1
|
Liu T, Ma M, Wu Y, Asif IM, Chen D, Liu L, Zhang M, Chen Y, Li B, Wang L. Protective Effects of Fucoidan on Iodoacetamide-Induced Functional Dyspepsia via Modulation of 5-HT Metabolism and Microbiota. Int J Mol Sci 2025; 26:3273. [PMID: 40244137 PMCID: PMC11989908 DOI: 10.3390/ijms26073273] [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: 02/06/2025] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
As the major polysaccharide in brown algae, fucoidan possesses broad biological abilities and has been reported to improve gastrointestinal health. Functional dyspepsia, a common non-organic disease, is a complex of symptoms mainly characterized by pathogenesis, such as visceral hypersensitivity, gastric dysmotility, and inflammation. To date, the effects of fucoidan in regulating functional dyspepsia with visceral sensitivity remains unclear. In the current study, iodoacetamide was employed to establish a mouse model of visceral hypersensitivity. Meanwhile, fucoidan was orally administrated for fourteen days. Indicators were conducted to evaluate the potential of fucoidan as the ingredient of complementary and alternative medicine for functional dyspepsia, such as levels of serum hormones, expression of receptors, and gut microbial profile. The results show that oral administration of fucoidan led to significant reductions in the secretion of 5-hydroxytryptamine, cortisol, and corticosterone. Additionally, it decreased the expression of 5-hydroxytryptamine-3 receptors, with regulation of 5-hydroxytryptamine metabolism and improvement of gut microbial imbalance. The above results suggest fucoidan could ameliorate visceral hypersensitivity by modulating 5-HT metabolism and microbiota. The current findings indicate that fucoidan has potential as a biological component in the adjuvant treatment of functional dyspepsia and for its expanded utilization in the food and medical fields.
Collapse
Affiliation(s)
- Tianxu Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
- Tianjin Key Laboratory of Food Science and Health, School of Medicine, Nankai University, Tianjin 300071, China
| | - Muyuan Ma
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Yonglin Wu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Ismail Muhammad Asif
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Daosen Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Lichong Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Minghui Zhang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Yijie Chen
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| | - Ling Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (T.L.); (M.M.)
- Key Laboratory of Environment Correlative Dietology (Huazhong Agricultural University), Ministry of Education, Wuhan 430070, China
| |
Collapse
|
2
|
Cui D, Geng X, Wang J, Han X, Huang Z, Jiang M, Gong Y. Efficacy and safety of LiWei capsule in chronic non-atrophic gastritis with erosions: a randomized controlled trial. Sci Rep 2025; 15:1620. [PMID: 39794448 PMCID: PMC11724083 DOI: 10.1038/s41598-025-85122-y] [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: 07/01/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
To assess the efficacy and safety of LiWei Capsule (LWC) in the treatment of chronic non-atrophic gastritis (CNG) with erosions and damp-heat stasis syndrome, based on Traditional Chinese Medicine (TCM) principles. This phase II, multicenter, randomized, double-blind, placebo- and positive-controlled trial enrolled patients diagnosed with CNG with erosions and damp-heat stasis syndrome. Participants were allocated to LWC, Sanjiu Weitai Capsule (SJWT, positive control), and placebo groups (2:1:1 ratio) and received corresponding treatment for 4 weeks, followed by a 16-week follow-up. The primary outcome was the curative rate of epigastric pain/bloating. Secondary outcomes included improvement in endoscopic examination, histopathological examination, and TCM symptom scores. Of 301 enrolled participants from five centers, 189 completed the study (95, 45, 49 cases in LWC, SJWT, and placebo groups, respectively). After 4 weeks of treatment, the curative rates of epigastric pain/bloating were 76.84%, 42.22%, and 22.45% in LWC, SJWT, and placebo groups, respectively (P < 0.001). The healing rates of endoscopic erosions were 65.3%, 46.7%, and 30.6% in LWC, SJWT, and placebo groups, respectively (P < 0.001). LWC effectively and safely alleviates epigastric pain/bloating and promotes endoscopic erosion healing in CNG patients with erosions (damp-heat stasis syndrome). LWC may be a promising treatment option for this condition.Trial registration: ChiCTR2100052010, assigned by the Chinese Clinical Trial Registry, registration date 13/10/2021.
Collapse
Affiliation(s)
- Danyang Cui
- General Hospital of Northern Theater Command, No.83 Cultural Road, Shenhe District, Shenyang, 110016, Liaoning, China
- Institute of Basic Research In Clinical Medicine, China Academy of Chinese Medical Sciences, Nanxiaojie 16#, Dongzhimennei, Beijing, 100700, China
| | - Xiao Geng
- General Hospital of Northern Theater Command, No.83 Cultural Road, Shenhe District, Shenyang, 110016, Liaoning, China
| | - Juan Wang
- Institute of Basic Research In Clinical Medicine, China Academy of Chinese Medical Sciences, Nanxiaojie 16#, Dongzhimennei, Beijing, 100700, China
| | - Xu Han
- Institute of Basic Research In Clinical Medicine, China Academy of Chinese Medical Sciences, Nanxiaojie 16#, Dongzhimennei, Beijing, 100700, China
| | - Zhijun Huang
- Jianmin Pharmaceutical Group Co., Ltd., Hubei, 430050, China
| | - Miao Jiang
- Institute of Basic Research In Clinical Medicine, China Academy of Chinese Medical Sciences, Nanxiaojie 16#, Dongzhimennei, Beijing, 100700, China.
| | - Yang Gong
- General Hospital of Northern Theater Command, No.83 Cultural Road, Shenhe District, Shenyang, 110016, Liaoning, China.
| |
Collapse
|
3
|
Chen L, Wang X, Li J, Zhang L, Wu W, Wei S, Zou W, Zhao Y. Elucidation of the mechanism of berberine against gastric mucosa injury in a rat model with chronic atrophic gastritis based on a combined strategy of multi-omics and molecular biology. Front Pharmacol 2025; 15:1499753. [PMID: 39834822 PMCID: PMC11743660 DOI: 10.3389/fphar.2024.1499753] [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: 09/21/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Background Berberine (BBR) is widely used to treat gastrointestinal diseases. However, the pharmacological mechanism of action of BBR in anti-chronic atrophic gastritis (CAG) remains unclear. This study aimed to investigate the mechanism of action of BBR in CAG by integration of molecular biology and multi-omics studies strategy. Methods The CAG model was established by alternating drinking water of 0.1% ammonia and 20 mmol/L sodium deoxycholate, accompanied by an irregular diet. Serum biochemical indices including PGI, PGII, GAS-17, IL-6, IL-1β, and TNF-α were analyzed. HE and AB-PAS staining were employed to assess pathological damage in gastric tissue. The underlying molecular mechanism of BBR in CAG treatment was explored via the integration of network pharmacology, transcriptomics, widely targeted metabolomics and intestinal flora analysis. Finally, relevant key targets and pathway were verified. Results The results showed that BBR exerted therapeutic effects in improving CAG via alleviating inflammation response, maintaining the gastric mucosal barrier's integrity and repairing gastric mucosal tissues. Network pharmacology showed that the treatment of CAG by BBR mainly involved in inflammatory response, apoptosis, angiogenesis and metabolic processes. Furthermore, 234 different expression genes were identified in the gastric tissue transcriptome, which were mainly involved in biological processes such as cell adhesion, angiogenesis, apoptosis, cell migration and lipids metabolism by regulating the MAPK signaling pathway. Metabolomics results showed that 125 differential metabolites were also identified, while the pathways were mainly involved in D-glutamine and D-glutamate metabolism, and tyrosine metabolism, etc. Integrating transcriptomics and metabolomics analyses indicated that BBR directly regulated Carnitine C3:0, LPC (0:0/20:3), L-Glutamic Acid and FFA (15:0) by acting on SLC25A20, PNLIPRP1, PLA2G4C, GSR, GFPT2, GCLM, CTPS1, ACSL1, ACOT4 and ACOT2. 16S rRNA sequencing revealed that BBR could restore the balance of gut microbiota dysbiosis by significantly regulating the relative abundance of unclassified_Muribaculaceae and Lactobacillus_johnsonii. Conclusion This study demonstrated that BBR alleviates CAG through the regulation of the MAPK signaling pathway, metabolic disorders and gut microbiota dysbiosis, thereby revealing the complex mechanism of BBR in relation to alleviating CAG from multiple levels and perspectives.
Collapse
Affiliation(s)
- Lisheng Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xin Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jianyu Li
- Department of Pharmacy, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lijuan Zhang
- Department of Pharmacy, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Wenbin Wu
- Healthcare Office of the Service Bureau of Agency for Offices Administration of the Central Military Commission, Beijing, China
| | - Shizhang Wei
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenjun Zou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanling Zhao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
4
|
Liu T, Chen Z, Sun L, Xiong L. Role of blood metabolites in mediating the effect of gut microbiota on chronic gastritis. Microbiol Spectr 2024; 12:e0149024. [PMID: 39404486 PMCID: PMC11537017 DOI: 10.1128/spectrum.01490-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 08/16/2024] [Indexed: 11/07/2024] Open
Abstract
Exploring the link between gut microbiota and chronic gastritis (CG), and assessing the potential mediating influence of blood metabolites. Using aggregated data from genome-wide association studies (GWAS), we performed a two-sample Mendelian randomization (MR) analysis to explore the genetic links between gut microbiota (412 types) and CG (623,822 cases). Furthermore, we utilized a two-step MR approach to measure the extent to which blood metabolites (1,400 types) mediate the impact of gut microbiota on CG. Through MR, we identified that three genetically predicted gut microbiota increased the risk of CG: the ubiquinol-8 biosynthesis pathway (OR 1.149, 95%CI 1.022-1.291), Odoribacter from the Porphyromonadaceae family (OR 1.260, 95%CI 1.044-1.523), and Coprococcus from the Lachnospiraceae family (OR 1.125, 95%CI 1.010-1.253). Currently, there is no evidence to suggest that genetically predicted CG affects the risk of gut microbiota. Four blood metabolites mediated the proportionate changes in genetically predicted gut microbiota: levels of 4-hydroxyphenylacetate levels by 14.9% (95% CI -0.559%, 30.3%), palmitoleate (16:1n7) levels, and the phosphate to alanine ratio together mediated the same microbiota by 6.97% (95% CI -1.61%, 15.6%) and 7.91% (95% CI -1.67%, 17.5%), while the phosphate to alanine ratio and X-12839 levels together mediated the same microbiota by 8.48% (95% CI -2.87%, 19.8%) and 10.7% (95% CI 0.353%, 21.1%). In conclusion, our research has confirmed a causal link between gut microbiota, blood metabolites, and CG. Metabolites such as 4-hydroxyphenylacetate levels, palmitoleate (16:1n7) levels, the phosphate to alanine ratio, and X-12839 levels have relatively significant mediating roles between gut microbiota and CG. These metabolites may influence the occurrence and development of CG by regulating inflammatory responses, energy metabolism, and gut barrier function. However, the majority of the influence of gut microbiota on CG remains unclear, necessitating further research into other potential mediating risk factors. Clinically, it is crucial to focus on patients suffering from CG who exhibit dysbiosis of gut microbiota.IMPORTANCEThe results indicate that interactions between particular gut microbiota and blood metabolites may significantly contribute to the onset and progression of CG. These findings offer new insights and potential targets for early diagnosis, personalized treatment, and prevention of CG.
Collapse
Affiliation(s)
- Tianying Liu
- College of Basic Medical Sciences, Changchun University of Traditional Medicine, Changchun, China
| | - Zhian Chen
- College of Integrative Medicine, Changchun University of Traditional Medicine, Changchun, China
| | - Li Sun
- Jilin Academy of Chinese Medical Sciences, Changchun, China
- Changchun University of Traditional Medicine, Changchun, China
| | - Lihui Xiong
- College of Basic Medical Sciences, Changchun University of Traditional Medicine, Changchun, China
| |
Collapse
|
5
|
Wang YM, Sun JH, Sun RX, Liu XY, Li JF, Li RZ, Du YR, Zhou XZ. Treating chronic atrophic gastritis: identifying sub-population based on real-world TCM electronic medical records. Front Pharmacol 2024; 15:1444733. [PMID: 39170704 PMCID: PMC11335612 DOI: 10.3389/fphar.2024.1444733] [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: 06/06/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Background and Objective Chronic atrophic gastritis (CAG) is a complex chronic disease caused by multiple factors that frequently occurs disease in the clinic. The worldwide prevalence of CAG is high. Interestingly, clinical CAG patients often present with a variety of symptom phenotypes, which makes it more difficult for clinicians to treat. Therefore, there is an urgent need to improve our understanding of the complexity of the clinical CAG population, obtain more accurate disease subtypes, and explore the relationship between clinical symptoms and medication. Therefore, based on the integrated platform of complex networks and clinical research, we classified the collected patients with CAG according to their different clinical characteristics and conducted correlation analysis on the classification results to identify more accurate disease subtypes to aid in personalized clinical treatment. Method Traditional Chinese medicine (TCM) offers an empirical understanding of the clinical subtypes of complicated disorders since TCM therapy is tailored to the patient's symptom profile. We gathered 6,253 TCM clinical electronic medical records (EMRs) from CAG patients and manually annotated, extracted, and preprocessed the data. A shared symptom-patient similarity network (PSN) was created. CAG patient subgroups were established, and their clinical features were determined through enrichment analysis employing community identification methods. Different clinical features of relevant subgroups were correlated based on effectiveness to identify symptom-botanical botanical drugs correspondence. Moreover, network pharmacology was employed to identify possible biological relationships between screened symptoms and medications and to identify various clinical and molecular aspects of the key subtypes using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. Results 5,132 patients were included in the study: 2,699 males (52.60%) and 2,433 females (47.41%). The population was divided into 176 modules. We selected the first 3 modules (M29, M3, and M0) to illustrate the characteristic phenotypes and genotypes of CAG disease subtypes. The M29 subgroup was characterized by gastric fullness disease and internal syndrome of turbidity and poison. The M3 subgroup was characterized by epigastric pain and disharmony between the liver and stomach. The M0 subgroup was characterized by epigastric pain and dampness-heat syndrome. In symptom analysis, The top symptoms for symptom improvement in all three subgroups were stomach pain, bloating, insomnia, poor appetite, and heartburn. However, the three groups were different. The M29 subgroup was more likely to have stomach distention, anorexia, and palpitations. Citrus medica, Solanum nigrum, Jiangcan, Shan ci mushrooms, and Dillon were the most popular botanical drugs. The M3 subgroup has a higher incidence of yellow urine, a bitter tongue, and stomachaches. Smilax glabra, Cyperus rotundus, Angelica sinensis, Conioselinum anthriscoides, and Paeonia lactiflora were the botanical drugs used. Vomiting, nausea, stomach pain, and appetite loss are common in the M0 subgroup. The primary medications are Scutellaria baicalensis, Smilax glabra, Picrorhiza kurroa, Lilium lancifolium, and Artemisia scoparia. Through GO and KEGG pathway analysis, We found that in the M29 subgroup, Citrus medica, Solanum nigrum, Jiangcan, Shan ci mushrooms, and Dillon may exert their therapeutic effects on the symptoms of gastric distension, anorexia, and palpitations by modulating apoptosis and NF-κB signaling pathways. In the M3 subgroup, Smilax glabra, Cyperus rotundus, Angelica sinensis, Conioselinum anthriscoides, and Paeonia lactiflora may be treated by NF-κB and JAK-STAT signaling pathway for the treatment of stomach pain, bitter mouth, and yellow urine. In the M0 subgroup, Scutellaria baicalensis, Smilax glabra, Picrorhiza kurroa, Lilium lancifolium, and Artemisia scoparia may exert their therapeutic effects on poor appetite, stomach pain, vomiting, and nausea through the PI3K-Akt signaling pathway. Conclusion Based on PSN identification and community detection analysis, CAG population division can provide useful recommendations for clinical CAG treatment. This method is useful for CAG illness classification and genotyping investigations and can be used for other complicated chronic diseases.
Collapse
Affiliation(s)
- Yu-man Wang
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Jian-hui Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, China
| | - Run-xue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, China
| | - Xiao-yu Liu
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Jing-fan Li
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Run-ze Li
- Graduate School of Hebei University of Traditional Chinese Medicine, Hebei, China
| | - Yan-ru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, China
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, China
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, China
| | - Xue-zhong Zhou
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| |
Collapse
|
6
|
Xu Y, Zhou C, Zong M, Zhu J, Guo X, Sun Z. High-protein high-konjac glucomannan diets changed glucose and lipid metabolism by modulating colonic microflora and bile acid profiles in healthy mouse models. Food Funct 2024; 15:4446-4461. [PMID: 38563504 DOI: 10.1039/d4fo00159a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
High protein and fiber diets are becoming increasingly popular for weight loss; however, the benefits or risks of high protein and fiber diets with a normal calorie level for healthy individuals still need to be elucidated. In this study, we explored the role and mechanisms of long-term high protein and/or konjac glucomannan diets on the metabolic health of healthy mouse models. We found that high konjac glucomannan contents improved the glucose tolerance of mice and both high protein and high konjac glucomannan contents improved the serum lipid profile but increased the TNF-α levels. In the liver, high dietary protein contents reduced the expression of the FASN gene related to fatty acid synthesis. Interactions of dietary protein and fiber were shown in the signaling pathways related to lipid and glucose metabolism of the liver and the inflammatory status of the colon, wherein the high protein and high konjac glucomannan diet downregulated the expression of the SREBF1 and FXR genes in the liver and downregulated the expression of TNF-α genes in the colon compared to the high protein diet. High konjac glucomannan contents reduced the colonic secondary bile acid levels including DCA and LCA; this was largely associated with the changed microbiota profile and also contributed to improved lipid and glucose homeostasis. In conclusion, high protein diets improved lipid homeostasis and were not a risk to metabolic health, while high fiber diets improved glucose and lipid homeostasis by modulating colonic microbiota and bile acid profiles, and a high protein diet supplemented with konjac glucomannan might improve hepatic lipid homeostasis and colonic inflammation in healthy mouse models through long-term intervention.
Collapse
Affiliation(s)
- Yetong Xu
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Chengyu Zhou
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Minyue Zong
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Junwei Zhu
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Xutong Guo
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| | - Zhihong Sun
- Laboratory for Bio-Feed and Molecular Nutrition, Department of Animal Science and Technology, Southwest University, Chongqing 400715, P. R. China.
| |
Collapse
|