1
|
den Bakker E, Smith DEC, Finken MJJ, Wamelink MMC, Salomons GS, van de Kamp JM, Bökenkamp A. Sulfate: a neglected (but potentially highly relevant) anion. Essays Biochem 2024; 68:391-399. [PMID: 38639060 DOI: 10.1042/ebc20230097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/20/2024]
Abstract
Sulfate is an important anion as sulfonation is essential in modulation of several compounds, such as exogens, polysaccharide chains of proteoglycans, cholesterol or cholesterol derivatives and tyrosine residues of several proteins. Sulfonation requires the presence of both the sulfate donor 3'-phosphoadenosine-5'-phosphosulfate (PAPS) and a sulfotransferase. Genetic disorders affecting sulfonation, associated with skeletal abnormalities, impaired neurological development and endocrinopathies, demonstrate the importance of sulfate. Yet sulfate is not measured in clinical practice. This review addresses sulfate metabolism and consequences of sulfonation defects, how to measure sulfate and why we should measure sulfate more often.
Collapse
Affiliation(s)
- Emil den Bakker
- Department of Pediatric Nephrology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | - Desiree E C Smith
- Department of Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
| | - Martijn J J Finken
- Department of Pediatric Endocrinology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| | | | - Gajja S Salomons
- Department of Metabolic Diseases, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, the Netherlands
| | - Jiddeke M van de Kamp
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam UMC, Amsterdam, the Netherlands
- Amsterdam Reproduction and Development, Amsterdam UMC, Amsterdam, the Netherlands
- Department of Human Genetics, Amsterdam UMC, Amsterdam, the Netherlands
| | - Arend Bökenkamp
- Department of Pediatric Nephrology, Emma Childrens Hospital, Amsterdam UMC, Amsterdam, the Netherlands
| |
Collapse
|
2
|
Chen X, Zhang Y, Yin J, Liu C, Xie M, Wang Y, Chen M, Zhang R, Yuan X, Li D, Chen X, Gao X, Cai G, Zhang S, Zhou B, Yang M. Structural basis for the reaction cycle and transport mechanism of human Na +-sulfate cotransporter NaS1 (SLC13A1). SCIENCE ADVANCES 2024; 10:eado6778. [PMID: 39576865 PMCID: PMC11584011 DOI: 10.1126/sciadv.ado6778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Sulfate (SO42-) is a pivotal inorganic anion with essential roles in mammalian physiology. NaS1, a member of solute carrier 13 family and divalent anion/sodium symporter family, functions as a Na+-sulfate cotransporter, facilitating sulfate (re)absorption across renal proximal tubule and small intestine epithelia. While previous studies have linked several human disorders to mutations in the NaS1 gene, its transport mechanism remains unclear. Here, we report the cryo-electron microscopy structures of five distinct conformations of the human NaS1 at resolutions of 2.7 to 3.3 angstroms, revealing the substrates recognition mechanism and the conformational change of NaS1 during the Na+-sulfate cotransport cycle. Our studies delineate the molecular basis of the detailed dynamic transport cycle of NaS1. These findings advance the current understanding of the Na+-sulfate cotransport mechanism, human sulfate (re)absorption, and the implications of disease-associated NaS1 mutations.
Collapse
Affiliation(s)
- Xudong Chen
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Youqi Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Jian Yin
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Chang Liu
- Beijing Life Science Academy, Beijing 102209, China
| | - Min Xie
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yixue Wang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Meiying Chen
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Rui Zhang
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xinyi Yuan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - De Li
- Radioisotope Laboratory of Center of Biomedical Analysis, Tsinghua University, Beijing 100084, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Xin Gao
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Computational Bioscience Research Center, King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing 100853, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Boda Zhou
- Department of Cardiology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Tsinghua-Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Cryo-EM Facility Center, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
3
|
Luo W, Zhao M, Dwidar M, Gao Y, Xiang L, Wu X, Medema MH, Xu S, Li X, Schäfer H, Chen M, Feng R, Zhu Y. Microbial assimilatory sulfate reduction-mediated H 2S: an overlooked role in Crohn's disease development. MICROBIOME 2024; 12:152. [PMID: 39152482 PMCID: PMC11328384 DOI: 10.1186/s40168-024-01873-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/13/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND H2S imbalances in the intestinal tract trigger Crohn's disease (CD), a chronic inflammatory gastrointestinal disorder characterized by microbiota dysbiosis and barrier dysfunction. However, a comprehensive understanding of H2S generation in the gut, and the contributions of both microbiota and host to systemic H2S levels in CD, remain to be elucidated. This investigation aimed to enhance comprehension regarding the sulfidogenic potential of both the human host and the gut microbiota. RESULTS Our analysis of a treatment-naive CD cohorts' fecal metagenomic and biopsy metatranscriptomic data revealed reduced expression of host endogenous H2S generation genes alongside increased abundance of microbial exogenous H2S production genes in correlation with CD. While prior studies focused on microbial H2S production via dissimilatory sulfite reductases, our metagenomic analysis suggests the assimilatory sulfate reduction (ASR) pathway is a more significant contributor in the human gut, given its high prevalence and abundance. Subsequently, we validated our hypothesis experimentally by generating ASR-deficient E. coli mutants ∆cysJ and ∆cysM through the deletion of sulfite reductase and L-cysteine synthase genes. This alteration significantly affected bacterial sulfidogenic capacity, colon epithelial cell viability, and colonic mucin sulfation, ultimately leading to colitis in murine model. Further study revealed that gut microbiota degrade sulfopolysaccharides and assimilate sulfate to produce H2S via the ASR pathway, highlighting the role of sulfopolysaccharides in colitis and cautioning against their use as food additives. CONCLUSIONS Our study significantly advances understanding of microbial sulfur metabolism in the human gut, elucidating the complex interplay between diet, gut microbiota, and host sulfur metabolism. We highlight the microbial ASR pathway as an overlooked endogenous H2S producer and a potential therapeutic target for managing CD. Video Abstract.
Collapse
Affiliation(s)
- Wanrong Luo
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Min Zhao
- Department of Gastroenterology, Shenzhen No.3 People's Hospital, Shenzhen, Guangdong, China
| | - Mohammed Dwidar
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Cleveland Clinic, Cleveland, OH, USA
| | - Yang Gao
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Liyuan Xiang
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China
| | - Xueting Wu
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China
| | - Marnix H Medema
- Bioinformatics Group, Wageningen University, Wageningen, The Netherlands
| | - Shu Xu
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China
| | - Xiaozhi Li
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China
| | - Hendrik Schäfer
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Minhu Chen
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China.
| | - Rui Feng
- Department of Gastroenterology, the First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Er Road, Room 1209, Guangzhou, 510080, China.
| | - Yijun Zhu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Sun HJ, Lu QB, Zhu XX, Ni ZR, Su JB, Fu X, Chen G, Zheng GL, Nie XW, Bian JS. Pharmacology of Hydrogen Sulfide and Its Donors in Cardiometabolic Diseases. Pharmacol Rev 2024; 76:846-895. [PMID: 38866561 DOI: 10.1124/pharmrev.123.000928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/13/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024] Open
Abstract
Cardiometabolic diseases (CMDs) are major contributors to global mortality, emphasizing the critical need for novel therapeutic interventions. Hydrogen sulfide (H2S) has garnered enormous attention as a significant gasotransmitter with various physiological, pathophysiological, and pharmacological impacts within mammalian cardiometabolic systems. In addition to its roles in attenuating oxidative stress and inflammatory response, burgeoning research emphasizes the significance of H2S in regulating proteins via persulfidation, a well known modification intricately associated with the pathogenesis of CMDs. This review seeks to investigate recent updates on the physiological actions of endogenous H2S and the pharmacological roles of various H2S donors in addressing diverse aspects of CMDs across cellular, animal, and clinical studies. Of note, advanced methodologies, including multiomics, intestinal microflora analysis, organoid, and single-cell sequencing techniques, are gaining traction due to their ability to offer comprehensive insights into biomedical research. These emerging approaches hold promise in characterizing the pharmacological roles of H2S in health and diseases. We will critically assess the current literature to clarify the roles of H2S in diseases while also delineating the opportunities and challenges they present in H2S-based pharmacotherapy for CMDs. SIGNIFICANCE STATEMENT: This comprehensive review covers recent developments in H2S biology and pharmacology in cardiometabolic diseases CMDs. Endogenous H2S and its donors show great promise for the management of CMDs by regulating numerous proteins and signaling pathways. The emergence of new technologies will considerably advance the pharmacological research and clinical translation of H2S.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Qing-Bo Lu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xue-Xue Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Zhang-Rong Ni
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Jia-Bao Su
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xiao Fu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Guo Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Guan-Li Zheng
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Xiao-Wei Nie
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| | - Jin-Song Bian
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China (H.-J.S., X.-X.Z., Z.-R.N., J.-B.S., X.F., G.C., G.-L.Z.); Department of Endocrinology, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, Jiangsu, China (Q.-B.L.); Shenzhen Key Laboratory of Respiratory Diseases, Shenzhen People's Hospital, Shenzhen, Guangdong, China (X.-W.N.); and Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, China (J.-S.B.)
| |
Collapse
|
5
|
Fricker AD, Yao T, Lindemann SR, Flores GE. Enrichment and characterization of human-associated mucin-degrading microbial consortia by sequential passage. FEMS Microbiol Ecol 2024; 100:fiae078. [PMID: 38794902 PMCID: PMC11180985 DOI: 10.1093/femsec/fiae078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 05/03/2024] [Accepted: 05/23/2024] [Indexed: 05/26/2024] Open
Abstract
Mucin is a glycoprotein secreted throughout the mammalian gastrointestinal tract that can support endogenous microorganisms in the absence of complex polysaccharides. While several mucin-degrading bacteria have been identified, the interindividual differences in microbial communities capable of metabolizing this complex polymer are not well described. To determine whether community assembly on mucin is deterministic across individuals or whether taxonomically distinct but functionally similar mucin-degrading communities are selected across fecal inocula, we used a 10-day in vitro sequential batch culture fermentation from three human donors with mucin as the sole carbon source. For each donor, 16S rRNA gene amplicon sequencing was used to characterize microbial community succession, and the short-chain fatty acid profile was determined from the final community. All three communities reached a steady-state by day 7 in which the community composition stabilized. Taxonomic comparisons amongst communities revealed that one of the final communities had Desulfovibrio, another had Akkermansia, and all three shared other members, such as Bacteroides. Metabolic output differences were most notable for one of the donor's communities, with significantly less production of acetate and propionate than the other two communities. These findings demonstrate the feasibility of developing stable mucin-degrading communities with shared and unique taxa. Furthermore, the mechanisms and efficiencies of mucin degradation across individuals are important for understanding how this community-level process impacts human health.
Collapse
Affiliation(s)
- Ashwana D Fricker
- Department of Biology, California State University, 18111 Nordhoff Street, Northridge, CA 91330, United States
| | - Tianming Yao
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, 745 Agriculture Mall Drive, West Lafayette, IN 47907, United States
| | - Stephen R Lindemann
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, 745 Agriculture Mall Drive, West Lafayette, IN 47907, United States
| | - Gilberto E Flores
- Department of Biology, California State University, 18111 Nordhoff Street, Northridge, CA 91330, United States
| |
Collapse
|
6
|
Wang X, Wang Z, Su S, Wu Y, Fan J, Hou X, Zhang K, Salama ES, Kulshrestha S, Ling Z, Liu P, Li X. Probiotics Pediococcus acidilactici GR-1 promotes the functional strains and remodels gut microbiota to reduce the Cr(VI) toxicity in a dual-chamber simulated intestinal system. CHEMOSPHERE 2024; 356:141927. [PMID: 38593954 DOI: 10.1016/j.chemosphere.2024.141927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 02/29/2024] [Accepted: 04/05/2024] [Indexed: 04/11/2024]
Abstract
Numerous animal studies have demonstrated the toxicity of hexavalent chromium [Cr(VI)] and the bioremediative effects of probiotics on the composition and functions of gut microbiota. Since the precise mechanisms of Cr(VI) detoxification and its interactions with human gut microbiota were unknown, a novel dual-chamber simulated intestinal (DCSI) system was developed to maintain both the stability of the simulated system and the composition of the gut microbiota. Probiotic GR-1 was found to regulate intestinal gut microbiota, thereby reducing the toxicity of Cr(VI) within the DCSI system. The results indicate that Cr(VI) levels were reduced from 2.260 ± 0.2438 μg/g to 1.7086 ± 0.1950 μg/g in the gut microbiota cell pellet, and Cr(VI) permeability decreased from 0.5521 ± 0.1132 μg/L to 0.3681 ± 0.0178 μg/L after 48 h in simulated gut fluid. Additionally, the removal rate of 1,1-Diphenyl-2-picrylhydrazyl (DPPH), reducibility (Vitamin C), and total antioxidant capacity (T-AOC) increased by 50.83%, 31.70%, and 27.56%, respectively, following probiotic treatment. The increase in antioxidant capacity correlated with total Cr removal (P < 0.05, r from -0.80 to 0.73). 16S rRNA sequencing analysis showed that gut microbiota composition was reshaped by the addition of probiotics, which regulated the recovery of the functional gut microbiota to normal levels, rather than restoring the entire gut microbiota composition for community function. Thus, this study not only demonstrates the feasibility and stability of culturing gut microbiota but also offers a new biotechnological approach to synthesizing functional communities with functional strains for environmental risk management.
Collapse
Affiliation(s)
- Xing Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Zemin Wang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Shaochen Su
- Healthy Examination & Management Center, First Hospital of Lanzhou University, Lanzhou, 730000, PR China.
| | - Ying Wu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Jingjing Fan
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Xiaoxiao Hou
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Kunyue Zhang
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - El-Sayed Salama
- Department of Occupational and Environmental Health, School of Public Health, Lanzhou University, Lanzhou University, Lanzhou, 730000, Gansu Province, PR China.
| | - Saurabh Kulshrestha
- Faculty of Applied Sciences and Biotechnology, Biotechnology and Management Sciences, Shoolini University, Bajhol, Solan, Himachal Pradesh, 173229, India.
| | - Zhenmin Ling
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Pu Liu
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| | - Xiangkai Li
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environment Pollution, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China; Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, PR China.
| |
Collapse
|
7
|
Stummer N, Feichtinger RG, Weghuber D, Kofler B, Schneider AM. Role of Hydrogen Sulfide in Inflammatory Bowel Disease. Antioxidants (Basel) 2023; 12:1570. [PMID: 37627565 PMCID: PMC10452036 DOI: 10.3390/antiox12081570] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 07/28/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Hydrogen sulfide (H2S), originally known as toxic gas, has now attracted attention as one of the gasotransmitters involved in many reactions in the human body. H2S has been assumed to play a role in the pathogenesis of many chronic diseases, of which the exact pathogenesis remains unknown. One of them is inflammatory bowel disease (IBD), a chronic intestinal disease subclassified as Crohn's disease (CD) and ulcerative colitis (UC). Any change in the amount of H2S seems to be linked to inflammation in this illness. These changes can be brought about by alterations in the microbiota, in the endogenous metabolism of H2S and in the diet. As both too little and too much H2S drive inflammation, a balanced level is needed for intestinal health. The aim of this review is to summarize the available literature published until June 2023 in order to provide an overview of the current knowledge of the connection between H2S and IBD.
Collapse
Affiliation(s)
- Nathalie Stummer
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (N.S.); (R.G.F.); (D.W.); (B.K.)
| | - René G. Feichtinger
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (N.S.); (R.G.F.); (D.W.); (B.K.)
| | - Daniel Weghuber
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (N.S.); (R.G.F.); (D.W.); (B.K.)
| | - Barbara Kofler
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (N.S.); (R.G.F.); (D.W.); (B.K.)
- Research Program for Receptor Biochemistry and Tumor Metabolism, Paracelsus Medical University (PMU), 5020 Salzburg, Austria
| | - Anna M. Schneider
- Department of Pediatrics, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria; (N.S.); (R.G.F.); (D.W.); (B.K.)
| |
Collapse
|
8
|
Egbueri JC. A multi-model study for understanding the contamination mechanisms, toxicity and health risks of hardness, sulfate, and nitrate in natural water resources. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:61626-61658. [PMID: 36928703 DOI: 10.1007/s11356-023-26396-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 03/07/2023] [Indexed: 05/10/2023]
Abstract
Several water quality contaminants have attracted the attention of numerous researchers globally, in recent times. Although the toxicity and health risk assessments of sulfate and water hardness have not received obvious attention, nitrate contamination has gained peculiar research interest globally. In the present paper, multiple data-driven indexical, graphical, and soft computational models were integrated for a detailed assessment and predictive modeling of the contamination mechanisms, toxicity, and human health risks of natural waters in Southeast Nigeria. Majority of the tested physicochemical parameters were within their satisfactory limits for drinking and other purposes. However, total hardness (TH), SO4, and NO3 were above stipulated limits in some locations. A nitrate health risk assessment revealed that certain areas present a chronic health risk to children, females, and males due to water intake. However, the dermal absorption route was found to have negligible health risks. SO4 in some locations was above the 100 mg/L Nigerian limit; thus, heightening the potential health effects due to intake of the contaminated water resources. Most samples had low TH values, which exposes users to health defects. There are mixed contamination mechanisms in the area, according to graphical plots, R-mode hierarchical dendrogram, factor analysis, and stoichiometry. However, geogenic mechanisms predominate over human-related mechanisms. Based on the results, a composite diagrammatic model was developed. Furthermore, predictive radial basis function (RBF) and multiple linear regression (MLR) models accurately predicted the TH, SO4, and NO3, with the RBF outperforming the MLR models. Insights from the RBF and MLR models were useful in validating the results of the hierarchical dendrogram, factor, stoichiometric, and graphical analyses.
Collapse
Affiliation(s)
- Johnbosco C Egbueri
- Department of Geology, Chukwuemeka Odumegwu Ojukwu University, Uli, Anambra State, Nigeria.
| |
Collapse
|
9
|
Yao CK, Sarbagili-Shabat C. Gaseous metabolites as therapeutic targets in ulcerative colitis. World J Gastroenterol 2023; 29:682-691. [PMID: 36742165 PMCID: PMC9896612 DOI: 10.3748/wjg.v29.i4.682] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/19/2022] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
Diet therapies are currently under-utilised in optimising clinical outcomes for patients with active ulcerative colitis (UC). Furthermore, existing dietary therapies are framed by poorly defined mechanistic targets to warrant its success. There is good evidence to suggest that microbial production of gaseous metabolites, hydrogen sulfide (H2S) and nitric oxide (NO) are implicated in the development of mucosal inflammation in UC. On a cellular level, exposure of the colonic epithelium to excessive concentrations of these gases are shown to promote functional defects described in UC. Hence, targeting bacterial production of these gases could provide an opportunity to formulate new dietary therapies in UC. Despite the paucity of evidence, there is epidemiological and clinical data to support the concept of reducing mucosal inflammation in UC via dietary strategies that reduce H2S. Several dietary components, namely sulphur-containing amino acids and inorganic sulphur have been shown to be influential in enhancing colonic H2S production. More recent data suggests increasing the supply of readily fermentable fibre as an effective strategy for H2S reduction. Conversely, very little is known regarding how diet alters microbial production of NO. Hence, the current evidence suggest that a whole diet approach is needed. Finally, biomarkers for assessing changes in microbial gaseous metabolites in response to dietary interventions are very much required. In conclusion, this review identifies a great need for high quality randomised-controlled trials to demonstrate the efficacy of a sulphide-reducing dietary therapy for patients with active UC.
Collapse
Affiliation(s)
- Chu K Yao
- Department of Gastroenterology, Monash University, Melbourne 3004, Australia
| | - Chen Sarbagili-Shabat
- Pediatric Gastroenterology Unit, PIBD Research Center, Wolfson Medical Center, Holon 5822012, Israel
| |
Collapse
|
10
|
Daniel H. Gut physiology meets microbiome science. GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 4:e1. [PMID: 39295899 PMCID: PMC11406389 DOI: 10.1017/gmb.2022.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/13/2022] [Accepted: 10/18/2022] [Indexed: 09/21/2024]
Abstract
Research on the gut microbiome has gained high popularity and almost every disease has meanwhile been linked to alterations in microbiome composition. Typically assessed via stool samples, the microbiome displays a huge diversity with a multitude of environmental parameters already identified as contributing to its character. Despite impressive scientific progress, normal microbiome diversity remains largely unexplained and it is tempting to speculate some of the yet unexplained variance is hidden in normal gut physiology. Although a few genome/phenome-wide associations studies have recently highlighted physiological parameters such as stool frequency, known as contributing to microbiome diversity, there is a large knowledge base from decades of basic research on gut functions that can be explored for possible links to stool features and microbiome characteristics. And, when extrapolating findings from faecal samples to the biology in the intestinal lumen or the mucosal microenvironment, gut anatomy and physiology features need to be considered. Similarly, differences in anatomy and physiology between rodents and humans need attention when discussing findings in animals in relation to human physiology and nutrition.
Collapse
Affiliation(s)
- Hannelore Daniel
- ex. School of Life Sciences, Technical University of Munich, Gregor-Mendel-Strasse 2, 85354 Freising, Germany
| |
Collapse
|
11
|
Kettle H, Louis P, Flint HJ. Process-based modelling of microbial community dynamics in the human colon. J R Soc Interface 2022; 19:20220489. [PMCID: PMC9554726 DOI: 10.1098/rsif.2022.0489] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/16/2022] [Indexed: 12/15/2024] Open
Abstract
The human colon contains a dynamic microbial community whose composition has important implications for human health. In this work, we build a process-based model of the colonic microbial ecosystem and compare with general empirical observations and the results of in vivo experiments. Our model comprises a complex microbial ecosystem along with absorption of short chain fatty acids (SCFA) and water by the host through the gut wall, variations in incoming dietary substrates (in the form of ‘meals’ whose composition varies in time), bowel movements, feedback on microbial growth from changes in pH resulting from SCFA production and multiple compartments to represent the proximal, transverse and distal colon. We verify our model against a number of observed criteria, e.g. total SCFA concentrations, SCFA ratios, mass of bowel movements, pH and water absorption over the transit time; and then run simulations investigating the effect of colonic transit time, and the composition and amount of indigestible carbohydrate in the host diet, which we compare with in vivo studies. The code is available as an R package (microPopGut) to aid future research.
Collapse
Affiliation(s)
- Helen Kettle
- Biomathematics and Statistics Scotland, James Clerk Maxwell Building, Peter Guthrie Tait Road, Edinburgh EH9 3FD, UK
| | - Petra Louis
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Harry J. Flint
- Gut Health Group, Rowett Institute, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
12
|
Duszka K. Versatile Triad Alliance: Bile Acid, Taurine and Microbiota. Cells 2022; 11:2337. [PMID: 35954180 PMCID: PMC9367564 DOI: 10.3390/cells11152337] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/21/2022] Open
Abstract
Taurine is the most abundant free amino acid in the body, and is mainly derived from the diet, but can also be produced endogenously from cysteine. It plays multiple essential roles in the body, including development, energy production, osmoregulation, prevention of oxidative stress, and inflammation. Taurine is also crucial as a molecule used to conjugate bile acids (BAs). In the gastrointestinal tract, BAs deconjugation by enteric bacteria results in high levels of unconjugated BAs and free taurine. Depending on conjugation status and other bacterial modifications, BAs constitute a pool of related but highly diverse molecules, each with different properties concerning solubility and toxicity, capacity to activate or inhibit receptors of BAs, and direct and indirect impact on microbiota and the host, whereas free taurine has a largely protective impact on the host, serves as a source of energy for microbiota, regulates bacterial colonization and defends from pathogens. Several remarkable examples of the interaction between taurine and gut microbiota have recently been described. This review will introduce the necessary background information and lay out the latest discoveries in the interaction of the co-reliant triad of BAs, taurine, and microbiota.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
13
|
Day AS, Yao CK, Costello SP, Ruszkiewicz A, Andrews JM, Gibson PR, Bryant RV. Therapeutic Potential of the 4 Strategies to SUlfide-REduction (4-SURE) Diet in Adults with Mild to Moderately Active Ulcerative Colitis: An Open-Label Feasibility Study. J Nutr 2022; 152:1690-1701. [PMID: 35451489 DOI: 10.1093/jn/nxac093] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 01/15/2022] [Accepted: 04/19/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diet therapy may bridge the therapeutic gap in ulcerative colitis (UC). OBJECTIVES The novel 4-SURE diet (4-strategies-to-SUlfide-REduction), designed to modulate colonic fermentation and influence production of excess hydrogen sulfide, was examined in a feasibility study for tolerability, clinical efficacy, and effects on microbial endpoints. METHODS Adults aged ≥18 y old with mild to moderately active UC were advised to increase intake of fermentable fibers, restrict total and sulfur-containing proteins, and avoid specific food additives for 8 wk. The primary outcome was tolerability of diet [100-mm visual analogue scale (VAS) with 100-mm being intolerable]. Secondary exploratory outcomes were self-reported adherence (always adherent ≥76-100%), clinical and endoscopic response (reduction in partial Mayo ≥2 and Mayo endoscopic subscore ≥1), modulation of fecal characteristics including markers of protein and carbohydrate fermentation, and food-related quality of life (IBD-FRQoL-29). Primary analysis was by intention to treat, performed using paired t and Wilcoxon signed-rank statistical tests. RESULTS Twenty-eight adults with UC [mean (range) age: 42 (22-72) y, 15 females, 3 proctitis, 14 left-sided, and 11 extensive] were studied. Prescribed dietary targets were achieved overall. The diet was well tolerated (VAS: 19 mm; 95% CI: 7, 31 mm) with 95% frequently or always adherent. Clinical response occurred in 13 of 28 (46%) and endoscopic improvement in 10 of 28 participants (36%). Two participants (7%) worsened. Fecal excretion of SCFAs increased by 69% (P < 0.0001), whereas the proportion of branched-chain fatty acids to SCFAs was suppressed by 27% (-1.34%; 95% CI: -2.28%, -0.40%; P = 0.007). The FRQoL improved by 10 points (95% CI: 4, 16; P < 0.001). CONCLUSIONS The 4-SURE dietary strategy is considered tolerable and an acceptable diet by adults with mild to moderately active UC. The dietary teachings achieved the prescribed dietary and fecal targets. Given signals of therapeutic efficacy, further evaluation of this diet is warranted in a placebo-controlled trial. This trial was registered at https://www.anzctr.org.au (Australian New Zealand Clinical Trials Registry) as ACTRN12619000063112.
Collapse
Affiliation(s)
- Alice S Day
- Inflammatory Bowel Disease Services, Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Basil Hetzel Research Institute, Woodville, Australia
| | - Chu Kion Yao
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Samuel P Costello
- Inflammatory Bowel Disease Services, Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Basil Hetzel Research Institute, Woodville, Australia
| | - Andrew Ruszkiewicz
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Gastroenterology Research Laboratory, SA Pathology, Adelaide, Australia
| | - Jane M Andrews
- School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Inflammatory Bowel Disease Service, Department of Gastroenterology and Hepatology, Royal Adelaide Hospital, Adelaide, Australia
| | - Peter R Gibson
- Department of Gastroenterology, Monash University and Alfred Health, Melbourne, Australia
| | - Robert V Bryant
- Inflammatory Bowel Disease Services, Department of Gastroenterology and Hepatology, The Queen Elizabeth Hospital, Woodville, Australia.,School of Medicine, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia.,Basil Hetzel Research Institute, Woodville, Australia
| |
Collapse
|
14
|
Wolf PG, Cowley ES, Breister A, Matatov S, Lucio L, Polak P, Ridlon JM, Gaskins HR, Anantharaman K. Diversity and distribution of sulfur metabolic genes in the human gut microbiome and their association with colorectal cancer. MICROBIOME 2022; 10:64. [PMID: 35440042 PMCID: PMC9016944 DOI: 10.1186/s40168-022-01242-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 02/01/2022] [Indexed: 05/05/2023]
Abstract
BACKGROUND Recent evidence implicates microbial sulfidogenesis as a potential trigger of colorectal cancer (CRC), highlighting the need for comprehensive knowledge of sulfur metabolism within the human gut. Microbial sulfidogenesis produces genotoxic hydrogen sulfide (H2S) in the human colon using inorganic (sulfate) and organic (taurine/cysteine/methionine) substrates; however, the majority of studies have focused on sulfate reduction using dissimilatory sulfite reductases (Dsr). RESULTS Here, we show that genes for microbial sulfur metabolism are more abundant and diverse than previously observed and are statistically associated with CRC. Using ~ 17,000 bacterial genomes from publicly available stool metagenomes, we studied the diversity of sulfur metabolic genes in 667 participants across different health statuses: healthy, adenoma, and carcinoma. Sulfidogenic genes were harbored by 142 bacterial genera and both organic and inorganic sulfidogenic genes were associated with carcinoma. Significantly, the anaerobic sulfite reductase (asr) genes were twice as abundant as dsr, demonstrating that Asr is likely a more important contributor to sulfate reduction in the human gut than Dsr. We identified twelve potential pathways for reductive taurine metabolism and discovered novel genera harboring these pathways. Finally, the prevalence of metabolic genes for organic sulfur indicates that these understudied substrates may be the most abundant source of microbially derived H2S. CONCLUSIONS Our findings significantly expand knowledge of microbial sulfur metabolism in the human gut. We show that genes for microbial sulfur metabolism in the human gut are more prevalent than previously known, irrespective of health status (i.e., in both healthy and diseased states). Our results significantly increase the diversity of pathways and bacteria that are associated with microbial sulfur metabolism in the human gut. Overall, our results have implications for understanding the role of the human gut microbiome and its potential contributions to the pathogenesis of CRC. Video abstract.
Collapse
Affiliation(s)
- Patricia G Wolf
- Institute for Health Research and Policy, University of Illinois at Chicago, Chicago, IL, USA
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Elise S Cowley
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Adam Breister
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Sarah Matatov
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Luke Lucio
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Paige Polak
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Jason M Ridlon
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - H Rex Gaskins
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Biomedical and Translational Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Pathobiology, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| | | |
Collapse
|
15
|
Aroca A, Gotor C. Hydrogen Sulfide: A Key Role in Autophagy Regulation from Plants to Mammalians. Antioxidants (Basel) 2022; 11:327. [PMID: 35204209 PMCID: PMC8868472 DOI: 10.3390/antiox11020327] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/27/2022] [Accepted: 02/06/2022] [Indexed: 02/01/2023] Open
Abstract
Autophagy is a degradative conserved process in eukaryotes to recycle unwanted cellular protein aggregates and damaged organelles. Autophagy plays an important role under normal physiological conditions in multiple biological processes, but it is induced under cellular stress. Therefore, it needs to be tightly regulated to respond to different cellular stimuli. In this review, the regulation of autophagy by hydrogen sulfide is described in both animal and plant systems. The underlying mechanism of action of sulfide is deciphered as the persulfidation of specific targets, regulating the pro- or anti-autophagic role of sulfide with a cell survival outcome. This review aims to highlight the importance of sulfide and persulfidation in autophagy regulation comparing the knowledge available in mammals and plants.
Collapse
Affiliation(s)
- Angeles Aroca
- Institute of Plant Biochemistry and Photosynthesis, University of Seville and CSIC, 41092 Seville, Spain;
| | | |
Collapse
|
16
|
Sikavi DR, Nguyen LH, Haruki K, Ugai T, Ma W, Wang DD, Thompson KN, Yan Y, Branck T, Wilkinson JE, Akimoto N, Zhong R, Lau MC, Mima K, Kosumi K, Morikawa T, Rimm EB, Garrett WS, Izard J, Cao Y, Song M, Huttenhower C, Ogino S, Chan AT. The Sulfur Microbial Diet and Risk of Colorectal Cancer by Molecular Subtypes and Intratumoral Microbial Species in Adult Men. Clin Transl Gastroenterol 2021; 12:e00338. [PMID: 34333506 PMCID: PMC8323793 DOI: 10.14309/ctg.0000000000000338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/05/2021] [Indexed: 01/22/2023] Open
Abstract
INTRODUCTION We recently described the sulfur microbial diet, a pattern of intake associated with increased gut sulfur-metabolizing bacteria and incidence of distal colorectal cancer (CRC). We assessed whether this risk differed by CRC molecular subtypes or presence of intratumoral microbes involved in CRC pathogenesis (Fusobacterium nucleatum and Bifidobacterium spp.). METHODS We performed Cox proportional hazards modeling to examine the association between the sulfur microbial diet and incidence of overall and distal CRC by molecular and microbial subtype in the Health Professionals Follow-Up Study (1986-2012). RESULTS We documented 1,264 incident CRC cases among 48,246 men, approximately 40% of whom had available tissue data. After accounting for multiple hypothesis testing, the relationship between the sulfur microbial diet and CRC incidence did not differ by subtype. However, there was a suggestion of an association by prostaglandin synthase 2 (PTGS2) status with a multivariable adjusted hazard ratio for highest vs lowest tertile of sulfur microbial diet scores of 1.31 (95% confidence interval: 0.99-1.74, Ptrend = 0.07, Pheterogeneity = 0.04) for PTGS2-high CRC. The association of the sulfur microbial diet with distal CRC seemed to differ by the presence of intratumoral Bifidobacterium spp. with an adjusted hazard ratio for highest vs lowest tertile of sulfur microbial diet scores of 1.65 (95% confidence interval: 1.14-2.39, Ptrend = 0.01, Pheterogeneity = 0.03) for Bifidobacterium-negative distal CRC. We observed no apparent heterogeneity by other tested molecular markers. DISCUSSION Greater long-term adherence to the sulfur microbial diet could be associated with PTGS2-high and Bifidobacterium-negative distal CRC in men. Additional studies are needed to further characterize the role of gut microbial sulfur metabolism and CRC.
Collapse
Affiliation(s)
- Daniel R. Sikavi
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Long H. Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Koichiro Haruki
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Dong D. Wang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kelsey N. Thompson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yan Yan
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Tobyn Branck
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jeremy E. Wilkinson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Naohiko Akimoto
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Rong Zhong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Mai Chan Lau
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kosuke Mima
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Keisuke Kosumi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Teppei Morikawa
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Eric B. Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Wendy S. Garrett
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Medicine, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jacques Izard
- Department of Food Science and Technology, University of Nebraska, Lincoln, Nebraska, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri, USA
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri, USA
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, Massachusetts, USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Interference of dietary polyphenols with potentially toxic amino acid metabolites derived from the colonic microbiota. Amino Acids 2021; 54:311-324. [PMID: 34235577 DOI: 10.1007/s00726-021-03034-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/24/2021] [Indexed: 02/07/2023]
Abstract
Each day, varying amounts of undigested or partially digested proteins reach the colon where they are metabolized by the microbiota, resulting in the formation of compounds such as ammonia, p-cresol, skatole, phenol, indole, and hydrogen sulfide (H2S). In farm animals, the excessive production of these metabolites can affect the quality of meat and milk and is a source of contaminating emissions from animal manure. In humans, their accumulation is potentially harmful, and it has been proposed that they could be involved in the development of pathologies such as colorectal cancer and ulcerative colitis, among others. This review assesses the evidence supporting the use of dietary polyphenols to reduce the production of these metabolites. Most studies have used condensed (proanthocyanidins) or hydrolyzable (ellagitannins and gallotannins) tannins, and have been carried out in farm animals. Several show that the administration of tannins in pigs, chicken, and ruminants decreases the levels of ammonia, p-cresol, skatole, and/or H2S, improving meat/milk quality and reducing manure odor. Direct application of tannins to manure also decreases ammonia emissions. Few studies were carried out in rats and humans and their results confirm, to a lesser extent, those reported in farm animals. These effects would be due to the capacity of tannins to trap ammonia and H2S, and to modify the composition of the microbiota, reducing the bacterial populations producing metabolites. In addition, PACs prevent p-cresol and H2S-induced alterations on intestinal cells in vitro. Tannins, therefore, appear as an interesting tool for improving the quality of animal products, human health, and the harmful emissions associated with breeding.
Collapse
|
18
|
Mitchell SC. Nutrition and sulfur. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 96:123-174. [PMID: 34112351 DOI: 10.1016/bs.afnr.2021.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sulfur is unusual in that it is a mineral that may be taken into the body in both inorganic and organic combinations. It has been available within the environment throughout the development of lifeforms and as such has become integrated into virtually every aspect of biochemical function. It is essential for the nature and maintenance of structure, assists in communication within the organism, is vital as a catalytic assistant in intermediary metabolism and the mechanism of energy flow as well as being involved in internal defense against potentially damaging reactive species and invading foreign chemicals. Recent studies have suggested extended roles for sulfur-containing molecules within living systems. As such, questions have been raised as to whether or not humans are receiving sufficient sulfur within their diet. Sulfur appears to have been the "poor relation" with regards to mineral nutrition. This may be because of difficulties encountered over its multifarious functions, the many chemical guises in which it may be ingested and its complex biochemical interconversions once taken into the body. No established daily requirements have been determined, unlike many minerals, although suggestions have been proposed. Owing to its widespread distribution within dietary components its intake has almost been taken for granted. In the majority of individuals partaking of a balanced diet the supply is deemed adequate, but those opting for specialized or restrictive diets may experience occasional and low-level shortages. In these instances, the careful use of sulfur supplements may be of benefit.
Collapse
Affiliation(s)
- Stephen C Mitchell
- Faculty of Medicine, Imperial College London, London, England, United Kingdom.
| |
Collapse
|
19
|
Sokolov AS, Nekrasov PV, Shaposhnikov MV, Moskalev AA. Hydrogen sulfide in longevity and pathologies: Inconsistency is malodorous. Ageing Res Rev 2021; 67:101262. [PMID: 33516916 DOI: 10.1016/j.arr.2021.101262] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/18/2021] [Accepted: 01/24/2021] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) is one of the biologically active gases (gasotransmitters), which plays an important role in various physiological processes and aging. Its production in the course of methionine and cysteine catabolism and its degradation are finely balanced, and impairment of H2S homeostasis is associated with various pathologies. Despite the strong geroprotective action of exogenous H2S in C. elegans, there are controversial effects of hydrogen sulfide and its donors on longevity in other models, as well as on stress resistance, age-related pathologies and aging processes, including regulation of senescence-associated secretory phenotype (SASP) and senescent cell anti-apoptotic pathways (SCAPs). Here we discuss that the translation potential of H2S as a geroprotective compound is influenced by a multiplicity of its molecular targets, pleiotropic biological effects, and the overlapping ranges of toxic and beneficial doses. We also consider the challenges of the targeted delivery of H2S at the required dose. Along with this, the complexity of determining the natural levels of H2S in animal and human organs and their ambiguous correlations with longevity are reviewed.
Collapse
|
20
|
Vrzáčková N, Ruml T, Zelenka J. Postbiotics, Metabolic Signaling, and Cancer. Molecules 2021; 26:molecules26061528. [PMID: 33799580 PMCID: PMC8000401 DOI: 10.3390/molecules26061528] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 12/19/2022] Open
Abstract
Postbiotics are health-promoting microbial metabolites delivered as a functional food or a food supplement. They either directly influence signaling pathways of the body or indirectly manipulate metabolism and the composition of intestinal microflora. Cancer is the second leading cause of death worldwide and even though the prognosis of patients is improving, it is still poor in the substantial part of the cases. The preventable nature of cancer and the importance of a complex multi-level approach in anticancer therapy motivate the search for novel avenues of establishing the anticancer environment in the human body. This review summarizes the principal findings demonstrating the usefulness of both natural and synthetic sources of postbotics in the prevention and therapy of cancer. Specifically, the effects of crude cell-free supernatants, the short-chain fatty acid butyrate, lactic acid, hydrogen sulfide, and β-glucans are described. Contradictory roles of postbiotics in healthy and tumor tissues are highlighted. In conclusion, the application of postbiotics is an efficient complementary strategy to combat cancer.
Collapse
|
21
|
Smith NW, Shorten PR, Altermann E, Roy NC, McNabb WC. Examination of hydrogen cross-feeders using a colonic microbiota model. BMC Bioinformatics 2021; 22:3. [PMID: 33407079 PMCID: PMC7789523 DOI: 10.1186/s12859-020-03923-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Hydrogen cross-feeding microbes form a functionally important subset of the human colonic microbiota. The three major hydrogenotrophic functional groups of the colon: sulphate-reducing bacteria (SRB), methanogens and reductive acetogens, have been linked to wide ranging impacts on host physiology, health and wellbeing. Results An existing mathematical model for microbial community growth and metabolism was combined with models for each of the three hydrogenotrophic functional groups. The model was further developed for application to the colonic environment via inclusion of responsive pH, host metabolite absorption and the inclusion of host mucins. Predictions of the model, using two existing metabolic parameter sets, were compared to experimental faecal culture datasets. Model accuracy varied between experiments and measured variables and was most successful in predicting the growth of high relative abundance functional groups, such as the Bacteroides, and short chain fatty acid (SCFA) production. Two versions of the colonic model were developed: one representing the colon with sequential compartments and one utilising a continuous spatial representation. When applied to the colonic environment, the model predicted pH dynamics within the ranges measured in vivo and SCFA ratios comparable to those in the literature. The continuous version of the model simulated relative abundances of microbial functional groups comparable to measured values, but predictions were sensitive to the metabolic parameter values used for each functional group. Sulphate availability was found to strongly influence hydrogenotroph activity in the continuous version of the model, correlating positively with SRB and sulphide concentration and negatively with methanogen concentration, but had no effect in the compartmentalised model version. Conclusions Although the model predictions compared well to only some experimental measurements, the important features of the colon environment included make it a novel and useful contribution to modelling the colonic microbiota.
Collapse
Affiliation(s)
- Nick W Smith
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand.,Riddet Institute, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand.,AgResearch, Ruakura Research Centre, Private Bag 3123, Hamilton, 3240, New Zealand
| | - Paul R Shorten
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand. .,AgResearch, Ruakura Research Centre, Private Bag 3123, Hamilton, 3240, New Zealand.
| | - Eric Altermann
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand.,AgResearch, Grasslands Research Centre, Private Bag 11008, Palmerston North, 4442, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C Roy
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Department of Human Nutrition, University of Otago, Dunedin, New Zealand.,Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Private Bag 11222, Palmerston North, 4442, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| |
Collapse
|
22
|
Gotteland M, Riveros K, Gasaly N, Carcamo C, Magne F, Liabeuf G, Beattie A, Rosenfeld S. The Pros and Cons of Using Algal Polysaccharides as Prebiotics. Front Nutr 2020; 7:163. [PMID: 33072794 PMCID: PMC7536576 DOI: 10.3389/fnut.2020.00163] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
Macroalgae stand out for their high content of dietary fiber (30–75%) that include soluble, sulfated (fucoidan, agaran, carrageenan, and ulvan) and non-sulfated (laminaran and alginate) polysaccharides. Many studies indicate that these compounds exert varied biological activities and health-promoting effects and for this reason, there is a growing interest for using them in food products. The aim of this review was to critically evaluate prebiotic properties of algal polysaccharides, i.e., their ability to exert biological activities by modulating the composition and/or diversity of gut microbiota (GM). Pre-clinical studies show that the non-sulfated alginate and laminaran are well-fermented by GM, promoting the formation of short chain fatty acids (SCFAs) including butyrate, and preventing that of harmful putrefactive compounds (NH3, phenol, p-cresol, indole and H2S). Alginate increases Bacteroides, Bifidobacterium, and Lactobacillus species while laminaran mostly stimulates Bacteroides sp. Results with sulfated polysaccharides are more questionable. Agarans are poorly fermentable but agarose-oligosaccharides exhibit an interesting prebiotic potential, increasing butyrate-producing bacteria and SCFAs. Though carrageenan-oligosaccharides are also fermented, their use is currently limited due to safety concerns. Regarding fucoidan, only one study reports SCFAs production, suggesting that it is poorly fermented. Its effect on GM does not indicate a clear pattern, making difficult to conclude whether it is beneficial or not. Notably, fucoidan impact on H2S production has not been evaluated, though some studies report it increases sulfate-reducing bacteria. Ulvan is badly fermented by GM and some studies show that part of its sulfate is dissimilated to H2S, which could affect colonic mitochondrial function. Accordingly, these results support the use of laminaran, alginate and agaro-oligosaccharides as prebiotics while more studies are necessary regarding that of fucoidan, carrageenan and ulvan. However, the realization of clinical trials is necessary to confirm such prebiotic properties in humans.
Collapse
Affiliation(s)
- Martin Gotteland
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile.,Department of Human Nutrition, Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile.,Millennium Nucleus in the Biology of Intestinal Microbiota, Santiago, Chile
| | - Karla Riveros
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Naschla Gasaly
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Constanza Carcamo
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fabien Magne
- Microbiology and Mycology Program, Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Gianella Liabeuf
- Department of Nutrition, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Alejandra Beattie
- Laboratorio de Ecosistemas Marinos Antárticos y Subantárticos, Universidad de Magallanes, Punta Arenas, Chile.,Centro de Investigación para la Conservación de Ecosistemas Australes, Punta Arenas, Chile
| | - Sebastián Rosenfeld
- Laboratorio de Ecosistemas Marinos Antárticos y Subantárticos, Universidad de Magallanes, Punta Arenas, Chile.,Laboratorio de Ecología Molecular, Departamento de Ciencias Ecológicas, Facultad de Ciencias, Universidad de Chile, Santiago, Chile.,Instituto de Ecología y Biodiversidad, Santiago, Chile
| |
Collapse
|
23
|
AROTARITEI D, TURNEA M, IONITE C, ROTARIU M. Artificial intelligence applied to model the sulphur absorption process - a possible application in cure with sulphurous mineral water. BALNEO RESEARCH JOURNAL 2020. [DOI: 10.12680/balneo.2020.358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The presence of Sulphur in human body is estimated to be around .25 percent from total body weight. The modern therapy includes among others the usage of Sulphur us spring waters as crenotherapy or external use like - therapeutic and preventive treatment for a large category of diseases. High levels of hydrogen sulphide are extremely toxic and as result the model of absorption of Sulphur could be useful as predictor in a daily treatment during a cure with sulphurous mineral water.
This quantity of chemical elements from a specific diet rich in these elements absorbed in human organism is one of the most important characteristics of a benefic cure in medical diseases. The most studied mechanisms of absorption were studied for Zinc, Magnesium, Iron and Calcium. The most common way to develop a mathematical model is to use the pharmacokinetic equations based on Michaelis-Menten approach (first order model) and to develop it thereafter for quantitative relations. In some cases, only experimental data can exists and the pharmacokinetic model is not completely elucidated or a simplified model doesn’t exist yet. A model based on genetic programming is proposed in order to discover a mathematical relationship between dietary sulphate (mmol/day) and total sulphate in ileostomy fluid (mmol/day) using experimental data published in literature.
The set of terminals that are used in genetic programing – GP (using Polish notation) is reduced to basic operation (sum, difference, multiplication and division) along with the most plausible operations that could have physical meaning: rooted square, exponential and power. The formulas discovered by GP using experimental proved a good fit of data with discovered mathematical formula, e.g. the rooted mean square error below 1.5% and R2 94.36%.
Mathematical formulas discovered by genetic programming can be used as an alternative to pharmacokinetic model in order to predict sulphate and sulphite absorption and excretion. The usage of this method is especially efficient when the mechanism of absorption in not elucidated enough to provide a compartmental model given by a set of equations. A correlation with pharmacokinetic equations in the case that these exist will help the improvement of terminals and alphabet used by genetic programming in order to have a model closer to one that have a physical meaning
Collapse
Affiliation(s)
- Dragos AROTARITEI
- 1 University of Medicine and Pharmacy “Grigore T. Popa, 16 University Street, Iasi, Romania
| | - Marius TURNEA
- 1 University of Medicine and Pharmacy “Grigore T. Popa, 16 University Street, Iasi, Romania
| | - Catalin IONITE
- 1 University of Medicine and Pharmacy “Grigore T. Popa, 16 University Street, Iasi, Romania
| | - Mariana ROTARIU
- 1 University of Medicine and Pharmacy “Grigore T. Popa, 16 University Street, Iasi, Romania
| |
Collapse
|
24
|
Chitosan-chelated zinc modulates cecal microbiota and attenuates inflammatory response in weaned rats challenged with Escherichia coli. J Microbiol 2020; 58:780-792. [PMID: 32870484 DOI: 10.1007/s12275-020-0056-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 02/08/2023]
Abstract
Escherichia coli (E. coli) infection is very common among young growing animals, and zinc supplementation is often used to alleviate inflammation induced by this disease. Therefore, the objective of this study was to evaluate whether chitosan-chelated zinc (CS-Zn) supplementation could attenuate gut injury induced by E. coli challenge and to explore how CS-Zn modulates cecal microbiota and alleviates intestinal inflammation in weaned rats challenged with E. coli. 36 weaned rats (55.65 ± 2.18 g of BW, n = 12) were divided into three treatment groups consisting of unchallenged rats fed a basal diet (Control) and two groups of rats challenged with E. coli and fed a basal diet or a diet containing 640 mg/kg CS-Zn (E. coli + CS-Zn, containing 50 mg/kg Zn) for a 14-day experiment. On days 10 to 12, each rat was given 4 ml of E. coli solution with a total bacteria count of 1010 CFU by oral gavage daily or normal saline of equal dosage. CS-Zn supplementation mitigated intestinal morphology impairment (e.g. higher crypt depth and lower macroscopic damage index) induced by E. coli challenge (P < 0.05), and alleviated the increase of Myeloperoxidase (MPO) activity after E. coli challenge (P < 0.05). 16S rRNA sequencing analyses revealed that E. coli challenge significantly increased the abundance of Verrucomicrobia and E. coli (P < 0.05). However, CS-Zn supplementation increased the abundance of Lactobacillus and decreased the relative abundance of Proteobacteria, Desulfovibrio and E. coli (P < 0.05). The concentrations of butyrate in the cecal digesta, which decreased due to the challenge, were higher in the E. coli + CS-Zn group (P < 0.05). In addition, CS-Zn supplementation significantly prevented the elevation of pro-inflammatory cytokines IL-6 concentration and up-regulated the level of anti-inflammatory cytokines IL-10 in cecal mucosa induced by E. coli infection (P < 0.05). In conclusion, these results indicate that CS-Zn produces beneficial effects in alleviating gut mucosal injury of E. coli challenged rats by enhancing the intestinal morphology and modulating cecal bacterial composition, as well as attenuating inflammatory response.
Collapse
|
25
|
Magnesium Sulfate-Rich Natural Mineral Waters in the Treatment of Functional Constipation-A Review. Nutrients 2020; 12:nu12072052. [PMID: 32664341 PMCID: PMC7400933 DOI: 10.3390/nu12072052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023] Open
Abstract
Functional constipation (FC) is a chronic constipation for which no physiological, anatomical or iatrogenic origin can be evidenced. This condition has a high impact on a patient’s quality of life and healthcare costs. Since FC is frequently associated with low physical activity and a diet low in fiber and/or water, first-line recommendations focus on sufficient activity, and sufficient fiber and water intake. In case of inefficacy of these measures, numerous drug treatments are available, either over the counter or on prescription. Magnesium sulfate has a long history in the treatment of FC, and magnesium sulfate-rich mineral waters have been used for centuries for their laxative properties. The laxative effect of magnesium and sulfate has since been widely demonstrated. Nevertheless, it appears that no clinical studies aiming at demonstrating their efficacy in FC had been conducted before the 21st century. In this paper, we reviewed the clinical data reporting the efficacy of magnesium sulfate-rich natural mineral waters. In view of their reported efficacy and safety, magnesium sulfate-rich natural mineral waters may represent a natural treatment for FC.
Collapse
|
26
|
Smith NW, Shorten PR, Altermann E, Roy NC, McNabb WC. Competition for Hydrogen Prevents Coexistence of Human Gastrointestinal Hydrogenotrophs in Continuous Culture. Front Microbiol 2020; 11:1073. [PMID: 32547517 PMCID: PMC7272605 DOI: 10.3389/fmicb.2020.01073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/29/2020] [Indexed: 01/24/2023] Open
Abstract
Understanding the metabolic dynamics of the human gastrointestinal tract (GIT) microbiota is of growing importance as research continues to link the microbiome to host health status. Microbial strains that metabolize hydrogen have been associated with a variety of both positive and negative host nutritional and health outcomes, but limited data exists for their competition in the GIT. To enable greater insight into the behaviour of these microbes, a mathematical model was developed for the metabolism and growth of the three major hydrogenotrophic groups: sulphate-reducing bacteria (SRB), methanogens and reductive acetogens. In batch culture simulations with abundant sulphate and hydrogen, the SRB outcompeted the methanogen for hydrogen due to having a half-saturation constant 106 times lower than that of the methanogen. The acetogen, with a high model threshold for hydrogen uptake of around 70 mM, was the least competitive. Under high lactate and zero sulphate conditions, hydrogen exchange between the SRB and the methanogen was the dominant interaction. The methanogen grew at 70% the rate of the SRB, with negligible acetogen growth. In continuous culture simulations, both the SRB and the methanogen were washed out at dilution rates above 0.15 h−1 regardless of substrate availability, whereas the acetogen could survive under abundant hydrogen conditions. Specific combinations of conditions were required for survival of more than one hydrogenotroph in continuous culture, and survival of all three was not possible. The stringency of these requirements and the inability of the model to simulate survival of all three hydrogenotrophs in continuous culture demonstrates that factors outside of those modelled are vital to allow hydrogenotroph coexistence in the GIT.
Collapse
Affiliation(s)
- Nick W Smith
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,AgResearch, Ruakura Research Centre, Hamilton, New Zealand.,AgResearch, Grasslands Research Centre, Palmerston North, New Zealand
| | - Paul R Shorten
- Riddet Institute, Massey University, Palmerston North, New Zealand.,AgResearch, Ruakura Research Centre, Hamilton, New Zealand
| | - Eric Altermann
- Riddet Institute, Massey University, Palmerston North, New Zealand.,AgResearch, Grasslands Research Centre, Palmerston North, New Zealand
| | - Nicole C Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand.,AgResearch, Grasslands Research Centre, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand.,Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Warren C McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| |
Collapse
|
27
|
Cires MJ, Navarrete P, Pastene E, Carrasco-Pozo C, Valenzuela R, Medina DA, Andriamihaja M, Beaumont M, Blachier F, Gotteland M. Protective Effect of an Avocado Peel Polyphenolic Extract Rich in Proanthocyanidins on the Alterations of Colonic Homeostasis Induced by a High-Protein Diet. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:11616-11626. [PMID: 31542929 DOI: 10.1021/acs.jafc.9b03905] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Avocado peel, a byproduct from the avocado pulp industry, is a promising source of polyphenolic compounds. We evaluated the effect of a proanthocyanidin-rich avocado peel polyphenol extract (AvPPE) on the composition and metabolic activity of human fecal microbiota cultured for 24 h in a bioreactor in the presence of high protein (HP) amounts and the effect of the resulting culture supernatants (CSs) on HT-29Glc-/+ and Caco-2 cells. AvPPE decreased the HP-induced production of ammonia, H2S, propionate, and isovalerate and increased that of indole and butyrate. Microbiota composition was marginally affected by HP, whileAvPPE increased the microorganisms/abundance of phylum Actinobacteria, families Coriobacteriaceae and Ruminococcaceae, and genus Faecalibacterium. AvPPE failed to prevent the HP-induced decrease of HT-29Glc-/+ cell viability and energy efficiency but prevented the HP-induced alterations of barrier function in Caco-2 cells. Additionally, the genotoxic effect of the CSs upon HT-29Glc-/+ was attenuated by AvPPE. Therefore, AvPPE may be considered as a promising product for improving colonic homeostasis.
Collapse
Affiliation(s)
- María José Cires
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
| | - Paola Navarrete
- Laboratory of Microbiology and Probiotics, Institute of Nutrition and Food Technology (INTA) , University of Chile , Santiago 7830490 , Chile
- Millennium Nucleus in the Biology of Intestinal Microbiota , Santiago , Chile
| | - Edgar Pastene
- Laboratory of Pharmacognosy, Faculty of Pharmacy , University of Concepción , Concepción 4030000 , Chile
- Laboratorio de Sı́ntesis y Biotransformación de Productos Naturales, Departamento de Ciencias Básicas, Facultad de Ciencias , Universidad del Bı́o-Bı́o , Chillán 3780000 , Chile
| | - Catalina Carrasco-Pozo
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
- Discovery Biology, Griffith Institute for Drug Discovery , Griffith University , Nathan , Queensland 4111 , Australia
| | - Rodrigo Valenzuela
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
| | - Daniel A Medina
- Laboratorio de Biotecnología Aplicada, Facultad de Medicina Veterinaria , Universidad San Sebastián , Lago Panguipulli 1390 , Puerto Montt 5480000 , Chile
| | | | - Martin Beaumont
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay , Paris 75005 , France
- GenPhySE, Université de Toulouse, INRA, INPT, ENVT , Toulouse 31326 , France
| | - François Blachier
- UMR PNCA, AgroParisTech, INRA, Université Paris-Saclay , Paris 75005 , France
| | - Martin Gotteland
- Department of Nutrition, Faculty of Medicine , University of Chile , Santiago 8380453 , Chile
- Laboratory of Microbiology and Probiotics, Institute of Nutrition and Food Technology (INTA) , University of Chile , Santiago 7830490 , Chile
- Millennium Nucleus in the Biology of Intestinal Microbiota , Santiago , Chile
| |
Collapse
|
28
|
Karauzum H, Updegrove TB, Kong M, Wu IL, Datta SK, Ramamurthi KS. Vaccine display on artificial bacterial spores enhances protective efficacy against Staphylococcus aureus infection. FEMS Microbiol Lett 2019; 365:5061626. [PMID: 30084923 DOI: 10.1093/femsle/fny190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022] Open
Abstract
Spores of Bacillus subtilis are encased in a protein coat composed of ∼80 different proteins. Recently, we reconstituted the basement layer of the coat, composed of two structural proteins (SpoVM and SpoIVA) around spore-sized silica beads encased in a lipid bilayer, to create synthetic spore-like particles termed 'SSHELs'. We demonstrated that SSHELs could display thousands of copies of proteins and small molecules of interest covalently linked to SpoIVA. In this study, we investigated the efficacy of SSHELs in delivering vaccines. We show that intramuscular vaccination of mice with undecorated one micron-diameter SSHELs elicited an antibody response against SpoIVA. We further demonstrate that SSHELs covalently modified with a catalytically inactivated staphylococcal alpha toxin variant (HlaH35L), without an adjuvant, resulted in improved protection against Staphylococcus aureus infection in a bacteremia model as compared to vaccination with the antigen alone. Although vaccination with either HlaH35L or HlaH35L conjugated to SSHELs similarly elicited the production of neutralizing antibodies to Hla, we found that a subset of memory T cells was differentially activated when the antigen was delivered on SSHELs. We propose that the particulate nature of SSHELs elicits a more robust immune response to the vaccine that results in superior protection against subsequent S. aureus infection.
Collapse
Affiliation(s)
- Hatice Karauzum
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Taylor B Updegrove
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Minsuk Kong
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - I-Lin Wu
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| | - Sandip K Datta
- Laboratory of Clinical Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Kumaran S Ramamurthi
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, 37 Convent Drive, Bethesda, MD 20892, USA
| |
Collapse
|
29
|
van den Born JC, Frenay ARS, Koning AM, Bachtler M, Riphagen IJ, Minovíc I, Feelisch M, Dekker MM, Bulthuis MLC, Gansevoort RT, Hillebrands JL, Pasch A, Bakker SJL, van Goor H. Urinary Excretion of Sulfur Metabolites and Risk of Cardiovascular Events and All-Cause Mortality in the General Population. Antioxid Redox Signal 2019; 30:1999-2010. [PMID: 29905081 DOI: 10.1089/ars.2017.7040] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Aims: Thiosulfate and sulfate are metabolites of hydrogen sulfide (H2S), a gaseous signaling molecule with cardiovascular (CV) protective properties. Urinary thiosulfate excretion and sulfate excretion are associated with favorable disease outcome in high-risk patient groups. We investigated the relationship between urinary excretion of sulfur metabolites, and risk of CV events and all-cause mortality in the general population. Results: Subjects (n = 6839) of the Prevention of Renal and Vascular End-stage Disease (PREVEND) study were followed prospectively. At baseline, 24-h urinary excretion of thiosulfate and sulfate was determined. Median urinary thiosulfate and sulfate excretion values were 1.27 (interquartile range [IQR] 0.89-2.37) μmol/24 h and 15.7 (IQR 12.0-20.3) mmol/24 h, respectively. Neither thiosulfate nor sulfate excretion showed an independent association with risk of CV events. Sulfate, but not thiosulfate, was inversely associated with risk of all-cause mortality, independent of potential confounders (hazard ratio 0.73 [95% confidence interval 0.63-0.84], p < 0.001). This association appeared most pronounced for normolipidemic subjects (pinteraction = 0.019). Innovation: The strong association between sulfate excretion and mortality in the general population emphasizes the (patho)physiological importance of sulfate or its precursor H2S. Conclusion: We hypothesize that urinary sulfate excretion, which is inversely associated with all-cause mortality in the general population, holds clinical relevance as a beneficial modulator in health and disease. Antioxid. Redox Signal. 30, 1999-2010.
Collapse
Affiliation(s)
- Joost C van den Born
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Anne-Roos S Frenay
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Anne M Koning
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands.,2 Department of Surgery, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Matthias Bachtler
- 3 Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Ineke J Riphagen
- 4 Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands.,5 Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Isidor Minovíc
- 4 Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Martin Feelisch
- 6 Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,7 NIHR Biomedical Research Centre, University of Southampton, Southampton, United Kingdom.,8 University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Marinda M Dekker
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Marian L C Bulthuis
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Ron T Gansevoort
- 4 Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Jan-Luuk Hillebrands
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| | - Andreas Pasch
- 3 Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Stephan J L Bakker
- 4 Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands.,5 Top Institute Food and Nutrition, Wageningen, The Netherlands
| | - Harry van Goor
- 1 Department of Pathology and Medical Biology, Division of Pathology, University Medical Center Groningen (UMCG), University of Groningen (RUG), Groningen, The Netherlands
| |
Collapse
|
30
|
Dietary Factors in Sulfur Metabolism and Pathogenesis of Ulcerative Colitis. Nutrients 2019; 11:nu11040931. [PMID: 31027194 PMCID: PMC6521024 DOI: 10.3390/nu11040931] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/20/2019] [Accepted: 04/23/2019] [Indexed: 02/07/2023] Open
Abstract
The biogeography of inflammation in ulcerative colitis (UC) suggests a proximal to distal concentration gradient of a toxin. Hydrogen sulfide (H2S) has long been considered one such toxin candidate, and dietary sulfur along with the abundance of sulfate reducing bacteria (SRB) were considered the primary determinants of H2S production and clinical course of UC. The metabolic milieu in the lumen of the colon, however, is the result of a multitude of factors beyond dietary sulfur intake and SRB abundance. Here we present an updated formulation of the H2S toxin hypothesis for UC pathogenesis, which strives to incorporate the interdependency of diet composition and the metabolic activity of the entire colon microbial community. Specifically, we suggest that the increasing severity of inflammation along the proximal-to-distal axis in UC is due to the dilution of beneficial factors, concentration of toxic factors, and changing detoxification capacity of the host, all of which are intimately linked to the nutrient flow from the diet.
Collapse
|
31
|
Dostal Webster A, Staley C, Hamilton MJ, Huang M, Fryxell K, Erickson R, Kabage AJ, Sadowsky MJ, Khoruts A. Influence of short-term changes in dietary sulfur on the relative abundances of intestinal sulfate-reducing bacteria. Gut Microbes 2019; 10:447-457. [PMID: 30810441 PMCID: PMC6748593 DOI: 10.1080/19490976.2018.1559682] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
High-protein diets may be linked to gut inflammation due to increased production of hydrogen sulfide (H2S), a potential toxin, as an end product of microbial fermentation in the colon by sulfidogenic sulfate-reducing bacteria (SRB). We hypothesized that dietary content of sulfur-containing amino acids (SAA) leads to variation in the relative abundances of intestinal SRB, which include Desulfovibrio and Bilophila taxa. To test this hypothesis we performed a pilot crossover study in four healthy volunteers, who consumed two interventional diets for 10-14 days, containing high or low SAA content. The total energy intake was similar between the two dietary extremes. Microbial communities were characterized by 16S rRNA gene amplicon and shotgun next-generation DNA sequencing. While the relative abundance of Desulfovibrio differed among participants (ANOVA P= 0.001), we could not detect a change with dietary treatments. Similarly, no differences in Bilophila abundance were observed among individuals or dietary arms. Inter-personal differences in microbial community composition and functional gene categories differed between subjects and these differences were maintained over the course of the study. These observations are consistent with re-analysis of two previously published dietary intervention studies. Finally, we found that inter-personal differences in the taxonomic composition of fecal microbiota, including the relative abundances of SRB, were maintained over time in 19 healthy individuals in our stool donor program. These results suggest that the use of dietary interventions alone may be insufficient for rapid therapeutic targeting of SRB. Nevertheless, these pilot data provide a foundation to inform future, statistically powered, studies.
Collapse
Affiliation(s)
- Allison Dostal Webster
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA,International Food Information Council, Washington, DC, USA
| | - Christopher Staley
- Biotechnology Institute, University of Minnesota, St. Paul, MN, USA,Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | | | - Merry Huang
- University of Minnesota Medical School, Minneapolis, MN, USA
| | - Kathryn Fryxell
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | - Amanda J. Kabage
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Michael J. Sadowsky
- Biotechnology Institute, University of Minnesota, St. Paul, MN, USA,University of Minnesota Medical School, Minneapolis, MN, USA,Department of Soil, Water, and Climate, University of Minnesota, St. Paul, MN, USA,Department of Plant and Microbial Biology, University of Minnesota, St. Paul, MN, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA,Biotechnology Institute, University of Minnesota, St. Paul, MN, USA,CONTACT Alexander Khoruts Division of Gastroenterology, Department of Medicine, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
32
|
Vedamurthy A, Ananthakrishnan AN. Influence of Environmental Factors in the Development and Outcomes of Inflammatory Bowel Disease. Gastroenterol Hepatol (N Y) 2019; 15:72-82. [PMID: 31011301 PMCID: PMC6469265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Inflammatory bowel disease (IBD), which mainly comprises Crohn's disease (CD) and ulcerative colitis (UC), is a term for chronic inflammatory diseases of the gut arising due to a dysregulated immune response to a dysbiotic gut microbiome on a background of genetic predisposition. However, genetics explains a small fraction of risk, and the external environment plays a large and important role in disease pathogenesis and natural history. Cigarette smoking, one of the earliest- and most-studied risk factors, increases the risk of CD onset and is associated with severe disease. In contrast, although smoking cessation increases the risk of UC onset and relapse, current smoking appears to be protective. Appendectomy is inversely associated with the risk of UC. Various dietary factors may influence risk of IBD, including an inverse association with dietary fiber. Exclusion diets have anecdotal reports of efficacy but lack evidence from rigorous clinical trials. Psychosocial factors such as depression, stress, and sleep disturbance play a role in onset as well as in determining natural history. There is an important need for rigorous studies robustly informing the association and mechanism(s) of influence of the external environment on IBD risk and for clinical trials examining the efficacy of alteration of the environment in ameliorating the course of IBD.
Collapse
Affiliation(s)
- Amar Vedamurthy
- Dr Vedamurthy is a hospitalist in the Division of General Medicine at Massachusetts General Hospital in Boston, Massachusetts and an instructor in medicine at Harvard Medical School in Boston, Massachusetts
- Dr Ananthakrishnan is an associate professor in the Division of Gastroenterology at Massachusetts General Hospital and at Harvard Medical School
| | - Ashwin N Ananthakrishnan
- Dr Vedamurthy is a hospitalist in the Division of General Medicine at Massachusetts General Hospital in Boston, Massachusetts and an instructor in medicine at Harvard Medical School in Boston, Massachusetts
- Dr Ananthakrishnan is an associate professor in the Division of Gastroenterology at Massachusetts General Hospital and at Harvard Medical School
| |
Collapse
|
33
|
Thandauthapani TD, Reeve AJ, Long AS, Turner IJ, Sharp JS. Exposing latent fingermarks on problematic metal surfaces using time of flight secondary ion mass spectroscopy. Sci Justice 2018; 58:405-414. [DOI: 10.1016/j.scijus.2018.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/26/2018] [Accepted: 08/10/2018] [Indexed: 11/24/2022]
|
34
|
Volumetric, viscometric and molecular simulation studies of glycine in aqueous sodium sulphate solutions at different temperatures. J Mol Liq 2018. [DOI: 10.1016/j.molliq.2018.06.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
35
|
Sattari Najafabadi Z, Skau Nielsen T, Skou Hedemann M. Dietary protein source and butyrylated high-amylose maize starch included in a high-protein diet determines the urinary metabolome of rats. Int J Food Sci Nutr 2018; 70:255-266. [PMID: 30160558 DOI: 10.1080/09637486.2018.1499711] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intake of red and processed meat increases the risk of colorectal cancer (CRC), whereas dairy product consumption and the intake of dietary fibre are negatively associated with this risk. We investigated the effect of (i) low-protein diets with either whey or cooked meat (beef) as the protein source and (ii) high-protein diets with cooked meat (beef) either without or with the inclusion of 10% butyrylated high-amylose maize starch (HAMSB), on the urinary metabolome of rats. Urine samples from rats were analysed using untargeted LC-MS metabolomics. The level and source of the dietary protein affected the urinary excretion of numerous metabolites indicating that several metabolic pathways were changed. The inclusion of HAMSB in a high-protein diet caused significant alterations in the excretion of several metabolites. HAMSB reduced urinary excretion of potentially harmful metabolites resulting from a high level of meat consumption.
Collapse
Affiliation(s)
- Zahra Sattari Najafabadi
- a Department of Animal Science, Faculty of Science and Technology , Aarhus University , Tjele , Denmark
| | - Tina Skau Nielsen
- a Department of Animal Science, Faculty of Science and Technology , Aarhus University , Tjele , Denmark
| | - Mette Skou Hedemann
- a Department of Animal Science, Faculty of Science and Technology , Aarhus University , Tjele , Denmark
| |
Collapse
|
36
|
Yao CK, Rotbart A, Ou JZ, Kalantar-Zadeh K, Muir JG, Gibson PR. Modulation of colonic hydrogen sulfide production by diet and mesalazine utilizing a novel gas-profiling technology. Gut Microbes 2018; 9:510-522. [PMID: 29561196 PMCID: PMC6287689 DOI: 10.1080/19490976.2018.1451280] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Excessive hydrogen sulfide (H2S) production from gut microbial metabolism may have clinically important relevance in the pathogenesis of gut disorders, including ulcerative colitis. However, little is known regarding factors that alter its production. Using a newly-designed in vitro gas-profiling technology, the study aimed to verify real-time H2S measurement reproducibility and thereafter, assess its production following exposure to dietary factors and 5-aminosalicylate acid (5-ASA). Measurements of H2S, carbon dioxide, hydrogen and methane measurements were compared between gas-profiling systems. Homogenized slurries were prepared from freshly-passed healthy human feces. Fifty ml slurries were aliquoted into separate fermentation chambers and substrates added including 1 g highly fermentable fructo-oligosaccharides (FOS) or resistant starch Hi-Maize (RS), or minimally fermentable psyllium or sterculia, 1 g cysteine, 0.9 g sodium sulfate or 1.2 mL of 1 M 5-ASA alone or in combinations. H2S release was sampled every 5 mins over 4-h and expressed relative to unspiked controls. RS suppressed H2S production by a mean 89.0 (SEM 4.8)% and FOS by 82.2 (6.2)% compared to <35 (17)% by psyllium and sterculia (p<0.001, two-way ANOVA). Cysteine stimulated H2S production by 1557 (532)%. The addition of FOS to slurries containing cysteine significantly suppressed H2S by 90 (2)% over the addition of 5-ASA (0.3 (2)%, p<0.001). Sulfate and 5-ASA had minimal overall effects. In conclusion, the H2S-profiling technology is a reproducible tool. Production of H2S is greatly enhanced by sulfur-amino acids but not inorganic sulfate, and is effectively suppressed by readily fermentable fibers. These findings inform potential designs of dietary therapies to reduce H2S production in vivo.
Collapse
Affiliation(s)
- Chu K. Yao
- Department of Gastroenterology, Central Clinical School, Alfred Centre, Monash University & Alfred Health, Melbourne, Australia,CONTACT Dr. Chu K. Yao Department of Gastroenterology, Central Clinical School, Monash University, Level 6, The Alfred Centre, 99 Commercial Road, Melbourne VIC 3004
| | - Asaf Rotbart
- School of Engineering, RMIT University, Melbourne, Australia
| | - Jian Z. Ou
- School of Engineering, RMIT University, Melbourne, Australia
| | | | - Jane G. Muir
- Department of Gastroenterology, Central Clinical School, Alfred Centre, Monash University & Alfred Health, Melbourne, Australia
| | - Peter R. Gibson
- Department of Gastroenterology, Central Clinical School, Alfred Centre, Monash University & Alfred Health, Melbourne, Australia
| |
Collapse
|
37
|
Perridon BW, Leuvenink HGD, Hillebrands JL, van Goor H, Bos EM. The role of hydrogen sulfide in aging and age-related pathologies. Aging (Albany NY) 2017; 8:2264-2289. [PMID: 27683311 PMCID: PMC5115888 DOI: 10.18632/aging.101026] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 09/13/2016] [Indexed: 12/14/2022]
Abstract
When humans grow older, they experience inevitable and progressive loss of physiological function, ultimately leading to death. Research on aging largely focuses on the identification of mechanisms involved in the aging process. Several proposed aging theories were recently combined as the 'hallmarks of aging'. These hallmarks describe (patho-)physiological processes that together, when disrupted, determine the aging phenotype. Sustaining evidence shows a potential role for hydrogen sulfide (H2S) in the regulation of aging. Nowadays, H2S is acknowledged as an endogenously produced signaling molecule with various (patho-) physiological effects. H2S is involved in several diseases including pathologies related to aging. In this review, the known, assumed and hypothetical effects of hydrogen sulfide on the aging process will be discussed by reviewing its actions on the hallmarks of aging and on several age-related pathologies.
Collapse
Affiliation(s)
- Bernard W Perridon
- Department of Pathology and Medical Biology, University Medical Center Groningen, the Netherlands
| | | | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, University Medical Center Groningen, the Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center Groningen, the Netherlands
| | - Eelke M Bos
- Department of Pathology and Medical Biology, University Medical Center Groningen, the Netherlands.,Department of Neurosurgery, Erasmus Medical Center Rotterdam, the Netherlands
| |
Collapse
|
38
|
Doleman JF, Grisar K, Van Liedekerke L, Saha S, Roe M, Tapp HS, Mithen RF. The contribution of alliaceous and cruciferous vegetables to dietary sulphur intake. Food Chem 2017; 234:38-45. [PMID: 28551250 PMCID: PMC5460521 DOI: 10.1016/j.foodchem.2017.04.098] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 03/17/2017] [Accepted: 04/17/2017] [Indexed: 01/01/2023]
Abstract
Despite its importance in many areas of human metabolism, there are no recommended daily intake guide lines for sulphur. It is generally assumed that most dietary sulphur originates from intake of methionine and cysteine. We estimated sulphur intake from food diaries, and validated the results with the use of a duplicate diet analyses. Sulphur intake estimations were highly correlated with that obtain through an elemental analysis of duplicate diets, with a mean±sd daily intakes of 956±327.9mg estimated from diet diary analyses and 935±329.9mg estimated by a duplicate diet analyses. Sulphur intake from alliaceous and cruciferous vegetables contributed up to 42% of total sulphur intake. Daily intake estimation comparisons through diet diary analyses and duplicate diet for other elements showed good agreement, except for sodium and zinc, in which analyses of 24h diet dairies overestimated intake by 35% and 52%, respectively.
Collapse
Affiliation(s)
- Joanne F Doleman
- Food & Health Programme, Institute of Food Research, Norwich, UK
| | - Katrijn Grisar
- Food & Health Programme, Institute of Food Research, Norwich, UK
| | | | - Shikha Saha
- Food & Health Programme, Institute of Food Research, Norwich, UK
| | - Mark Roe
- Food Databanks, Institute of Food Research, Norwich NR4 7UA, UK
| | - Henri S Tapp
- Analytical Sciences Unit, Institute of Food Research, Norwich NR4 7UA, UK
| | - Richard F Mithen
- Food & Health Programme, Institute of Food Research, Norwich, UK.
| |
Collapse
|
39
|
Pichette J, Fynn-Sackey N, Gagnon J. Hydrogen Sulfide and Sulfate Prebiotic Stimulates the Secretion of GLP-1 and Improves Glycemia in Male Mice. Endocrinology 2017; 158:3416-3425. [PMID: 28977605 DOI: 10.1210/en.2017-00391] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
Recently, the gastrointestinal microbiome, and its metabolites, has emerged as a potential regulator of host metabolism. However, to date little is known on the precise mechanisms of how this regulation occurs. Hydrogen sulfide (H2S) is abundantly produced in the colon by sulfate-reducing bacteria (SRB). H2S is a bioactive gas that plays regulatory roles in many systems, including metabolic hormone regulation. This gas metabolite is produced in close proximity to the glucagonlike peptide-1 (GLP-1)-secreting cells in the gut epithelium. GLP-1 is a peptide hormone that plays pivotal roles in both glucose homeostasis and appetite regulation. We hypothesized that H2S can directly regulate GLP-1 secretion. We demonstrated that H2S donors (NaHS and GYY4137) directly stimulate GLP-1 secretion in murine L-cells (GLUTag) and that this occurs through p38 mitogen-activated protein kinase without affecting cell viability. We then increased SRB in mice by supplementing the diet with a prebiotic chondroitin sulfate for 4 weeks. Mice treated with chondroitin sulfate had elevated Desulfovibrio piger levels in the feces and increased colonic and fecal H2S concentration. These animals also had enhanced GLP-1 and insulin secretion, improved oral glucose tolerance, and reduced food consumption. These results indicate that H2S plays a stimulatory role in GLP-1 secretion and that sulfate prebiotics can enhance GLP-1 release and its downstream metabolic actions.
Collapse
Affiliation(s)
- Jennifer Pichette
- Laurentian University, Department of Biology, Sudbury, Ontario P3E 2C6, Canada
| | - Nancy Fynn-Sackey
- Laurentian University, Department of Biology, Sudbury, Ontario P3E 2C6, Canada
| | - Jeffrey Gagnon
- Laurentian University, Department of Biology, Sudbury, Ontario P3E 2C6, Canada
| |
Collapse
|
40
|
Barton LL, Ritz NL, Fauque GD, Lin HC. Sulfur Cycling and the Intestinal Microbiome. Dig Dis Sci 2017; 62:2241-2257. [PMID: 28766244 DOI: 10.1007/s10620-017-4689-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 07/19/2017] [Indexed: 02/08/2023]
Abstract
In this review, we focus on the activities transpiring in the anaerobic segment of the sulfur cycle occurring in the gut environment where hydrogen sulfide is produced. While sulfate-reducing bacteria are considered as the principal agents for hydrogen sulfide production, the enzymatic desulfhydration of cysteine by heterotrophic bacteria also contributes to production of hydrogen sulfide. For sulfate-reducing bacteria respiration, molecular hydrogen and lactate are suitable as electron donors while sulfate functions as the terminal electron acceptor. Dietary components provide fiber and macromolecules that are degraded by bacterial enzymes to monomers, and these are fermented by intestinal bacteria with the production to molecular hydrogen which promotes the metabolic dominance by sulfate-reducing bacteria. Sulfate is also required by the sulfate-reducing bacteria, and this can be supplied by sulfate- and sulfonate-containing compounds that are hydrolyzed by intestinal bacterial with the release of sulfate. While hydrogen sulfide in the intestinal biosystem may be beneficial to bacteria by increasing resistance to antibiotics, and protecting them from reactive oxygen species, hydrogen sulfide at elevated concentrations may become toxic to the host.
Collapse
Affiliation(s)
- Larry L Barton
- Department of Biology, MSCO3 2020, University of New Mexico, Albuquerque, NM, 87131, USA.
| | - Nathaniel L Ritz
- New Mexico VA Health Care System, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Guy D Fauque
- CNRS, IRD, Mediterranean Institute of Oceanography (MIO) UM 110, Aix-Marseille Université, Université de Toulon, Campus de Luminy, Case 901, 13288, Marseille Cedex 09, France
| | - Henry C Lin
- New Mexico VA Health Care System, University of New Mexico, Albuquerque, NM, 87131, USA
| |
Collapse
|
41
|
Kellingray L, Tapp HS, Saha S, Doleman JF, Narbad A, Mithen RF. Consumption of a diet rich in Brassica vegetables is associated with a reduced abundance of sulphate-reducing bacteria: A randomised crossover study. Mol Nutr Food Res 2017; 61:1600992. [PMID: 28296348 PMCID: PMC5600105 DOI: 10.1002/mnfr.201600992] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/16/2017] [Accepted: 02/24/2017] [Indexed: 02/06/2023]
Abstract
SCOPE We examined whether a Brassica-rich diet was associated with an increase in the relative abundance of intestinal lactobacilli and sulphate-reducing bacteria (SRB), or alteration to the composition of the gut microbiota, in healthy adults. METHODS AND RESULTS A randomised crossover study was performed with ten healthy adults who were fed a high- and a low-Brassica diet for 2-wk periods, with a 2-wk washout phase separating the diets. The high-Brassica diet consisted of six 84 g portions of broccoli, six 84 g portions of cauliflower and six 300 g portions of a broccoli and sweet potato soup. The low-Brassica diet consisted of one 84 g portion of broccoli and one 84 g portion of cauliflower. Faecal microbiota composition was measured in samples collected following 2-wk Brassica-free periods (consumption of all Brassica prohibited), and after each diet, whereby the only Brassica consumed was that supplied by the study team. No significant changes to the relative abundance of lactobacilli were observed (p = 0.8019). The increased consumption of Brassica was associated with a reduction in the relative abundance of SRB (p = 0.0215), and members of the Rikenellaceae, Ruminococcaceae, Mogibacteriaceae, Clostridium and unclassified Clostridiales (p < 0.01). CONCLUSION The increased consumption of Brassica vegetables was linked to a reduced relative abundance of SRB, and therefore may be potentially beneficial to gastrointestinal health.
Collapse
Affiliation(s)
- Lee Kellingray
- Food and Health ProgrammeInstitute of Food ResearchNorwich Research ParkNorwichUK
| | - Henri S. Tapp
- Analytical Sciences UnitInstitute of Food ResearchNorwich Research ParkNorwichUK
| | - Shikha Saha
- Food and Health ProgrammeInstitute of Food ResearchNorwich Research ParkNorwichUK
| | - Joanne F. Doleman
- Food and Health ProgrammeInstitute of Food ResearchNorwich Research ParkNorwichUK
| | - Arjan Narbad
- Gut Health and Food Safety ProgrammeInstitute of Food ResearchNorwich Research ParkNorwichUK
| | - Richard F. Mithen
- Food and Health ProgrammeInstitute of Food ResearchNorwich Research ParkNorwichUK
| |
Collapse
|
42
|
Lee T, Clavel T, Smirnov K, Schmidt A, Lagkouvardos I, Walker A, Lucio M, Michalke B, Schmitt-Kopplin P, Fedorak R, Haller D. Oral versus intravenous iron replacement therapy distinctly alters the gut microbiota and metabolome in patients with IBD. Gut 2017; 66:863-871. [PMID: 26848182 PMCID: PMC5531225 DOI: 10.1136/gutjnl-2015-309940] [Citation(s) in RCA: 219] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Revised: 12/02/2015] [Accepted: 12/21/2015] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Iron deficiency is a common complication in patients with IBD and oral iron therapy is suggested to exacerbate IBD symptoms. We performed an open-labelled clinical trial to compare the effects of per oral (PO) versus intravenous (IV) iron replacement therapy (IRT). DESIGN The study population included patients with Crohn's disease (CD; N=31), UC (N=22) and control subjects with iron deficiency (non-inflamed, NI=19). After randomisation, participants received iron sulfate (PO) or iron sucrose (IV) over 3 months. Clinical parameters, faecal bacterial communities and metabolomes were assessed before and after intervention. RESULTS Both PO and IV treatments ameliorated iron deficiency, but higher ferritin levels were observed with IV. Changes in disease activity were independent of iron treatment types. Faecal samples in IBD were characterised by marked interindividual differences, lower phylotype richness and proportions of Clostridiales. Metabolite analysis also showed separation of both UC and CD from control anaemic participants. Major shifts in bacterial diversity occurred in approximately half of all participants after IRT, but patients with CD were most susceptible. Despite individual-specific changes in phylotypes due to IRT, PO treatment was associated with decreased abundances of operational taxonomic units assigned to the species Faecalibacterium prausnitzii, Ruminococcus bromii, Dorea sp. and Collinsella aerofaciens. Clear IV-specific and PO-specific fingerprints were evident at the level of metabolomes, with changes affecting cholesterol-derived host substrates. CONCLUSIONS Shifts in gut bacterial diversity and composition associated with iron treatment are pronounced in IBD participants. Despite similar clinical outcome, oral administration differentially affects bacterial phylotypes and faecal metabolites compared with IV therapy. TRIAL REGISTRATION NUMBER clinicaltrial.gov (NCT01067547).
Collapse
Affiliation(s)
- Thomas Lee
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Canada,Department of Gastroenterology, Wollongong Hospital, Wollongong, NSW, Australia
| | - Thomas Clavel
- ZIEL Institute for Food and Health, Technische Universität München, Freising, Germany
| | - Kirill Smirnov
- Research Unit Analytical BioGeoChemistry, German Research Center for Environmental Health, Neuherberg, Germany
| | - Annemarie Schmidt
- Chair of Nutrition and Immunology, Technische Universität München, Freising, Germany
| | - Ilias Lagkouvardos
- ZIEL Institute for Food and Health, Technische Universität München, Freising, Germany
| | - Alesia Walker
- Research Unit Analytical BioGeoChemistry, German Research Center for Environmental Health, Neuherberg, Germany
| | - Marianna Lucio
- Research Unit Analytical BioGeoChemistry, German Research Center for Environmental Health, Neuherberg, Germany
| | - Bernhard Michalke
- Research Unit Analytical BioGeoChemistry, German Research Center for Environmental Health, Neuherberg, Germany
| | - Philippe Schmitt-Kopplin
- ZIEL Institute for Food and Health, Technische Universität München, Freising, Germany,Research Unit Analytical BioGeoChemistry, German Research Center for Environmental Health, Neuherberg, Germany
| | - Richard Fedorak
- Division of Gastroenterology, Department of Medicine, University of Alberta, Edmonton, Canada
| | - Dirk Haller
- ZIEL Institute for Food and Health, Technische Universität München, Freising, Germany,Chair of Nutrition and Immunology, Technische Universität München, Freising, Germany
| |
Collapse
|
43
|
Lewis JD, Abreu MT. Diet as a Trigger or Therapy for Inflammatory Bowel Diseases. Gastroenterology 2017; 152:398-414.e6. [PMID: 27793606 DOI: 10.1053/j.gastro.2016.10.019] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/15/2016] [Accepted: 10/19/2016] [Indexed: 02/07/2023]
Abstract
The most common question asked by patients with inflammatory bowel disease (IBD) is, "Doctor, what should I eat?" Findings from epidemiology studies have indicated that diets high in animal fat and low in fruits and vegetables are the most common pattern associated with an increased risk of IBD. Low levels of vitamin D also appear to be a risk factor for IBD. In murine models, diets high in fat, especially saturated animal fats, also increase inflammation, whereas supplementation with omega 3 long-chain fatty acids protect against intestinal inflammation. Unfortunately, omega 3 supplements have not been shown to decrease the risk of relapse in patients with Crohn's disease. Dietary intervention studies have shown that enteral therapy, with defined formula diets, helps children with Crohn's disease and reduces inflammation and dysbiosis. Although fiber supplements have not been shown definitively to benefit patients with IBD, soluble fiber is the best way to generate short-chain fatty acids such as butyrate, which has anti-inflammatory effects. Addition of vitamin D and curcumin has been shown to increase the efficacy of IBD therapy. There is compelling evidence from animal models that emulsifiers in processed foods increase risk for IBD. We discuss current knowledge about popular diets, including the specific carbohydrate diet and diet low in fermentable oligo-, di-, and monosaccharides and polyols. We present findings from clinical and basic science studies to help gastroenterologists navigate diet as it relates to the management of IBD.
Collapse
Affiliation(s)
- James D Lewis
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Maria T Abreu
- Crohn's and Colitis Center, Department of Medicine, Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida.
| |
Collapse
|
44
|
Dawson PA, Richard K, Perkins A, Zhang Z, Simmons DG. Review: Nutrient sulfate supply from mother to fetus: Placental adaptive responses during human and animal gestation. Placenta 2017; 54:45-51. [PMID: 28089504 DOI: 10.1016/j.placenta.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/23/2016] [Accepted: 01/04/2017] [Indexed: 01/20/2023]
Abstract
Nutrient sulfate has numerous roles in mammalian physiology and is essential for healthy fetal growth and development. The fetus has limited capacity to generate sulfate and relies on sulfate supplied from the maternal circulation via placental sulfate transporters. The placenta also has a high sulfate requirement for numerous molecular and cellular functions, including sulfate conjugation (sulfonation) to estrogen and thyroid hormone which leads to their inactivation. Accordingly, the ratio of sulfonated (inactive) to unconjugated (active) hormones modulates endocrine function in fetal, placental and maternal tissues. During pregnancy, there is a marked increase in the expression of genes involved in transport and generation of sulfate in the mouse placenta, in line with increasing fetal and placental demands for sulfate. The maternal circulation also provides a vital reservoir of sulfate for the placenta and fetus, with maternal circulating sulfate levels increasing by 2-fold from mid-gestation. However, despite evidence from animal studies showing the requirement of maternal sulfate supply for placental and fetal physiology, there are no routine clinical measurements of sulfate or consideration of dietary sulfate intake in pregnant women. This is also relevant to certain xenobiotics or pharmacological drugs which when taken by the mother use significant quantities of circulating sulfate for detoxification and clearance, and thereby have the potential to decrease sulfonation capacity in the placenta and fetus. This article will review the physiological adaptations of the placenta for maintaining sulfate homeostasis in the fetus and placenta, with a focus on pathophysiological outcomes in animal models of disturbed sulfate homeostasis.
Collapse
Affiliation(s)
- P A Dawson
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia.
| | - K Richard
- Conjoint Endocrine Laboratory, Chemical Pathology, Pathology Queensland, Queensland Health, Herston, Australia
| | - A Perkins
- School of Medical Science, Menzies Health Institute Queensland, Griffith University, Gold Coast Campus, Australia
| | - Z Zhang
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| | - D G Simmons
- Mater Research Institute, The University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
45
|
Langford R, Hurrion E, Dawson PA. Genetics and pathophysiology of mammalian sulfate biology. J Genet Genomics 2017; 44:7-20. [DOI: 10.1016/j.jgg.2016.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 12/23/2022]
|
46
|
Metagenomic evidence for taxonomic dysbiosis and functional imbalance in the gastrointestinal tracts of children with cystic fibrosis. Sci Rep 2016; 6:22493. [PMID: 26940651 PMCID: PMC4778032 DOI: 10.1038/srep22493] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 02/12/2016] [Indexed: 01/07/2023] Open
Abstract
Cystic fibrosis (CF) results in inflammation, malabsorption of fats and other nutrients, and obstruction in the gastrointestinal (GI) tract, yet the mechanisms linking these disease manifestations to microbiome composition remain largely unexplored. Here we used metagenomic analysis to systematically characterize fecal microbiomes of children with and without CF, demonstrating marked CF-associated taxonomic dysbiosis and functional imbalance. We further showed that these taxonomic and functional shifts were especially pronounced in young children with CF and diminished with age. Importantly, the resulting dysbiotic microbiomes had significantly altered capacities for lipid metabolism, including decreased capacity for overall fatty acid biosynthesis and increased capacity for degrading anti-inflammatory short-chain fatty acids. Notably, these functional differences correlated with fecal measures of fat malabsorption and inflammation. Combined, these results suggest that enteric fat abundance selects for pro-inflammatory GI microbiota in young children with CF, offering novel strategies for improving the health of children with CF-associated fat malabsorption.
Collapse
|
47
|
Sarin H. Permeation thresholds for hydrophilic small biomolecules across microvascular and epithelial barriers are predictable on basis of conserved biophysical properties. In Silico Pharmacol 2016; 3:5. [PMID: 26820890 PMCID: PMC4471070 DOI: 10.1186/s40203-015-0009-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
Purpose Neutral small hydrophiles are permeable to varying degrees, across the aqueous pores of phospholipid bilayer protein channels, with their potential for permeation into cells being predictable, on the basis of hydrophilicity and size. Here, it is hypothesized that permeation thresholds for small hydrophiles, across capillary zona occludens tight junction and inter-epithelial junction pore complexes are predictable, on the basis of predicted hydrophilicity in context of predicted molecular size and charge distribution, as are those of cations and anions, on the basis of predicted ionization in context of predicted atomic size. Methods Small hydrophiles are categorized by charge distribution. 2-dimensional plots of predicted hydrophilic octanol-to-water partition coefficient (HOWPC; unitless) and predicted van der Waals diameter (vdWD; nm) are generated for each category. The predicted HOWPC-to-vdWD ratio (nm-1), and vdWDs for permeable hydrophile at the maximum and minimum HOWPC-to-vdWD, vdWD @ MAXimum HOWPC-to-vdWD and vdWD @ MINimum HOWPC-to-vdWD are determined. For cations and anions, the ionization-to-atomic diameter ratios (CI or AI-to-AD ratios; nm-1) are determined. Results Per sizes of mixed and pure polyneutral hydrophiles, the permeation size maximum for hydrophiles across tight junction pore complexes is >0.69 ≤ 0.73 nanometers and across inter-epithelial junction pore complexes is ≥ 0.81 nanometers. For hydrophiles with anionicity or cationicity, the vdWDs @ MAXimum HOWPC-to-vdWD are less than those of mixed and polyneutral hydrophiles across both tight and inter-epithelial junctions, ranges specific to category and junction type. For cations, the permeation threshold across tight junctions is between the CI-to-AD ratio of Na+ (+2.69 nm-1) and CH3-Hg+ (+2.36 nm-1), with CH3-Hg+ and K+ (+2.20 nm-1) being permeable; and for divalent cations, the threshold across inter-epithelial junctions is between the CI-to-AD ratio of Mg2+ (+6.25 nm-1) and Ca2+ (+5.08 nm-1) , Ca2+ being semi-permeable. For anions, the permeation threshold across tight junctions is between the AI-to-AD ratio of Cl- (-4.91 nm-1) and Br- (-4.17 nm-1), and the threshold across inter-epithelial junctions is between the AI-to-AD ratio of F- (-7.81 nm-1) and Cl- (-4.91 nm-1). Conclusions In silico modeling reveals that permeation thresholds, of small molecule hydrophiles, cations and anions across junctional pore complexes, are conserved in the physiologic state. Electronic supplementary material The online version of this article (doi:10.1186/s40203-015-0009-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hemant Sarin
- Freelance Investigator in Translational Science and Medicine, Charleston, WV, USA.
| |
Collapse
|
48
|
Epithelial Electrolyte Transport Physiology and the Gasotransmitter Hydrogen Sulfide. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:4723416. [PMID: 26904165 PMCID: PMC4745330 DOI: 10.1155/2016/4723416] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/17/2015] [Indexed: 11/18/2022]
Abstract
Hydrogen sulfide (H2S) is a well-known environmental chemical threat with an unpleasant smell of rotten eggs. Aside from the established toxic effects of high-dose H2S, research over the past decade revealed that cells endogenously produce small amounts of H2S with physiological functions. H2S has therefore been classified as a "gasotransmitter." A major challenge for cells and tissues is the maintenance of low physiological concentrations of H2S in order to prevent potential toxicity. Epithelia of the respiratory and gastrointestinal tract are especially faced with this problem, since these barriers are predominantly exposed to exogenous H2S from environmental sources or sulfur-metabolising microbiota. In this paper, we review the cellular mechanisms by which epithelial cells maintain physiological, endogenous H2S concentrations. Furthermore, we suggest a concept by which epithelia use their electrolyte and liquid transport machinery as defence mechanisms in order to eliminate exogenous sources for potentially harmful H2S concentrations.
Collapse
|
49
|
Abstract
1. Sulphonation is unusual amongst the common Phase II (condensation; synthetic) reactions experienced by xenobiotics, in that the availability of the conjugating agent, sulphate, may become a rate-limiting factor. This sulphate is derived within the body via the oxygenation of sulphur moieties liberated from numerous ingested compounds including the sulphur-containing amino acids. Preformed inorganic sulphate also makes a considerable contribution to this pool. 2. There has been a divergence of opinion as to whether or not inorganic sulphate may be readily absorbed from the gastrointestinal tract and this controversy still continues in some quarters. Even more so, is the vexing question of potential absorption of inorganic sulphate via the lungs and through the skin. 3. This review examines the relevant diverse literature and concludes that sulphate ions may move across biological membranes by means of specific transporters and, although the gastrointestinal tract is by far the major portal of entry, some absorption across the lungs and the skin may take place under appropriate circumstances.
Collapse
Affiliation(s)
- Stephen C Mitchell
- a Computational and Systems Medicine, Department of Surgery and Cancer, Faculty of Medicine , Imperial College London , South Kensington , London , UK and
| | - Rosemary H Waring
- b Birmingham Toxicology Consortium, School of Biosciences, University of Birmingham , Edgbaston , Birmingham , UK
| |
Collapse
|
50
|
Rakoczy J, Lee S, Weerasekera SJ, Simmons DG, Dawson PA. Placental and fetal cysteine dioxygenase gene expression in mouse gestation. Placenta 2015; 36:956-9. [PMID: 26119969 DOI: 10.1016/j.placenta.2015.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/15/2015] [Accepted: 06/10/2015] [Indexed: 01/01/2023]
Abstract
Nutrient sulfate is important for fetal development. The fetus has a limited capacity to generate sulfate and relies on maternal sulfate supplied via the placenta. The gestational age when fetal sulfate generation begins is unknown but would require cysteine dioxygenase (CDO1) which mediates a major step of sulfate production from cysteine. We investigated the ontogeny of Cdo1 mRNA expression in mouse fetal and placental tissues, which showed increasing levels from embryonic day 10.5 and was localised to the decidua and several fetal tissues including nasal cavities and brain. These findings suggest a role for Cdo1 in sulfate generation from mid-gestation.
Collapse
Affiliation(s)
- J Rakoczy
- Mater Research Institute, University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, University of Queensland, St. Lucia, Australia
| | - S Lee
- Mater Research Institute, University of Queensland, Woolloongabba, Australia
| | - S J Weerasekera
- Mater Research Institute, University of Queensland, Woolloongabba, Australia
| | - D G Simmons
- School of Biomedical Sciences, University of Queensland, St. Lucia, Australia
| | - P A Dawson
- Mater Research Institute, University of Queensland, Woolloongabba, Australia; School of Biomedical Sciences, University of Queensland, St. Lucia, Australia.
| |
Collapse
|