1
|
Fiorucci S, Urbani G, Biagioli M, Sepe V, Distrutti E, Zampella A. Bile acids and bile acid activated receptors in the treatment of Covid-19. Biochem Pharmacol 2024; 228:115983. [PMID: 38081371 DOI: 10.1016/j.bcp.2023.115983] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 09/20/2024]
Abstract
Since its first outbreak in 2020, the pandemic caused by the Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) has caused the death of almost 7 million people worldwide. Vaccines have been fundamental in disease prevention and to reduce disease severity especially in patients with comorbidities. Nevertheless, treatment of COVID-19 has been proven difficult and several approaches have failed to prevent disease onset or disease progression, particularly in patients with comorbidities. Interrogation of drug data bases has been widely used since the beginning of pandemic to repurpose existing drugs/natural substances for the prevention/treatment of COVID-19. Steroids, including bile acids such as ursodeoxycholic acid (UDCA) and chenodeoxycholic acid (CDCA) have shown to be promising for their potential in modulating SARS-CoV-2/host interaction. Bile acids have proven to be effective in preventing binding of spike protein with the Angiotensin Converting Enzyme II (ACE2), thus preventing virus uptake by the host cells and inhibiting its replication, as well as in indirectly modulating immune response. Additionally, the two main bile acid activated receptors, GPBAR1 and FXR, have proven effective in modulating the expression of ACE2, suggesting an indirect role for these receptors in regulating SARS-CoV-2 infectiveness and immune response. In this review we have examined how the potential of bile acids and their receptors as anti-COVID-19 therapies and how these biochemical mechanisms translate into clinical efficacy.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy.
| | - Ginevra Urbani
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Michele Biagioli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Valentina Sepe
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | | | - Angela Zampella
- Department of Pharmacy, University of Naples Federico II, Naples, Italy
| |
Collapse
|
2
|
Alenezi T, Alrubaye B, Fu Y, Shrestha J, Algehani S, Wang H, Liyanage R, Sun X. Recombinant Bile Salt Hydrolase Enhances the Inhibition Efficiency of Taurodeoxycholic Acid against Clostridium perfringens Virulence. Pathogens 2024; 13:464. [PMID: 38921762 PMCID: PMC11206707 DOI: 10.3390/pathogens13060464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/27/2024] Open
Abstract
Clostridium perfringens is the main pathogen of chicken necrotic enteritis (NE) causing huge economic losses in the poultry industry. Although dietary secondary bile acid deoxycholic acid (DCA) reduced chicken NE, the accumulation of conjugated tauro-DCA (TDCA) raised concerns regarding DCA efficacy. In this study, we aimed to deconjugate TDCA by bile salt hydrolase (BSH) to increase DCA efficacy against the NE pathogen C. perfringens. Assays were conducted to evaluate the inhibition of C. perfringens growth, hydrogen sulfide (H2S) production, and virulence gene expression by TDCA and DCA. BSH activity and sequence alignment were conducted to select the bsh gene for cloning. The bsh gene from Bifidobacterium longum was PCR-amplified and cloned into plasmids pET-28a (pET-BSH) and pDR111 (pDR-BSH) for expressing the BSH protein in E. coli BL21 and Bacillus subtilis 168 (B-sub-BSH), respectively. His-tag-purified BSH from BL21 cells was evaluated by SDS-PAGE, Coomassie blue staining, and a Western blot (WB) assays. Secretory BSH from B. subtilis was analyzed by a Dot-Blot. B-sub-BSH was evaluated for the inhibition of C. perfringens growth. C. perfringens growth reached 7.8 log10 CFU/mL after 24 h culture. C. perfringens growth was at 8 vs. 7.4, 7.8 vs. 2.6 and 6 vs. 0 log10 CFU/mL in 0.2, 0.5, and 1 mM TDCA vs. DCA, respectively. Compared to TDCA, DCA reduced C. perfringens H2S production and the virulence gene expression of asrA1, netB, colA, and virT. BSH activity was observed in Lactobacillus johnsonii and B. longum under anaerobe but not L. johnsonii under 10% CO2 air. After the sequence alignment of bsh from ten bacteria, bsh from B. longum was selected, cloned into pET-BSH, and sequenced at 951 bp. After pET-BSH was transformed in BL21, BSH expression was assessed around 35 kDa using Coomassie staining and verified for His-tag using WB. After the subcloned bsh and amylase signal peptide sequence was inserted into pDR-BSH, B. subtilis was transformed and named B-sub-BSH. The transformation was evaluated using PCR with B. subtilis around 3 kb and B-sub-BSH around 5 kb. Secretory BSH expressed from B-sub-BSH was determined for His-tag using Dot-Blot. Importantly, C. perfringens growth was reduced greater than 59% log10 CFU/mL in the B-sub-BSH media precultured with 1 vs. 0 mM TDCA. In conclusion, TDCA was less potent than DCA against C. perfringens virulence, and recombinant secretory BSH from B-sub-BSH reduced C. perfringens growth, suggesting a new potential intervention against the pathogen-induced chicken NE.
Collapse
Affiliation(s)
- Tahrir Alenezi
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
- College of Medical Applied Sciences, The Northern Border University, Arar 91431, Saudi Arabia
| | - Bilal Alrubaye
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
| | - Ying Fu
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Janashrit Shrestha
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
| | - Samar Algehani
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Hong Wang
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| | - Rohana Liyanage
- Department of Chemistry, University of Arkansas, Fayetteville, AR 72701, USA;
| | - Xiaolun Sun
- Center of Excellence for Poultry Science, University of Arkansas, Fayetteville, AR 72701, USA; (B.A.); (J.S.); (S.A.); (H.W.)
- Cell and Molecular Biology Program, University of Arkansas, Fayetteville, AR 72701, USA
| |
Collapse
|
3
|
Tian S, Jiang Y, Han Q, Meng C, Ji F, Zhou B, Ye M. Putative Probiotic Ligilactobacillus salivarius Strains Isolated from the Intestines of Meat-Type Pigeon Squabs. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10289-1. [PMID: 38805143 DOI: 10.1007/s12602-024-10289-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 05/29/2024]
Abstract
This study aims to screen for potential probiotic lactic acid bacteria from the intestines of meat-type pigeon squabs. Ligilactobacillus salivarius YZU37 was identified as the best comprehensive performed strain. Being acid- and bile salt-tolerant, it displayed growth-inhibition activities against Staphylococcus aureus ATCC25923, Escherichia coli ATCC25922, and Salmonella typhimurium SL1344, exhibited sensitivity to 6 commonly used antibiotics, and endowed with good cell surface hydrophobicity, auto-aggregation property, and anti-oxidant activities. Results of in vitro experiments indicated that the bacteriostatic effects of this strain were related to the production of proteinaceous substances that depend on acidic conditions. Whole-genome sequencing of L. salivarius YZU37 was performed to elucidate the genetic basis underlying its probiotic potential. Pangenome analysis of L. salivarius YZU37 and other 212 L. salivarius strains available on NCBI database revealed a pigeon-unique gene coding choloylglycine hydrolase (CGH), which had higher enzyme-substrate binding affinity than that of the common CGH shared by L. salivarius strains of other sources. Annotation of the functional genes in the genome of L. salivarius YZU37 revealed genes involved in responses to acid, bile salt, heat, cold, heavy metal, and oxidative stresses. The whole genome analysis also revealed the absence of virulence and toxin genes and the presence of 65 genes distributed under 4 CAZymes classes, 2 CRISPR-cas regions, and 3 enterolysin A clusters which may confer the acid-dependent antimicrobial potential of L. salivarius YZU37. Altogether, our results highlighted the probiotic potential of L. salivarius YZU37. Further in vivo investigations are required to elucidate its beneficial effects on pigeons.
Collapse
Affiliation(s)
- Shaoqi Tian
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Yinhong Jiang
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Qiannan Han
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Chuang Meng
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Feng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100089, China
| | - Bin Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Manhong Ye
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
4
|
Zheng Y, Zhang S, Zhang Z, Zhang T, Teng X, Xiao G, Huang S. Isolation of Lactobacillus acidophilus strain and its anti-obesity effect in a diet induced obese murine model. Lett Appl Microbiol 2024; 77:ovae021. [PMID: 38400571 DOI: 10.1093/lambio/ovae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/15/2024] [Accepted: 02/22/2024] [Indexed: 02/25/2024]
Abstract
Intestinal microbiota is a potential determinant of obesity, with probiotic bile salt hydrolase (BSH) as one of the key mechanisms in the anti-obesity effects. In this study, we present a Lactobacillus acidophilus GOLDGUT-LA100 (LA100) with high BSH activity, good gastric acid and bile salt tolerance, and a potential anti-obesity effect. LA100's anti-obesity effects were evaluated in a high-fat diet-induced, obese mouse model. LA100 administration alleviates high-fat diet-induced pathophysiological symptoms, such as body weight gain, high serum glucose and cholesterol level, hepatic lipid accumulation, and adipose inflammation. These results demonstrate concrete anti-obesity benefit in animal models and show promising applications in future clinical studies.
Collapse
Affiliation(s)
- Yanyi Zheng
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | - Silu Zhang
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | - Zhizhu Zhang
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | | | - Xin Teng
- Bluepha Co., Ltd, Shenzhen 518000, China
| | - Guoxun Xiao
- Wonderlab Innovation Centre for Healthcare, Shenzhen Porshealth Bioengineering Co., Ltd, Shenzhen 518000, China
| | - Song Huang
- Bluepha Co., Ltd, Shenzhen 518000, China
- Department of Chemical and Biological Engineering, Xiamen University, Xiamen 361102, China
- School of Public Health, Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
5
|
Xu H, Yuan M, Niu K, Yang W, Jiang M, Zhang L, Zhou J. Involvement of Bile Acid Metabolism and Gut Microbiota in the Amelioration of Experimental Metabolism-Associated Fatty Liver Disease by Nobiletin. Molecules 2024; 29:976. [PMID: 38474489 DOI: 10.3390/molecules29050976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolism-associated fatty liver disease (MAFLD), a growing health problem worldwide, is one of the major risks for the development of cirrhosis and liver cancer. Oral administration of nobiletin (NOB), a natural citrus flavonoid, modulates the gut microbes and their metabolites in mice. In the present study, we established a mouse model of MAFLD by subjecting mice to a high-fat diet (HFD) for 12 weeks. Throughout this timeframe, NOB was administered to investigate its potential benefits on gut microbial balance and bile acid (BA) metabolism using various techniques, including 16S rRNA sequencing, targeted metabolomics of BA, and biological assays. NOB effectively slowed the progression of MAFLD by reducing serum lipid levels, blood glucose levels, LPS levels, and hepatic IL-1β and TNF-α levels. Furthermore, NOB reinstated diversity within the gut microbial community, increasing the population of bacteria that produce bile salt hydrolase (BSH) to enhance BA excretion. By exploring further, we found NOB downregulated hepatic expression of the farnesoid X receptor (FXR) and its associated small heterodimer partner (SHP), and it increased the expression of downstream enzymes, including cholesterol 7α-hydroxylase (CYP7A1) and cytochrome P450 27A1 (CYP27A1). This acceleration in cholesterol conversion within the liver contributes to mitigating MAFLD. The present findings underscore the significant role of NOB in regulating gut microbial balance and BA metabolism, revealing that long-term intake of NOB plays beneficial roles in the prevention or intervention of MAFLD.
Collapse
Affiliation(s)
- Hongling Xu
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Mingming Yuan
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Kailin Niu
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Wei Yang
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Maoyuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, China
| | - Lei Zhang
- School of Traditional Chinese Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| | - Jing Zhou
- Laboratory Animal Center Affiliate from Research Office, Sichuan Academy of Chinese Medicine Sciences, Chengdu 610041, China
| |
Collapse
|
6
|
Chen C, Gao K, Chen Z, Zhang Q, Ke X, Mao B, Fan Q, Li Y, Chen S. The supplementation of the multi-strain probiotics WHHPRO™ alleviates high-fat diet-induced metabolic symptoms in rats via gut-liver axis. Front Nutr 2024; 10:1324691. [PMID: 38274203 PMCID: PMC10808617 DOI: 10.3389/fnut.2023.1324691] [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: 10/20/2023] [Accepted: 12/27/2023] [Indexed: 01/27/2024] Open
Abstract
Metabolic syndrome (MS) has emerged as one of the major global health concerns, accompanied by a series of related complications, such as obesity and type-2 diabetes. The gut-liver axis (GLA) is a bidirectional communication between the gut and the liver. The GLA alterations have been revealed to be closely associated with the development of MS. Probiotics within Lactobacillus and Bifidobacterium confer beneficial effects on improving MS symptoms. WHHPRO™ is a mixture of four probiotic strains, with potential MS-improving abilities. This study aimed to investigate the effects of WHHPRO™ on MS symptoms using a high-fat diet (HFD) rat model. Oral administration of WHHPRO™ for 12 weeks improved glucose tolerance, blood lipid, body weight, and liver index in HFD rats. WHHPRO™ shaped the gut microbiome composition by increasing the abundance of Lactobacillus and Akkermansia and normalized the reduced SCFA levels in HFD rats. Besides, WHHPRO™ modulated the fecal bile acids (BAs) profile, with decreased levels of T-b-MCA and 12-KDCA and increased levels of LCA and ILCA. Meanwhile, WHHPRO™ increased total unconjugated BAs in feces and liver and reduced the accumulation of total hepatic BA pool size in HFD rats. Moreover, WHHPRO™ reversed the expression of genes associated with impaired BA metabolism signaling in the ileum and liver. Our findings suggest that WHHPRO™ exerted beneficial effects on improving MS symptoms, involving the modulation of the gut microbiome composition, SCFAs, and the FXR-FGF15 signaling along the GLA. Supplementation of WHHPRO™ may serve as a novel strategy for improving MS symptoms.
Collapse
Affiliation(s)
- Cailing Chen
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Kan Gao
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Zuoguo Chen
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Qiwen Zhang
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Xueqin Ke
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Bingyong Mao
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiuling Fan
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Yanjun Li
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| | - Su Chen
- Key Laboratory of Food and Biological Engineering of Zhejiang Province, Hangzhou, China
- Research and Development Department, Hangzhou Wahaha Group Co., Ltd., Hangzhou, China
- Hangzhou Wahaha Technology Co., Ltd., Hangzhou, China
| |
Collapse
|
7
|
Wu L, Zhou J, Zhou A, Lei Y, Tang L, Hu S, Wang S, Xiao X, Chen Q, Tu D, Lu C, Lai Y, Li Y, Zhang X, Tang B, Yang S. Lactobacillus acidophilus ameliorates cholestatic liver injury through inhibiting bile acid synthesis and promoting bile acid excretion. Gut Microbes 2024; 16:2390176. [PMID: 39205654 PMCID: PMC11364073 DOI: 10.1080/19490976.2024.2390176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/13/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Gut microbiota dysbiosis is involved in cholestatic liver diseases. However, the mechanisms remain to be elucidated. The purpose of this study was to examine the effects and mechanisms of Lactobacillus acidophilus (L. acidophilus) on cholestatic liver injury in both animals and humans. Bile duct ligation (BDL) was performed to mimic cholestatic liver injury in mice and serum liver function was tested. Gut microbiota were analyzed by 16S rRNA sequencing. Fecal bacteria transplantation (FMT) was used to evaluate the role of gut microbiota in cholestasis. Bile acids (BAs) profiles were analyzed by targeted metabolomics. Effects of L. acidophilus in cholestatic patients were evaluated by a randomized controlled clinical trial (NO: ChiCTR2200063330). BDL induced different severity of liver injury, which was associated with gut microbiota. 16S rRNA sequencing of feces confirmed the gut flora differences between groups, of which L. acidophilus was the most distinguished genus. Administration of L. acidophilus after BDL significantly attenuated hepatic injury in mice, decreased liver total BAs and increased fecal total BAs. Furthermore, after L. acidophilus treatment, inhibition of hepatic Cholesterol 7α-hydroxylase (CYP7α1), restored ileum Fibroblast growth factor 15 (FGF15) and Small heterodimer partner (SHP) accounted for BAs synthesis decrease, whereas enhanced BAs excretion was attributed to the increase of unconjugated BAs by enriched bile salt hydrolase (BSH) enzymes in feces. Similarly, in cholestasis patients, supplementation of L. acidophilus promoted the recovery of liver function and negatively correlated with liver function indicators, possibly in relationship with the changes in BAs profiles and gut microbiota composition. L. acidophilus treatment ameliorates cholestatic liver injury through inhibited hepatic BAs synthesis and enhances fecal BAs excretion.
Collapse
Affiliation(s)
- Lingyi Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jianchun Zhou
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - An Zhou
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuanyuan Lei
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiping Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Sumin Wang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xu Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Qiao Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Dianji Tu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Cheng Lu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yi Lai
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yiding Li
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Tibet, China
| | - Xiao Zhang
- Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Tibet, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
8
|
Chen CY, Ho HC. Roles of gut microbes in metabolic-associated fatty liver disease. Tzu Chi Med J 2023; 35:279-289. [PMID: 38035063 PMCID: PMC10683521 DOI: 10.4103/tcmj.tcmj_86_23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/04/2023] [Accepted: 05/31/2023] [Indexed: 12/02/2023] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is the most common chronic liver disease. Gut dysbiosis is considered a significant contributing factor in disease development. Increased intestinal permeability can be induced by gut dysbiosis, followed by the entry of lipopolysaccharide into circulation to reach peripheral tissue and result in chronic inflammation. We reviewed how microbial metabolites push host physiology toward MAFLD, including short-chain fatty acids (SCFAs), bile acids, and tryptophan metabolites. The effects of SCFAs are generally reported as anti-inflammatory and can improve intestinal barrier function and restore gut microbiota. Gut microbes can influence intestinal barrier function through SCFAs produced by fermentative bacteria, especially butyrate and propionate producers. This is achieved through the activation of free fatty acid sensing receptors. Bile is directly involved in lipid absorption. Gut microbes can alter bile acid composition by bile salt hydrolase-producing bacteria and bacterial hydroxysteroid dehydrogenase-producing bacteria. These bile acids can affect host physiology by activating farnesoid X receptor Takeda G protein-coupled receptor 5. Gut microbes can also induce MAFLD-associated symptoms by producing tryptophan metabolites kynurenine, serotonin, and indole-3-propionate. A summary of bacterial genera involved in SCFAs production, bile acid transformation, and tryptophan metabolism is provided. Many bacteria have demonstrated efficacy in alleviating MAFLD in animal models and are potential therapeutic candidates for MAFLD.
Collapse
Affiliation(s)
- Chun-Yao Chen
- Department of Biomedical Sciences and Engineering, Tzu Chi University, Hualien, Taiwan
| | - Han-Chen Ho
- Department of Anatomy, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
9
|
Liu L, Deng L, Wei W, Li C, Lu Y, Bai J, Li L, Zhang H, Jin N, Li C, Zhao C. Lactiplantibacillus plantarum LPJZ-658 Improves Non-Alcoholic Steatohepatitis by Modulating Bile Acid Metabolism and Gut Microbiota in Mice. Int J Mol Sci 2023; 24:13997. [PMID: 37762300 PMCID: PMC10531215 DOI: 10.3390/ijms241813997] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is one of the most prevalent diseases worldwide; it is characterized by hepatic lipid accumulation, inflammation, and progressive fibrosis. Here, a Western diet combined with low-dose weekly carbon tetrachloride was fed to C57BL/6J mice for 12 weeks to build a NASH model to investigate the attenuating effects and possible mechanisms of Lactiplantibacillus plantarum LPJZ-658. Hepatic pathology, lipid profiles, and gene expression were assessed. The metabolomic profiling of the serum was performed. The composition structure of gut microbiota was profiled using 16s rRNA sequencing. The results show that LPJZ-658 treatment significantly attenuated liver injury, steatosis, fibrosis, and inflammation in NASH mice. Metabolic pathway analysis revealed that several pathways, such as purine metabolism, glycerophospholipid metabolism, linoleic acid metabolism, and primary bile acid biosynthesis, were associated with NASH. Notably, we found that treatment with LPJZ-658 regulated the levels of bile acids (BAs) in the serum. Moreover, LPJZ-658 restored NASH-induced gut microbiota dysbiosis. The correlation analysis deduced obvious interactions between BAs and gut microbiota. The current study indicates that LPJZ-658 supplementation protects against NASH progression, which is accompanied by alternating BA metabolic and modulating gut microbiota.
Collapse
Affiliation(s)
- Liming Liu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Liquan Deng
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wei Wei
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China;
| | - Chunhua Li
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Yuting Lu
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| | - Jieying Bai
- College of Future Technology, Peking University, Beijing 100871, China;
| | - Letian Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Ningyi Jin
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Chang Li
- Research Unit of Key Technologies for Prevention and Control of Virus Zoonoses, Chinese Academy of Medical Sciences, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China; (L.L.); (N.J.)
| | - Cuiqing Zhao
- College of Animal Science and Technology, Jilin Agricultural Science and Technology University, Jilin 132101, China; (L.L.); (C.L.); (Y.L.)
| |
Collapse
|
10
|
Deng X, Lin B, Wang F, Xu P, Wang N. Specnuezhenide Ameliorates Age-Related Hepatic Lipid Accumulation via Modulating Bile Acid Homeostasis and Gut Microbiota in D-Galactose-Induced Mice. Metabolites 2023; 13:960. [PMID: 37623903 PMCID: PMC10456809 DOI: 10.3390/metabo13080960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/09/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023] Open
Abstract
Age-related hepatic lipid accumulation has become a major health problem in the elderly population. Specnuezhenide (SPN) is a major active iridoid glycoside from an edible herb Fructus Ligustri Lucidi, which is commonly used for preventing age-related diseases. However, the beneficial effects of SPN on age-related liver injury remain unknown. This study aimed to reveal the effect of SPN on age-related hepatic lipid accumulation and the underlying mechanism. D-galactose (D-gal)-induced aging mice were treated with vehicle or SPN for 12 weeks. Treatment of SPN decreased lipid accumulation and inflammation in the liver of D-gal-induced mice. Untargeted and targeted metabolomics showed that the SPN could regulate the bile acid (BA) synthesis pathway and restore the BA compositions in serum, livers, and feces of the D-gal-induced mice. Furthermore, SPN enhanced the protein and mRNA levels of hepatic BAs synthesis enzymes cytochrome P45027A1, cytochrome P4507A1, cytochrome P4507B1, and cytochrome P4508B1. Meanwhile, SPN alleviated D-gal-induced gut dysbiosis and reversed the proportions of microbes associated with bile salt hydrolase activity, including Lactobacillus, Ruminiclostridium, and Butyrivibrio. Our study revealed that SPN attenuated age-related hepatic lipid accumulation by improving BA profiles via modulating hepatic BA synthesis enzymes and gut microbiota.
Collapse
Affiliation(s)
- Xuehui Deng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310007, China; (X.D.)
| | - Bingfeng Lin
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China; (B.L.)
| | - Fang Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310007, China; (X.D.)
| | - Pingcui Xu
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China; (B.L.)
| | - Nani Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou 310007, China; (X.D.)
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou 310007, China; (B.L.)
| |
Collapse
|
11
|
Mezhibovsky E, Tveter KM, Villa-Rodriguez JA, Bacalia K, Kshatriya D, Desai N, Cabales A, Wu Y, Sui K, Duran RM, Bello NT, Roopchand DE. Grape Polyphenols May Prevent High-Fat Diet-Induced Dampening of the Hypothalamic-Pituitary-Adrenal Axis in Male Mice. J Endocr Soc 2023; 7:bvad095. [PMID: 37538101 PMCID: PMC10396072 DOI: 10.1210/jendso/bvad095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Indexed: 08/05/2023] Open
Abstract
Context Chronic high-fat diet (HFD) consumption causes obesity associated with retention of bile acids (BAs) that suppress important regulatory axes, such as the hypothalamic-pituitary-adrenal axis (HPAA). HFD impairs nutrient sensing and energy balance due to a dampening of the HPAA and reduced production and peripheral metabolism of corticosterone (CORT). Objective We assessed whether proanthocyanidin-rich grape polyphenol (GP) extract can prevent HFD-induced energy imbalance and HPAA dysregulation. Methods Male C57BL6/J mice were fed HFD or HFD supplemented with 0.5% w/w GPs (HFD-GP) for 17 weeks. Results GP supplementation reduced body weight gain and liver fat while increasing circadian rhythms of energy expenditure and HPAA-regulating hormones, CORT, leptin, and PYY. GP-induced improvements were accompanied by reduced mRNA levels of Il6, Il1b, and Tnfa in ileal or hepatic tissues and lower cecal abundance of Firmicutes, including known BA metabolizers. GP-supplemented mice had lower concentrations of circulating BAs, including hydrophobic and HPAA-inhibiting BAs, but higher cecal levels of taurine-conjugated BAs antagonistic to farnesoid X receptor (FXR). Compared with HFD-fed mice, GP-supplemented mice had increased mRNA levels of hepatic Cyp7a1 and Cyp27a1, suggesting reduced FXR activation and more BA synthesis. GP-supplemented mice also had reduced hepatic Abcc3 and ileal Ibabp and Ostβ, indicative of less BA transfer into enterocytes and circulation. Relative to HFD-fed mice, CORT and BA metabolizing enzymes (Akr1d1 and Srd5a1) were increased, and Hsd11b1 was decreased in GP supplemented mice. Conclusion GPs may attenuate HFD-induced weight gain by improving hormonal control of the HPAA and inducing a BA profile with less cytotoxicity and HPAA inhibition, but greater FXR antagonism.
Collapse
Affiliation(s)
- Esther Mezhibovsky
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Kevin M Tveter
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Jose A Villa-Rodriguez
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Karen Bacalia
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Dushyant Kshatriya
- Department of Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Nikhil Desai
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Alrick Cabales
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Yue Wu
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Ke Sui
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Rocio M Duran
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Nicholas T Bello
- Department of Animal Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| | - Diana E Roopchand
- Department of Food Science and NJ Institute for Food Nutrition and Health (Rutgers Center for Lipid Research; Center for Nutrition Microbiome and Health), Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
- Department of Nutritional Sciences Graduate Program, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA
| |
Collapse
|
12
|
Gadaleta RM, Cariello M, Crudele L, Moschetta A. Bile Salt Hydrolase-Competent Probiotics in the Management of IBD: Unlocking the "Bile Acid Code". Nutrients 2022; 14:3212. [PMID: 35956388 PMCID: PMC9370712 DOI: 10.3390/nu14153212] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/02/2022] [Accepted: 08/03/2022] [Indexed: 01/18/2023] Open
Abstract
Bile acid (BA) species and the gut microbiota (GM) contribute to intestinal mucosa homeostasis. BAs shape the GM and, conversely, intestinal bacteria with bile salt hydrolase (BSH) activity modulate the BA pool composition. The mutual interaction between BAs and intestinal microorganisms also influences mucosal barrier integrity, which is important for inflammatory bowel disease (IBD) pathogenesis, prevention and therapy. High levels of secondary BAs are detrimental for the intestinal barrier and increase the intestinal inflammatory response and dysbiosis. Additionally, a lack of BSH-active bacteria plays a role in intestinal inflammation and BA dysmetabolism. Thus, BSH-competent bacteria in probiotic formulations are being actively studied in IBD. At the same time, studies exploring the modulation of the master regulator of BA homeostasis, the Farnesoid X Receptor (FXR), in intestinal inflammation and how this impacts the GM are gaining significant momentum. Overall, the choice of probiotic supplementation should be a peculiar issue of personalized medicine, considering not only the disease but also the specific BA and metabolic signatures of a given patient.
Collapse
Affiliation(s)
- Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Lucilla Crudele
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
- INBB National Instituto for Biostructure and Biosystems, Viale delle Medaglie d’Oro 305, 00136 Rome, Italy
| |
Collapse
|
13
|
Rahman MRT, Fliss I, Biron E. Insights in the Development and Uses of Alternatives to Antibiotic Growth Promoters in Poultry and Swine Production. Antibiotics (Basel) 2022; 11:766. [PMID: 35740172 PMCID: PMC9219610 DOI: 10.3390/antibiotics11060766] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
The overuse and misuse of antibiotics has contributed to the rise and spread of multidrug-resistant bacteria. To address this global public health threat, many countries have restricted the use of antibiotics as growth promoters and promoted the development of alternatives to antibiotics in human and veterinary medicine and animal farming. In food-animal production, acidifiers, bacteriophages, enzymes, phytochemicals, probiotics, prebiotics, and antimicrobial peptides have shown hallmarks as alternatives to antibiotics. This review reports the current state of these alternatives as growth-promoting factors for poultry and swine production and describes their mode of action. Recent findings on their usefulness and the factors that presently hinder their broader use in animal food production are identified by SWOT (strength, weakness, opportunity, and threat) analysis. The potential for resistance development as well as co- and cross-resistance with currently used antibiotics is also discussed. Using predetermined keywords, we searched specialized databases including Scopus, Web of Science, and Google Scholar. Antibiotic resistance cannot be stopped, but its spreading can certainly be hindered or delayed with the development of more alternatives with innovative modes of action and a wise and careful use of antimicrobials in a One Health approach.
Collapse
Affiliation(s)
- Md Ramim Tanver Rahman
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada;
- Laboratory of Medicinal Chemistry, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC G1V 0A6, Canada;
| | - Ismail Fliss
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC G1V 0A6, Canada;
- Food Science Department, Faculty of Agriculture and Food Sciences, Université Laval, Québec, QC G1V 0A6, Canada
| | - Eric Biron
- Faculty of Pharmacy, Université Laval, Québec, QC G1V 0A6, Canada;
- Laboratory of Medicinal Chemistry, CHU de Québec Research Center, Québec, QC G1V 4G2, Canada
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC G1V 0A6, Canada;
| |
Collapse
|
14
|
Hari S, Burns GL, Hoedt EC, Keely S, Talley NJ. Eosinophils, Hypoxia-Inducible Factors, and Barrier Dysfunction in Functional Dyspepsia. FRONTIERS IN ALLERGY 2022; 3:851482. [PMID: 35769556 PMCID: PMC9234913 DOI: 10.3389/falgy.2022.851482] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Functional dyspepsia (FD) is a highly prevalent disorder of gut-brain interaction (DGBI), previously known as a functional gastrointestinal disorder. Characterized by early satiety, postprandial fullness, and/or epigastric pain or burning, diagnosis depends on positive symptomatology and exclusion of obvious structural diseases. A subtle inflammatory phenotype has been identified in FD patients, involving an increase in duodenal mucosal eosinophils, and imbalances in the duodenal gut microbiota. A dysregulated epithelial barrier has also been well described in FD and is thought to be a contributing factor to the low-grade duodenal inflammation observed, however the mechanisms underpinning this are poorly understood. One possible explanation is that alterations in the microbiota and increased immune cells can result in the activation of cellular stress response pathways to perpetuate epithelial barrier dysregulation. One such cellular response pathway involves the stabilization of hypoxia-inducible factors (HIF). HIF, a transcriptional protein involved in the cellular recognition and adaptation to hypoxia, has been identified as a critical component of various pathologies, from cancer to inflammatory bowel disease (IBD). While the contribution of HIF to subtle inflammation, such as that seen in FD, is unknown, HIF has been shown to have roles in regulating the inflammatory response, particularly the recruitment of eosinophils, as well as maintaining epithelial barrier structure and function. As such, we aim to review our present understanding of the involvement of eosinophils, barrier dysfunction, and the changes to the gut microbiota including the potential pathways and mechanisms of HIF in FD. A combination of PubMed searches using the Mesh terms functional dyspepsia, functional gastrointestinal disorders, disorders of gut-brain interaction, duodenal eosinophilia, barrier dysfunction, gut microbiota, gut dysbiosis, low-grade duodenal inflammation, hypoxia-inducible factors (or HIF), and/or intestinal inflammation were undertaken in the writing of this narrative review to ensure relevant literature was included. Given the findings from various sources of literature, we propose a novel hypothesis involving a potential role for HIF in the pathophysiological mechanisms underlying FD.
Collapse
Affiliation(s)
- Suraj Hari
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, TAS, Australia
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
| | - Grace L. Burns
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| | - Emily C. Hoedt
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| | - Simon Keely
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| | - Nicholas J. Talley
- NHMRC Centre of Research Excellence in Digestive Health, University of Newcastle, Newcastle, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- *Correspondence: Nicholas J. Talley
| |
Collapse
|
15
|
Poudel S, Tabler GT, Lin J, Zhai W, Zhang L. Riboflavin and Bacillus subtilis effects on growth performance and
woody-breast of Ross 708 broilers with or without Eimeria spp.
challenge. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:443-461. [PMID: 35709099 PMCID: PMC9184709 DOI: 10.5187/jast.2022.e24] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/02/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
This study was conducted to assess the effects of the dietary supplementation of
riboflavin (as a bile salt hydrolase [BSH] inhibitor) and Bacillus
subtilis on growth performance and woody breast of male broilers
challenged with Eimeria spp. Intestinal bacteria, including
supplemented probiotics, can produce BSH enzymes that deconjugate conjugated
bile salts and reduce fat digestion. A 3 × 2 × 2 (riboflavin
× Bacillus subtilis × Eimeria
spp. challenge) factorial arrangement of treatments in randomized complete block
design was used. On d 14, birds were gavaged with 20× doses of commercial
cocci vaccine (CoccivacR-B52, Merck Animal Health, Omaha, NE).
Dietary treatment of riboflavin and B. subtilis did not affect
body weight (BW), body weight gain (BWG), and feed conversion (FCR) d 0 to 14
and overall d 0 to 41. Eimeria spp challenge reduced BWG, feed
intake (FI), and increased FCR between d 14 to 28, but increased BWG and lowered
FCR between d 28 to 35. There were no effects of the Eimeria
spp. challenge on the overall d 0 to 41 FCR and FI, but BWG was reduced.
Eimeria spp. challenge increased the abdominal fat pad
weight and slight woody breast incidences on processed birds on d 42. Dietary
inclusion of B. subtilis and riboflavin at tested levels did
not help birds to mitigate the negative impact of Eimeria spp.
challenge to enhance the growth performance.
Collapse
Affiliation(s)
- Sabin Poudel
- Department of Poultry Science, Mississippi
State University, MS 39762, USA
| | - George T. Tabler
- Department of Poultry Science, Mississippi
State University, MS 39762, USA
| | - Jun Lin
- Department of Animal Science, University
of Tennessee, Knoxville, TN 37996, USA
| | - Wei Zhai
- Department of Poultry Science, Mississippi
State University, MS 39762, USA
| | - Li Zhang
- Department of Poultry Science, Mississippi
State University, MS 39762, USA
- Corresponding author: Li Zhang, Department of
Poultry Science, Mississippi State University, MS 39762, USA. Tel:
+1-662-325-3416, E-mail:
| |
Collapse
|
16
|
Zhao WW, Xiao M, Wu X, Li XW, Li XX, Zhao T, Yu L, Chen XQ. Ilexsaponin A 1 Ameliorates Diet-Induced Nonalcoholic Fatty Liver Disease by Regulating Bile Acid Metabolism in Mice. Front Pharmacol 2022; 12:771976. [PMID: 34970143 PMCID: PMC8712733 DOI: 10.3389/fphar.2021.771976] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Bile acid (BA) metabolism is an attractive therapeutic target in nonalcoholic fatty liver disease (NAFLD). We aimed to investigate the effect of ilexsaponin A1 (IsA), a major bioactive ingredient of Ilex, on high-fat diet (HFD)-induced NAFLD in mice with a focus on BA homeostasis. Male C57BL/6J mice were fed an HFD to induce NAFLD and were treated with IsA (120 mg/kg) for 8 weeks. The results showed that administration of IsA significantly decreased serum total cholesterol (TC), attenuated liver steatosis, and decreased total hepatic BA levels in HFD-induced NAFLD mice. IsA-treated mice showed increased BA synthesis in the alternative pathway by upregulating the gene expression levels of sterol 27-hydroxylase (CYP27A1) and cholesterol 7b-hydroxylase (CYP7B1). IsA treatment accelerated efflux and decreased uptake of BA in liver by increasing hepatic farnesoid X receptor (FXR) and bile salt export pump (BSEP) expression, and reducing Na+-taurocholic acid cotransporting polypeptide (NTCP) expression. Alterations in the gut microbiota and increased bile salt hydrolase (BSH) activity might be related to enhanced fecal BA excretion in IsA-treated mice. This study demonstrates that consumption of IsA may prevent HFD-induced NAFLD and exert cholesterol-lowering effects, possibly by regulating the gut microbiota and BA metabolism.
Collapse
Affiliation(s)
- Wen-Wen Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children Health, Beijing, China
| | - Meng Xiao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,National Institutes for Food and Drug Control, Beijing, China
| | - Xia Wu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiu-Wei Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiao-Xi Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ting Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Lan Yu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiao-Qing Chen
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Ocaña-Wilhelmi L, Martín-Núñez GM, Ruiz-Limón P, Alcaide J, García-Fuentes E, Gutiérrez-Repiso C, Tinahones FJ, Moreno-Indias I. Gut Microbiota Metabolism of Bile Acids Could Contribute to the Bariatric Surgery Improvements in Extreme Obesity. Metabolites 2021; 11:metabo11110733. [PMID: 34822391 PMCID: PMC8620296 DOI: 10.3390/metabo11110733] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/03/2023] Open
Abstract
Bariatric surgery is the only procedure to obtain and maintain weight loss in the long term, although the mechanisms driving these benefits are not completely understood. In the last years, gut microbiota has emerged as one of the drivers through its metabolites, especially secondary bile acids. In the current study, we have compared the gut microbiota and the bile acid pool, as well as anthropometric and biochemical parameters, of patient with morbid obesity who underwent bariatric surgery by two different techniques, namely Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG). Gut microbiota populations differed after the respective procedures, particularly with respect to the Enterobacteriaceae family. Both techniques resulted in changes in the bile acids pool, but RYGB was the procedure which suffered the greatest changes, with a reduction in most of their levels. Blautia and Veillonella were the two genera that more relationships showed with secondary bile acids, indicating a possible role in their formation and inhibition, respectively. Correlations with the anthropometric and biochemical variables showed that secondary bile acids could have a role in the amelioration of the glucose and HDL-cholesterol levels. Thus, we have observed a possible relationship between the interaction of the bile acids pool metabolized by the gut microbiota in the metabolic improvements obtained by bariatric surgery in the frame of morbid obesity, deserving further investigation in greater cohorts to decipher the role of each bile acid in the homeostasis of the host for their possible use in the development of microbiota-based therapeutics, such as new drugs, postbiotics or probiotics.
Collapse
Affiliation(s)
- Luis Ocaña-Wilhelmi
- Departamento de Especialidades Quirúrgicas, Bioquímica e Inmunología, Universidad de Málaga, 29010 Málaga, Spain;
- Unidad de Gestión Clínica de Cirugía General y del Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Málaga, Spain
| | - Gracia María Martín-Núñez
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
| | - Patricia Ruiz-Limón
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
| | - Juan Alcaide
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
| | - Eduardo García-Fuentes
- Department of Gastroenterology, Virgen de la Victoria University Hospital, Institute of Biomedical Research in Málaga (IBIMA), Málaga University, 29010 Málaga, Spain;
- CIBER Enfermedades Hepáticas y Digestivas-CIBEREHD, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Carolina Gutiérrez-Repiso
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
- Correspondence: (C.G.-R.); (F.J.T.); (I.M.-I.)
| | - Francisco J. Tinahones
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
- Correspondence: (C.G.-R.); (F.J.T.); (I.M.-I.)
| | - Isabel Moreno-Indias
- Department of Endocrinology and Nutrition, Virgen de la Victoria Hospital, Instituto de Investigación Biomédica de Málaga (IBIMA), Málaga University, 29010 Málaga, Spain; (G.M.M.-N.); (P.R.-L.); (J.A.)
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 29029 Madrid, Spain
- Correspondence: (C.G.-R.); (F.J.T.); (I.M.-I.)
| |
Collapse
|
18
|
Importance of Conjugation of the Bile Salt on the Mechanism of Lipolysis. Molecules 2021; 26:molecules26195764. [PMID: 34641309 PMCID: PMC8510408 DOI: 10.3390/molecules26195764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 12/17/2022] Open
Abstract
We aim to advance the discussion on the significance of the conjugation of bile salts (BS) in our organism. We hypothesize that conjugation influences the rate of lipolysis. Since the rate of lipolysis is a compound parameter, we compare the effect of conjugation on four surface parameters, which contribute to the rate. Since deconjugation is due to gut microbiota, we hypothesize that microbiota may affect the rate of lipolysis. A meta-analysis of literature data of critical micelle concentration, β, aggregation number, and molar solubilization ratio has been performed for the first time. In addition, critical micelle concentration (CMC), interfacial tension, and lipolysis rate measurements were performed. It was found that the unconjugated BS in mixed micelles increases the antagonism between the BS, therefore, increasing the CMC. This correlated with the effect of unconjugated BS on the solubilization capacity of mixed micelles. The collected literature information indicates that the role of the BS and its conjugation in our organism is a key factor influencing the functioning of our organism, where too high levels of unconjugated BS may lead to malabsorption of fat-soluble nutrients. The experimental lipolysis results irrevocably showed that conjugation is a significant factor influencing the rate.
Collapse
|
19
|
Bile Salt Hydrolases: At the Crossroads of Microbiota and Human Health. Microorganisms 2021; 9:microorganisms9061122. [PMID: 34067328 PMCID: PMC8224655 DOI: 10.3390/microorganisms9061122] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota has been increasingly linked to metabolic health and disease over the last few decades. Several factors have been suggested to be involved in lipid metabolism and metabolic responses. One mediator that has gained great interest as a clinically important enzyme is bile salt hydrolase (BSH). BSH enzymes are widely distributed in human gastrointestinal microbial communities and are believed to play key roles in both microbial and host physiology. In this review, we discuss the current evidence related to the role of BSHs in health and provide useful insights that may pave the way for new therapeutic targets in human diseases.
Collapse
|
20
|
Poudel S, Zhang L, Tabler GT, Lin J, Zhai W. Effects of riboflavin and Bacillus subtilis on internal organ development and intestinal health of Ross 708 male broilers with or without coccidial challenge. Poult Sci 2021; 100:100973. [PMID: 33588345 PMCID: PMC7896149 DOI: 10.1016/j.psj.2020.12.070] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 11/28/2022] Open
Abstract
In a companion study, we found that inclusion of different doses of riboflavin affected growth performance of Ross 708 male broilers' responses to coccidial challenge (by 5 Eimeria spp on day 14 of age) and dietary Bacillus subtilis (B. subtilis) supplementation. The current study was conducted to further test whether supplementation of B. subtilis and riboflavin will reduce negative impact and inflammation caused by Eimeria spp proliferation and help proper function of internal organs. A total of 1,248 Ross × Ross 708 male broiler chicks were randomly placed in 96 floor pens (8 blocks, 12 treatments). Treatments were arranged in a 3 (riboflavin) × 2 (B. subtilis) × 2 (Coccidial challenge) factorial arrangement in a randomized complete block design. Coccidial challenge reduced the weight of sampled birds on day 27 and day 36 and increased the relative weights of the internal organs of proventriculus, duodenum, jejunum, ileum, and spleen to BW on day 27, which may be because of inflammation caused by proliferation of Eimeria spp. The increased relative weights of duodenum, jejunum, ileum, and spleen on coccidial challenged birds were lost on day 36. Correlation analysis also indicated that the jejunum weight was positively related to villus height, Eimeria acervulina, and Eimeria maxima on day 27 but was not on day 36. The loss of the positive relationships may be because of recovery of the birds from coccidiosis on day 36. Even though the coccidial challenge and riboflavin interactively affected feed conversion ratio and BW gain and supplementation of dietary B. subtilis reduced mortality from day 35 to 42 in the companion study, the same response of internal organs was not observed in the current study. Coccidial challenge compromised development of internal organs of Ross 708 male broilers at an early age, but the negative effects subsided with age of birds rather than supplementation of riboflavin and B. subtilis at current tested levels under our experimental set up.
Collapse
Affiliation(s)
- Sabin Poudel
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA
| | - Li Zhang
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA
| | - George T Tabler
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA
| | - Jun Lin
- Department of Animal Science, University of Tennessee, Knoxville, TN, USA
| | - Wei Zhai
- Department of Poultry Science, Mississippi State University, Mississippi State, MS, USA.
| |
Collapse
|
21
|
Fiorucci S, Distrutti E, Carino A, Zampella A, Biagioli M. Bile acids and their receptors in metabolic disorders. Prog Lipid Res 2021; 82:101094. [PMID: 33636214 DOI: 10.1016/j.plipres.2021.101094] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/03/2021] [Accepted: 02/12/2021] [Indexed: 02/08/2023]
Abstract
Bile acids are a large family of atypical steroids which exert their functions by binding to a family of ubiquitous cell membrane and nuclear receptors. There are two main bile acid activated receptors, FXR and GPBAR1, that are exclusively activated by bile acids, while other receptors CAR, LXRs, PXR, RORγT, S1PR2and VDR are activated by bile acids in addition to other more selective endogenous ligands. In the intestine, activation of FXR and GPBAR1 promotes the release of FGF15/19 and GLP1 which integrate their signaling with direct effects exerted by theother receptors in target tissues. This network is tuned in a time ordered manner by circadian rhythm and is critical for the regulation of metabolic process including autophagy, fast-to-feed transition, lipid and glucose metabolism, energy balance and immune responses. In the last decade FXR ligands have entered clinical trials but development of systemic FXR agonists has been proven challenging because their side effects including increased levels of cholesterol and Low Density Lipoproteins cholesterol (LDL-c) and reduced High-Density Lipoprotein cholesterol (HDL-c). In addition, pruritus has emerged as a common, dose related, side effect of FXR ligands. Intestinal-restricted FXR and GPBAR1 agonists and dual FXR/GPBAR1 agonists have been developed. Here we review the last decade in bile acids physiology and pharmacology.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy.
| | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Adriana Carino
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Napoli, Federico II, Napoli, Italy
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
22
|
Abstract
Bile acids are a group of chemically different steroids generated at the host/microbial interface. Indeed, while primary bile acids are the end-product of cholesterol breakdown in the host liver, secondary bile acids are the products of microbial metabolism. Primary and secondary bile acids along with their oxo derivatives have been identified as signaling molecules acting on a family of cell membrane and nuclear receptors collectively known as "bile acid-activated receptors." Members of this group of receptors are highly expressed throughout the gastrointestinal tract and mediate the bilateral communications of the intestinal microbiota with the host immune system. The expression and function of bile acid-activated receptors FXR, GPBAR1, PXR, VDR, and RORγt are highly dependent on the structure of the intestinal microbiota and negatively regulated by intestinal inflammation. Studies from gene ablated mice have demonstrated that FXR and GPBAR1 are essential to maintain a tolerogenic phenotype in the intestine, and their ablation promotes the polarization of intestinal T cells and macrophages toward a pro-inflammatory phenotype. RORγt inhibition by oxo-bile acids is essential to constrain Th17 polarization of intestinal lymphocytes. Gene-wide association studies and functional characterizations suggest a potential role for impaired bile acid signaling in development inflammatory bowel diseases (IBD). In this review, we will focus on how bile acids and their receptors mediate communications of intestinal microbiota with the intestinal immune system, describing dynamic changes of bile acid metabolism in IBD and the potential therapeutic application of targeting bile acid signaling in these disorders.
Collapse
|
23
|
Sato K, Kakiyama G, Suzuki M, Naritaka N, Takei H, Sato H, Kimura A, Murai T, Kurosawa T, Pandak WM, Nittono H, Shimizu T. Changes in conjugated urinary bile acids across age groups. Steroids 2020; 164:108730. [PMID: 32961239 DOI: 10.1016/j.steroids.2020.108730] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 08/07/2020] [Accepted: 09/06/2020] [Indexed: 11/26/2022]
Abstract
Bile acid compositions are known to change dramatically after birth with aging. However, no reports have described the transition of conjugated urinary bile acids from the neonatal period to adulthood, and such findings would noninvasively offer insights into hepatic function. The aim of this study was to investigate differences in bile acid species, conjugation rates, and patterns, and to pool characteristics for age groups. We measured urinary bile acids in spot urine samples from 92 healthy individuals ranging from birth to 58 years old using liquid chromatography tandem mass spectrometry (LC/ESI-MS/MS). Sixty-six unconjugated and conjugated bile acids were systematically determined. After birth, urinary bile acids dramatically changed from fetal (i.e., Δ4-, Δ5-, and polyhydroxy-bile acids) to mature (i.e., CA and CDCA) bile acids. Peak bile acid excretion was 6-8 days after birth, steadily decreasing thereafter. A major change in bile acid conjugation pattern (taurine to glycine) also occurred at 2-4 months old. Our data provide important information regarding transitions of bile acid biosynthesis, including conjugation. The data also support the existence of physiologic cholestasis in the neonatal period and the establishment of the intestinal bacterial flora in infants.
Collapse
Affiliation(s)
- Keiko Sato
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Genta Kakiyama
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and McGuire VA Medical Center, 1201 Broad Rock Blvd., Richmond, VA 23249, USA.
| | - Mitsuyoshi Suzuki
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Nakayuki Naritaka
- Junshin Clinic BA Institute, 2-1-22 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Hajime Takei
- Junshin Clinic BA Institute, 2-1-22 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Hiroaki Sato
- Department of Perinatal and Neonatal Medicine, Saitama Medical Center, Jichi Medical University, 1-847 Amanuma-cho, Omiya-ku, Saitama 330-8503, Japan.
| | - Akihiko Kimura
- Department of Pediatrics and Child Health, Kurume University School of Medicine, 67 Asahi-cho, Kurume, Fukuoka 830-0011, Japan.
| | - Tsuyoshi Murai
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Tohbetsu-cho, Ishikari, Hokkaido 061-0293, Japan.
| | - Takao Kurosawa
- Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, 1757 Kanazawa, Tohbetsu-cho, Ishikari, Hokkaido 061-0293, Japan.
| | - William M Pandak
- Division of Gastroenterology, Hepatology, and Nutrition, Virginia Commonwealth University and McGuire VA Medical Center, 1201 Broad Rock Blvd., Richmond, VA 23249, USA.
| | - Hiroshi Nittono
- Junshin Clinic BA Institute, 2-1-22 Haramachi, Meguro-ku, Tokyo 152-0011, Japan.
| | - Toshiaki Shimizu
- Department of Pediatrics, Juntendo University, Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
24
|
Viñado A, Castillejos L, Barroeta AC. Soybean lecithin as an alternative energy source for grower and finisher broiler chickens: impact on performance, fatty acid digestibility, gut health, and abdominal fat saturation degree. Poult Sci 2020; 99:5653-5662. [PMID: 33142483 PMCID: PMC7647704 DOI: 10.1016/j.psj.2020.06.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/20/2020] [Accepted: 06/17/2020] [Indexed: 11/16/2022] Open
Abstract
An experiment was performed to assess the inclusion of soybean lecithin (SL) in the replacement of soybean oil (SO), for grower and finisher broiler chicken diets (up to 15 d of life), and its effects on performance, fatty acid (FA) absorption, gut health, and saturation degree of the abdominal fat pad (AFP). A total of 1,440 female Ross-308 chickens were distributed in 60 pens and were fed 5 experimental diets. The control diet (T1) was supplemented with SO (grower and finisher diets at 2.00%), and 4 levels of SL were included in replacement: T2 (0.25% in grower and 0.50% in finisher diets), T3 (0.50% in grower and 1.00% in finisher diets), T4 (0.75% in grower and 1.50% in finisher diets), and T5 (1.00% in grower and 2.00% in finisher diets). At day 39, titanium dioxide was added to finisher diets at 5 g/kg to perform a digestibility balance. At day 46, AFP, tissue, and gut digesta samples were collected to characterize FA digestibility, adipose saturation degree, microbial groups, and histomorphometry. No effects were associated with SO replacement by SL on performance (P > 0.05), ileal digestibility of total, saturated and monounsaturated FA (P > 0.05), nor jejunal morphology (P > 0.05). Total replacement of SO by SL reduced ileal absorption of polyunsaturated FA (P < 0.02) and increased jejunal Lactobacillus spp. counts (P = 0.049). Higher levels of SL inclusion (T4 and T5) lowered polyunsaturated FA concentration of the AFP (P = 0.002) and, thus, slightly reduced its unsaturated-to-saturated FA ratio (P = 0.005). Soybean lecithin inclusion did not modify performance parameters, total FA absorption, nor jejunal morphology, however caused changes on polyunsaturated FA absorption, jejunal microbiota, and saturation degree of the AFP. The study demonstrates that soybean lecithin can be included, in combination with or in replacement of soybean oil, as an alternative energy source for grower (up to a 1%) and finisher broiler diets (up to 2%).
Collapse
Affiliation(s)
- A Viñado
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Facultat de Veterinària, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Barcelona, Spain
| | - L Castillejos
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Facultat de Veterinària, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Barcelona, Spain.
| | - A C Barroeta
- Animal Nutrition and Welfare Service, Department of Animal and Food Science, Facultat de Veterinària, Universitat Autònoma de Barcelona, E-08193 Bellaterra, Barcelona, Spain
| |
Collapse
|
25
|
Li C, Ji Q, He T, Liu Y, Ma Y. Characterization of a recombinant bile salt hydrolase (BSH) from Bifidobacterium bifidum for its glycine-conjugated bile salts specificity. BIOCATAL BIOTRANSFOR 2020. [DOI: 10.1080/10242422.2020.1804881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Chou Li
- College of Marine and Bio-engineering, Yancheng Teachers' University, Yancheng, PR China
| | - Qingzhi Ji
- School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng, PR China
| | - Tongyao He
- School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng, PR China
| | - Yingying Liu
- School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng, PR China
| | - Yunqing Ma
- School of Pharmacy, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, Yancheng Teachers' University, Yancheng, PR China
| |
Collapse
|
26
|
Abstract
Vertebrates synthesize a diverse set of steroids and bile acids that undergo bacterial biotransformations. The endocrine literature has principally focused on the biochemistry and molecular biology of host synthesis and tissue-specific metabolism of steroids. Host-associated microbiota possess a coevolved set of steroid and bile acid modifying enzymes that match the majority of host peripheral biotransformations in addition to unique capabilities. The set of host-associated microbial genes encoding enzymes involved in steroid transformations is known as the sterolbiome. This review focuses on the current knowledge of the sterolbiome as well as its importance in medicine and agriculture.
Collapse
|
27
|
Liu Y, Chen K, Li F, Gu Z, Liu Q, He L, Shao T, Song Q, Zhu F, Zhang L, Jiang M, Zhou Y, Barve S, Zhang X, McClain CJ, Feng W. Probiotic Lactobacillus rhamnosus GG Prevents Liver Fibrosis Through Inhibiting Hepatic Bile Acid Synthesis and Enhancing Bile Acid Excretion in Mice. Hepatology 2020; 71:2050-2066. [PMID: 31571251 PMCID: PMC7317518 DOI: 10.1002/hep.30975] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/22/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS Cholestatic liver disease is characterized by gut dysbiosis and excessive toxic hepatic bile acids (BAs). Modification of gut microbiota and repression of BA synthesis are potential strategies for the treatment of cholestatic liver disease. The purpose of this study was to examine the effects and to understand the mechanisms of the probiotic Lactobacillus rhamnosus GG (LGG) on hepatic BA synthesis, liver injury, and fibrosis in bile duct ligation (BDL) and multidrug resistance protein 2 knockout (Mdr2-/- ) mice. APPROACH AND RESULTS Global and intestine-specific farnesoid X receptor (FXR) inhibitors were used to dissect the role of FXR. LGG treatment significantly attenuated liver inflammation, injury, and fibrosis with a significant reduction of hepatic BAs in BDL mice. Hepatic concentration of taurine-β-muricholic acid (T-βMCA), an FXR antagonist, was markedly increased in BDL mice and reduced in LGG-treated mice, while chenodeoxycholic acid, an FXR agonist, was decreased in BDL mice and normalized in LGG-treated mice. LGG treatment significantly increased the expression of serum and ileum fibroblast growth factor 15 (FGF-15) and subsequently reduced hepatic cholesterol 7α-hydroxylase and BA synthesis in BDL and Mdr2-/- mice. At the molecular level, these changes were reversed by global and intestine-specific FXR inhibitors in BDL mice. In addition, LGG treatment altered gut microbiota, which was associated with increased BA deconjugation and increased fecal and urine BA excretion in both BDL and Mdr2-/- mice. In vitro studies showed that LGG suppressed the inhibitory effect of T-βMCA on FXR and FGF-19 expression in Caco-2 cells. CONCLUSION LGG supplementation decreases hepatic BA by increasing intestinal FXR-FGF-15 signaling pathway-mediated suppression of BA de novo synthesis and enhances BA excretion, which prevents excessive BA-induced liver injury and fibrosis in mice.
Collapse
Affiliation(s)
- Yunhuan Liu
- Department of MedicineUniversity of LouisvilleLouisvilleKY
| | - Kefei Chen
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Department of Liver Surgery and Liver Transplantation CenterWest China HospitalSichuan UniversityChengduChina
| | - Fengyuan Li
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKY
| | - Zelin Gu
- Department of MedicineUniversity of LouisvilleLouisvilleKY
| | - Qi Liu
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- The Second Affiliated HospitalWenzhou Medical UniversityWenzhouChina
| | - Liqing He
- Department of ChemistryUniversity of LouisvilleLouisvilleKY
| | - Tuo Shao
- Department of MedicineUniversity of LouisvilleLouisvilleKY
| | - Qing Song
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- The First Affiliated HospitalXi'an Jiaotong UniversityXi'anChina
| | - Fenxia Zhu
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Affiliated Hospital of Integrated Traditional Chinese and Western MedicineNanjing University of Chinese MedicineNanjingChina
| | - Lihua Zhang
- Department of MedicineUniversity of LouisvilleLouisvilleKY
| | - Mengwei Jiang
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKY
| | - Yun Zhou
- Department of MedicineUniversity of LouisvilleLouisvilleKY
| | - Shirish Barve
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKY
- Alcohol Research CenterUniversity of LouisvilleLouisvilleKY
- Hepatobiology & Toxicology CenterUniversity of LouisvilleLouisvilleKY
| | - Xiang Zhang
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKY
- Department of ChemistryUniversity of LouisvilleLouisvilleKY
- Alcohol Research CenterUniversity of LouisvilleLouisvilleKY
- Hepatobiology & Toxicology CenterUniversity of LouisvilleLouisvilleKY
| | - Craig J. McClain
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKY
- Alcohol Research CenterUniversity of LouisvilleLouisvilleKY
- Hepatobiology & Toxicology CenterUniversity of LouisvilleLouisvilleKY
- Robley Rex VA Medical CenterLouisvilleKY
| | - Wenke Feng
- Department of MedicineUniversity of LouisvilleLouisvilleKY
- Department of Pharmacology and ToxicologyUniversity of LouisvilleLouisvilleKY
- Alcohol Research CenterUniversity of LouisvilleLouisvilleKY
- Hepatobiology & Toxicology CenterUniversity of LouisvilleLouisvilleKY
| |
Collapse
|
28
|
Geng W, Long SL, Chang YJ, Saxton AM, Joyce SA, Lin J. Evaluation of bile salt hydrolase inhibitor efficacy for modulating host bile profile and physiology using a chicken model system. Sci Rep 2020; 10:4941. [PMID: 32188876 PMCID: PMC7080769 DOI: 10.1038/s41598-020-61723-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 02/27/2020] [Indexed: 11/24/2022] Open
Abstract
Gut microbial enzymes, bile salt hydrolases (BSHs) are the gateway enzymes for bile acid (BA) modification in the gut. This activity is a promising target for developing innovative non-antibiotic growth promoters to enhance animal production and health. Compelling evidence has shown that inhibition of BSH activity should enhance weight gain by altering the BA pool, host signalling and lipid metabolism. We recently identified a panel of promising BSH inhibitors. Here, we address the potential of them as alternative, effective, non-antibiotic feed additives, for commercial application, to promote animal growth using a chicken model. In this study, the in vivo efficacy of three BSH inhibitors (caffeic acid phenethylester, riboflavin, carnosic acid) were evaluated. 7-day old chicks (10 birds/group) were either untreated or they received one of the specific BSH inhibitors (25 mg/kg body weight) via oral gavage for 17 days. The chicks in treatment groups consistently displayed higher body weight gain than the untreated chicks. Metabolomic analysis demonstrated that BSH inhibitor treatment led to significant changes in both circulating and intestinal BA signatures in support of blunted intestinal BSH activity. Consistent with this finding, liver and intestinal tissue RNA-Seq analysis showed that carnosic acid treatment significantly altered expression of genes involved in lipid and bile acid metabolism. Taken together, this study validates microbial BSH activity inhibition as an alternative target and strategy to antibiotic treatment for animal growth promotion.
Collapse
Affiliation(s)
- Wenjing Geng
- Department of Animal Science, The University of Tennessee, 2506, River Drive, Knoxville, USA
| | - Sarah L Long
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Yun-Juan Chang
- Department of High Performance Computing and Research, University of Rutgers, Newark, USA
| | - Arnold M Saxton
- Department of Animal Science, The University of Tennessee, 2506, River Drive, Knoxville, USA
| | - Susan A Joyce
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| | - Jun Lin
- Department of Animal Science, The University of Tennessee, 2506, River Drive, Knoxville, USA.
| |
Collapse
|
29
|
Bogatyrev SR, Rolando JC, Ismagilov RF. Self-reinoculation with fecal flora changes microbiota density and composition leading to an altered bile-acid profile in the mouse small intestine. MICROBIOME 2020; 8:19. [PMID: 32051033 PMCID: PMC7017497 DOI: 10.1186/s40168-020-0785-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/05/2020] [Indexed: 05/06/2023]
Abstract
BACKGROUND The upper gastrointestinal tract plays a prominent role in human physiology as the primary site for enzymatic digestion and nutrient absorption, immune sampling, and drug uptake. Alterations to the small intestine microbiome have been implicated in various human diseases, such as non-alcoholic steatohepatitis and inflammatory bowel conditions. Yet, the physiological and functional roles of the small intestine microbiota in humans remain poorly characterized because of the complexities associated with its sampling. Rodent models are used extensively in microbiome research and enable the spatial, temporal, compositional, and functional interrogation of the gastrointestinal microbiota and its effects on the host physiology and disease phenotype. Classical, culture-based studies have documented that fecal microbial self-reinoculation (via coprophagy) affects the composition and abundance of microbes in the murine proximal gastrointestinal tract. This pervasive self-reinoculation behavior could be a particularly relevant study factor when investigating small intestine microbiota. Modern microbiome studies either do not take self-reinoculation into account, or assume that approaches such as single housing mice or housing on wire mesh floors eliminate it. These assumptions have not been rigorously tested with modern tools. Here, we used quantitative 16S rRNA gene amplicon sequencing, quantitative microbial functional gene content inference, and metabolomic analyses of bile acids to evaluate the effects of self-reinoculation on microbial loads, composition, and function in the murine upper gastrointestinal tract. RESULTS In coprophagic mice, continuous self-exposure to the fecal flora had substantial quantitative and qualitative effects on the upper gastrointestinal microbiome. These differences in microbial abundance and community composition were associated with an altered profile of the small intestine bile acid pool, and, importantly, could not be inferred from analyzing large intestine or stool samples. Overall, the patterns observed in the small intestine of non-coprophagic mice (reduced total microbial load, low abundance of anaerobic microbiota, and bile acids predominantly in the conjugated form) resemble those typically seen in the human small intestine. CONCLUSIONS Future studies need to take self-reinoculation into account when using mouse models to evaluate gastrointestinal microbial colonization and function in relation to xenobiotic transformation and pharmacokinetics or in the context of physiological states and diseases linked to small intestine microbiome and to small intestine dysbiosis. Video abstract.
Collapse
Affiliation(s)
- Said R Bogatyrev
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Justin C Rolando
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, USA
| | - Rustem F Ismagilov
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Division of Chemistry and Chemical Engineering, California Institute of Technology, 1200 E. California Blvd, Pasadena, CA, USA.
| |
Collapse
|
30
|
Fucoidan and galactooligosaccharides ameliorate high-fat diet–induced dyslipidemia in rats by modulating the gut microbiota and bile acid metabolism. Nutrition 2019; 65:50-59. [PMID: 31029922 DOI: 10.1016/j.nut.2019.03.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 01/23/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
|
31
|
The complex structure of bile salt hydrolase from Lactobacillus salivarius reveals the structural basis of substrate specificity. Sci Rep 2019; 9:12438. [PMID: 31455813 PMCID: PMC6711994 DOI: 10.1038/s41598-019-48850-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 08/09/2019] [Indexed: 01/23/2023] Open
Abstract
The gut bacterial bile salt hydrolase (BSH) plays a critical role in host lipid metabolism and energy harvest. Therefore, BSH is a promising microbiome target to develop new therapies to regulate obesity in humans and novel non-antibiotic growth promoters for food animals. We previously reported the 1.90 Å apo crystal structure of BSH from Lactobacillus salivarius (lsBSH). In this study, we soaked the lsBSH crystal with glycocholic acid (GCA), a substrate, and obtained a 2.10 Å structure containing complex of lsBSH bound to GCA and cholic acid (CA), a product. The substrate/product sits in the water-exposed cavity molded by Loops 2 and 3. While the glycine moiety of GCA is exposed into a highly polar pocket, the sterane core of GCA is stabilized by aromatic and hydrophobic interactions. Comparison of product binding with BSH from Clostridium perfringenes reveals a distinct orientation of the sterane core in the binding site. The stability of the substrate-lsBSH complex and the putative catalytic mechanism were explored with molecular dynamics simulations. Site-directed mutagenesis of lsBSH demonstrated that Cys2 and Asn171 are critical for enzymatic activity, while Tyr24, Phe65 and Gln257 contribute to the substrate specificity. Together, this study provides structural insights into BSH-substrate interaction, the mechanism of catalysis and substrate specificity, which facilitate rational design of BSH inhibitors.
Collapse
|
32
|
Song Z, Cai Y, Lao X, Wang X, Lin X, Cui Y, Kalavagunta PK, Liao J, Jin L, Shang J, Li J. Taxonomic profiling and populational patterns of bacterial bile salt hydrolase (BSH) genes based on worldwide human gut microbiome. MICROBIOME 2019; 7:9. [PMID: 30674356 PMCID: PMC6345003 DOI: 10.1186/s40168-019-0628-3] [Citation(s) in RCA: 277] [Impact Index Per Article: 46.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/16/2019] [Indexed: 05/07/2023]
Abstract
BACKGROUND Bile salt hydrolase plays an important role in bile acid-mediated signaling pathways, which regulate lipid absorption, glucose metabolism, and energy homeostasis. Several reports suggest that changes in the composition of bile acids are found in many diseases caused by dysbacteriosis. RESULTS Here, we present the taxonomic identification of bile salt hydrolase (BSH) in human microbiota and elucidate the abundance and activity differences of various bacterial BSH among 11 different populations from six continents. For the first time, we revealed that bile salt hydrolase protein sequences (BSHs) are distributed in 591 intestinal bacterial strains within 117 genera in human microbiota, and 27.52% of these bacterial strains containing BSH paralogs. Significant variations are observed in BSH distribution patterns among different populations. Based on phylogenetic analysis, we reclassified these BSHs into eight phylotypes and investigated the abundance patterns of these phylotypes among different populations. From the inspection of enzyme activity among different BSH phylotypes, BSH-T3 showed the highest enzyme activity and is only found in Lactobaclillus. The phylotypes of BSH-T5 and BSH-T6 mainly from Bacteroides with high percentage of paralogs exhibit different enzyme activity and deconjugation activity. Furthermore, we found that there were significant differences between healthy individuals and patients with atherosclerosis and diabetes in some phylotypes of BSHs though the correlations were pleiotropic. CONCLUSION This study revealed the taxonomic and abundance profiling of BSH in human gut microbiome and provided a phylogenetic-based system to assess BSHs activity by classifying the target sequence into specific phylotype. Furthermore, the present work disclosed the variation patterns of BSHs among different populations of geographical regions and health/disease cohorts, which is essential to understand the role of BSH in the development and progression of related diseases.
Collapse
Affiliation(s)
- Ziwei Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
| | - Yuanyuan Cai
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009 China
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
| | - Xue Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
| | - Xiaoxuan Lin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
| | - Yingyun Cui
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
| | | | - Jun Liao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009 China
| | - Liang Jin
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009 China
| | - Jing Shang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009 China
| | - Jing Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009 China
| |
Collapse
|
33
|
A new method for the in vitro determination of the bile tolerance of potentially probiotic lactobacilli. Appl Microbiol Biotechnol 2018; 102:1903-1910. [DOI: 10.1007/s00253-018-8742-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 12/21/2017] [Accepted: 12/26/2017] [Indexed: 12/22/2022]
|
34
|
DiMarzio M, Rusconi B, Yennawar NH, Eppinger M, Patterson AD, Dudley EG. Identification of a mouse Lactobacillus johnsonii strain with deconjugase activity against the FXR antagonist T-β-MCA. PLoS One 2017; 12:e0183564. [PMID: 28910295 PMCID: PMC5598929 DOI: 10.1371/journal.pone.0183564] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 08/07/2017] [Indexed: 12/13/2022] Open
Abstract
Bile salt hydrolase (BSH) activity against the bile acid tauro-beta-muricholic acid (T-β-MCA) was recently reported to mediate host bile acid, glucose, and lipid homeostasis via the farnesoid X receptor (FXR) signaling pathway. An earlier study correlated decreased Lactobacillus abundance in the cecum with increased concentrations of intestinal T-β-MCA, an FXR antagonist. While several studies have characterized BSHs in lactobacilli, deconjugation of T-β-MCA remains poorly characterized among members of this genus, and therefore it was unclear what strain(s) were responsible for this activity. Here, a strain of L. johnsonii with robust BSH activity against T-β-MCA in vitro was isolated from the cecum of a C57BL/6J mouse. A screening assay performed on a collection of 14 Lactobacillus strains from nine different species identified BSH substrate specificity for T-β-MCA only in two of three L. johnsonii strains. Genomic analysis of the two strains with this BSH activity revealed the presence of three bsh genes that are homologous to bsh genes in the previously sequenced human-associated strain L. johnsonii NCC533. Heterologous expression of several bsh genes in E. coli followed by enzymatic assays revealed broad differences in substrate specificity even among closely related bsh homologs, and suggests that the phylogeny of these enzymes does not closely correlate with substrate specificity. Predictive modeling allowed us to propose a potential mechanism driving differences in BSH activity for T-β-MCA in these homologs. Our data suggests that L. johnsonii regulates T-β-MCA levels in the mouse intestinal environment, and that this species may play a central role in FXR signaling in the mouse.
Collapse
Affiliation(s)
- Michael DiMarzio
- Department of Food Science, The Pennsylvania State University, University Park, PA, United States of America
| | - Brigida Rusconi
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States of America
| | - Neela H. Yennawar
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, United States of America
| | - Mark Eppinger
- Department of Biology and South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, United States of America
| | - Andrew D. Patterson
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA, United States of America
| | - Edward G. Dudley
- Department of Food Science, The Pennsylvania State University, University Park, PA, United States of America
- * E-mail:
| |
Collapse
|
35
|
Novoa-Garrido M, Rebours C, Aanensen L, Torp T, Lind V, Steinshamn H. Effect of seaweed on gastrointestinal microbiota isolated from Norwegian White sheep. ACTA AGR SCAND A-AN 2017. [DOI: 10.1080/09064702.2017.1310287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- M. Novoa-Garrido
- Faculty of Biosciences and Aquaculture, Nord University, Bodø, Norway
- NIBIO – Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - C. Rebours
- NIBIO – Norwegian Institute of Bioeconomy Research, Ås, Norway
- Møreforsking Ålesund AS, Ålesund, Norway
| | - L. Aanensen
- NIBIO – Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - T. Torp
- NIBIO – Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - V. Lind
- NIBIO – Norwegian Institute of Bioeconomy Research, Ås, Norway
| | - H. Steinshamn
- NIBIO – Norwegian Institute of Bioeconomy Research, Ås, Norway
| |
Collapse
|