key Laboratory of Emergency and Disaster Medicine, Wenzhou, China
| |
Collapse
|
7
|
Wen X, Wan F, Zhong R, Chen L, Zhang H. Hydroxytyrosol Alleviates Intestinal Oxidative Stress by Regulating Bile Acid Metabolism in a Piglet Model. Int J Mol Sci 2024; 25:5590. [PMID: 38891778 PMCID: PMC11171822 DOI: 10.3390/ijms25115590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/16/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
Infants and young animals often suffer from intestinal damage caused by oxidative stress, which may adversely affect their overall health. Hydroxytyrosol, a plant polyphenol, has shown potential in decreasing intestinal oxidative stress, but its application and mechanism of action in infants and young animals are still inadequately documented. This study selected piglets as a model to investigate the alleviating effects of hydroxytyrosol on intestinal oxidative stress induced by diquat and its potential mechanism. Hydroxytyrosol improved intestinal morphology, characterized by higher villus height and villus height/crypt depth. Meanwhile, hydroxytyrosol led to higher expression of Occludin, MUC2, Nrf2, and its downstream genes, and lower expression of cytokines IL-1β, IL-6, and TNF-α. Both oxidative stress and hydroxytyrosol resulted in a higher abundance of Clostridium_sensu_stricto_1, and a lower abundance of Lactobacillus and Streptococcus, without a significant effect on short-chain fatty acids levels. Oxidative stress also led to disorders in bile acid (BA) metabolism, such as the lower levels of primary BAs, hyocholic acid, hyodeoxycholic acid, and tauroursodeoxycholic acid, which were partially restored by hydroxytyrosol. Correlation analysis revealed a positive correlation between these BA levels and the expression of Nrf2 and its downstream genes. Collectively, hydroxytyrosol may reduce oxidative stress-induced intestinal damage by regulating BA metabolism.
Collapse
Affiliation(s)
| | | | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (X.W.); (F.W.); (H.Z.)
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (X.W.); (F.W.); (H.Z.)
| | | |
Collapse
|
8
|
Song W, Wen R, Liu T, Zhou L, Wang G, Dai X, Shi L. Oat-based postbiotics ameliorate high-sucrose induced liver injury and colitis susceptibility by modulating fatty acids metabolism and gut microbiota. J Nutr Biochem 2024; 125:109553. [PMID: 38147914 DOI: 10.1016/j.jnutbio.2023.109553] [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: 10/12/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 12/28/2023]
Abstract
High-sucrose (HS) consumption leads to metabolic disorders and increases susceptibility to colitis. Postbiotics hold great potentials in combating metabolic diseases and offer advantages in safety and processability, compared with living probiotics. We developed innovative oat-based postbiotics and extensively explored how they could benefit in rats with long-term high-sucrose consumption. The postbiotics fermented with Lactiplantibacillus plantarum (OF-1) and OF-5, the one fermented with the optimal selection of five probiotics (i.e., L. plantarum, Limosilactobacillus reuteri, Lacticaseibacillus rhamnosus, Lactobacillus acidophilus, and Bifidobacterium lactis) alleviated HS induced liver injury, impaired fatty acid metabolism and inflammation through activating AMPK/SREBP-1c pathways. Moreover, oat-based postbiotics restored detrimental effects of HS on fatty acid profiles in liver, as evidenced by the increases in polyunsaturated fatty acids and decreases in saturated fatty acids, with OF-5 showing most pronounced effects. Furthermore, oat-based postbiotics prevented HS exacerbated susceptibility to dextran sodium sulfate caused colitis and reconstructed epithelial tight junction proteins in colons. Oat-based postbiotics, in particular OF-5 notably remodeled gut microbiota composition, e.g., enriching the relative abundances of Akkermansia, Bifidobacterium, Alloprevotella and Prevotella, which may play an important role in the liver-colon axis responsible for improvements of liver functions and reduction of colitis susceptibility. The heat-inactivated probiotics protected against HS-induced liver and colon damage, but such effects were less pronounced compared with oat-based postbiotics. Our findings emphasize the great value of oat-based postbiotics as nutritional therapeutics to combat unhealthy diet induced metabolic dysfunctions.
Collapse
Affiliation(s)
- Wei Song
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Ruixue Wen
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Lanqi Zhou
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi, China
| | - Guoze Wang
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, China.
| | - Xiaoshuang Dai
- BGI Institute of Applied Agriculture, BGI-Agro, Shenzhen, Guangdong, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, Shaanxi, China.
| |
Collapse
|
9
|
Zhang S, Lv H, Cai X, Tang S, Zhong R, Chen L, Zhang H. Effects of the compound extracts of Caprifoliaceae and Scutellaria baicalensis Georgi on the intestinal microbiota and antioxidant function. Front Microbiol 2024; 14:1289490. [PMID: 38282732 PMCID: PMC10822692 DOI: 10.3389/fmicb.2023.1289490] [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: 09/06/2023] [Accepted: 12/22/2023] [Indexed: 01/30/2024] Open
Abstract
According to the Chinese encyclopedia "Ben Cao Gang Mu" (AD 1552-1578), Caprifoliaceae and Scutellaria baicalensis Georgi are used in traditional Chinese medicine to clear heat, detoxify, and treat wind-heat colds, upper respiratory tract infections, and pneumonia. However, the mechanism and the effects of the compound extracts of Caprifoliaceae and Scutellaria baicalensis Georgi on intestinal health remain unclear. From the perspective of intestinal microbes, this study assessed the antioxidant, anti-inflammatory, and intestinal protective properties of Caprifoliaceae and Scutellaria baicalensis Georgi. Mice received diets with or without Caprifoliaceae and Scutellaria baicalensis Georgi extractive (BCA) for 2 weeks in this study. The results showed that BCA increased body weight gain, feed intake, and catalase (CAT) content in the mice but reduced γ-glutamyl transpeptidase (γ-GT) content in the serum (p < 0.05). BCA improved the Sobs, Chao, and Ace indices, as well as the number of Campylobacterota, Patercibacteria, and Desulfobacterota in the colon microbiota, while it decreased the Firmicutes phylum (p < 0.05). At the genus level, BCA increased Candidatus_Saccharimonas, Helicobacter, unclassified_f_Lachnospiraceae, Alistipes, norank_f_norank_o_Clostridia_vadinBB60_group, norank_f_Ruminococcaceae, unclassified_f_Ruminococcaceae, etc. abundance (p < 0.05), but it significantly decreased Lactobacillus and Lachnospiraceae_UCG_001 abundance (p < 0.05). Moreover, BCA improved the concentration of acetic acid, butyric acid, propionic acid, valeric acid, and isovaleric acid and diminished the concentration of isobutyric acid (p < 0.05). Correlation analysis shows that the changes in short-chain fatty acids and antioxidant and inflammatory indices in the serum were significantly correlated with the BCA-enriched microbiota. This study supplemented a database for the application of Caprifoliaceae and Scutellaria baicalensis Georgi in clinical and animal production.
Collapse
Affiliation(s)
- Shunfen Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Huiyuan Lv
- College of Animal Science and Technology, China Agricultural University, Beijing, China
- Beijing Centre Biology Co., Ltd., Beijing, China
| | - Xueying Cai
- Hangzhou First People's Hospital, Hangzhou, China
| | - Shanlong Tang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liang Chen
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
10
|
Shen J, Wang S, Xia H, Han S, Wang Q, Wu Z, Zhuge A, Li S, Chen H, Lv L, Chen Y, Li L. Akkermansia muciniphila attenuated lipopolysaccharide-induced acute lung injury by modulating the gut microbiota and SCFAs in mice. Food Funct 2023; 14:10401-10417. [PMID: 37955584 DOI: 10.1039/d3fo04051h] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Gut microbiota are closely related to lipopolysaccharide (LPS)-induced acute lung injury (ALI). Akkermansia muciniphila (A. muciniphila) maintains the intestinal barrier function and regulates the balance of reduced glutathione/oxidized glutathione. However, it may be useful as a treatment strategy for LPS-induced lung injury. Our study aimed to explore whether A. muciniphila could improve lung injury by affecting the gut microbiota. The administration of A. muciniphila effectively attenuated lung injury tissue damage and significantly decreased the oxidative stress and inflammatory reaction induced by LPS, with lower levels of myeloperoxidase (MDA), enhanced superoxide dismutase (SOD) activity, decreased pro-inflammatory cytokine levels, and reduced macrophage and neutrophil infiltration. Moreover, A. muciniphila maintained the intestinal barrier function, reshaped the disordered microbial community, and promoted the secretion of short-chain fatty acids (SCFAs). A. muciniphila significantly downregulated the expression of TLR2, MyD88 and NF-kappa B (P < 0.05). Butyrate supplementation demonstrated a significant improvement in the inflammatory response (P < 0.05) and mitigation of histopathological damage in mice with ALI, thereby restoring the intestinal butyric acid concentration. In conclusion, our findings indicate that A. muciniphila inhibits the accumulation of inflammatory cytokines and attenuates the activation of the TLR2/Myd88/NF-κB pathway due to exerting anti-inflammatory effects through butyrate. This study provides an experimental foundation for the potential application of A. muciniphila and butyrate in the prevention and treatment of ALI.
Collapse
Affiliation(s)
- Jian Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Shuting Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang 330006, China
| | - He Xia
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Qiangqiang Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Zhengjie Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Aoxiang Zhuge
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Shengjie Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Hui Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Longxian Lv
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Yanfei Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Rd., Hangzhou City 310003, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| |
Collapse
|
11
|
He Y, Wang D, Liu K, Deng S, Liu Y. Sodium humate alleviates LPS-induced intestinal barrier injury by improving intestinal immune function and regulating gut microbiota. Mol Immunol 2023; 161:61-73. [PMID: 37499314 DOI: 10.1016/j.molimm.2023.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 07/15/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Sodium humate (HNa), known for its abundant functional active groups, is extensively utilized in food dietary supplements due to its versatile properties. Furthermore, HNa possesses notable anti-inflammatory, antioxidant, and anti-diarrheal properties. This research endeavor aimed to elucidate the protective effects of HNa against intestinal barrier injury induced by lipopolysaccharide (LPS). The findings of this study demonstrated that pretreatment with HNa effectively mitigated intestinal barrier injury in the jejunum. HNa exhibited inhibitory effects on the activation of the NLRP3 inflammasome and the production of inflammatory factors within the intestine. HNa supplementation also contributed to the upregulation of mucin and tight junctions (TJs) expression, consequently enhancing the integrity of the intestinal barrier. Notably, our investigation revealed that HNa shared comparable efficacy with the TLR4 inhibitor TAK-242 in inhibiting the TLR4/NFκB signaling pathway. Furthermore, an in-depth analysis of the gut microbiota demonstrated that HNa exerted a regulatory influence on LPS-induced microflora disturbance. In conclusion, these findings collectively indicate that HNa mitigates LPS-induced mucosal damage in the jejunum and preserves the integrity of the intestinal barrier by modulating intestinal immune function and regulating gut microbiota.
Collapse
Affiliation(s)
- Yanjun He
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Dong Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; College of Veterinary Medicine, Shandong Agricultural University, Tai'an 271000, PR China
| | - Kexin Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shouxiang Deng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Yun Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
12
|
Treatment of Dyslipidemia through Targeted Therapy of Gut Microbiota. Nutrients 2023; 15:nu15010228. [PMID: 36615885 PMCID: PMC9823358 DOI: 10.3390/nu15010228] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Dyslipidemia is a multifaceted condition with various genetic and environmental factors contributing to its pathogenesis. Further, this condition represents an important risk factor for its related sequalae including cardiovascular diseases (CVD) such as coronary artery disease (CAD) and stroke. Emerging evidence has shown that gut microbiota and their metabolites can worsen or protect against the development of dyslipidemia. Although there are currently numerous treatment modalities available including lifestyle modification and pharmacologic interventions, there has been promising research on dyslipidemia that involves the benefits of modulating gut microbiota in treating alterations in lipid metabolism. In this review, we examine the relationship between gut microbiota and dyslipidemia, the impact of gut microbiota metabolites on the development of dyslipidemia, and the current research on dietary interventions, prebiotics, probiotics, synbiotics and microbiota transplant as therapeutic modalities in prevention of cardiovascular disease. Overall, understanding the mechanisms by which gut microbiota and their metabolites affect dyslipidemia progression will help develop more precise therapeutic targets to optimize lipid metabolism.
Collapse
|