1
|
Wu X, Yu D, Ma Y, Fang X, Sun P. Function and therapeutic potential of Amuc_1100, an outer membrane protein of Akkermansia muciniphila: A review. Int J Biol Macromol 2025; 308:142442. [PMID: 40157674 DOI: 10.1016/j.ijbiomac.2025.142442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/16/2025] [Accepted: 03/21/2025] [Indexed: 04/01/2025]
Abstract
The gut microbiota-derived protein Amuc_1100, a key outer membrane component of Akkermansia muciniphila, has emerged as a groundbreaking therapeutic agent with unique structural and functional properties. Amuc_1100 exerts multifaceted immune-metabolic effects through novel mechanisms, including modulation of TLR2/4 and JAK/STAT pathways. This review highlights its unique multi-component structure that enables synergistic biological activity, and its pharmacological properties, which underlies its ability to enhance intestinal barrier integrity, restore microbiota balance, and suppress systemic inflammation. Crucially, Amuc_1100 demonstrates unprecedented therapeutic versatility across both intestinal disorders (e.g., inflammatory bowel disease, antibiotic-associated diarrhea) and extraintestinal conditions-notably improving neuropsychiatric symptoms via gut-serotonin axis regulation, combating cancer through CD8+ T cell activation, and mitigating cardiotoxicity via gut-heart immune crosstalk. Emerging innovations in targeted delivery systems, including gut-retentive nano-formulations and engineered probiotic vectors, further amplify its clinical potential. We critically evaluate recent advances distinguishing Amuc_1100's mechanisms from live bacterial interventions. By synthesizing evidence from preclinical models, this work positions Amuc_1100 as a prototype for next-generation microbiome-derived therapeutics, bridging microbial ecology with precision medicine.
Collapse
Affiliation(s)
- Xuhui Wu
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, PR China
| | - Yunkun Ma
- Department of Gastroenterology and Digestive Endoscopy Center, The Second Hospital of Jilin University, Changchun 130041, PR China
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, PR China.
| | - Pengda Sun
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun 130041, PR China.
| |
Collapse
|
2
|
Jin X, Huo J, Yao Y, Li R, Sun M, Li J, Wu Z. A multi-dimensional validation strategy of pharmacological effects of Radix Isatidis Mixtures against the co-infection of Mycoplasma gallisepticum and Escherichia coli in poultry. Poult Sci 2025; 104:104576. [PMID: 39616681 PMCID: PMC11648751 DOI: 10.1016/j.psj.2024.104576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/21/2024] [Accepted: 11/21/2024] [Indexed: 01/25/2025] Open
Abstract
Natural drugs possess exceptional pharmacological properties, yet their development is often hindered by a lack of clarity regarding the mechanisms of their pharmacological actions. Building on our previous research, we employed a co-infection model with Mycoplasma gallisepticum (MG) and Escherichia coli (E. coli) to investigate the pharmacological action of Radix Isatidis Mixtures (RIM). To further demonstrate the various mechanisms underlying the pharmacological effects of RIM, we conducted a validation study focusing on gene expression, protein interactions, metabolic pathways, and molecular docking. Through a multi-omics joint analysis network, we identified key targets and metabolites associated with co-infection and conducted targeted verification experiments with RIM aqueous extracts. The experimental results indicated that, compared to the co-infection group, the RIM treatment group significantly modulated the expression of select genes and proteins, particularly MMP2 and TLR4, with a high level of statistical significance (p < 0.01). At the metabolic level, the treatment group exhibited significantly reduced expression levels of Dopamine and γ-Aminobutyric acid. Notably, the molecular docking results highlighted compounds with the most favorable binding affinities: Salvianolic acid A (-10.1 kcal/mol), Licorice (-9.3 kcal/mol), and Isoglycyrrhiza (-8.7 kcal/mol). In conclusion, our multi-level experiments demonstrated that RIM possesses the characteristics of multi-pathway and multi-target treatment for co-infection.
Collapse
Affiliation(s)
- Xiaodi Jin
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Jinhai Huo
- Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, PR China
| | - Yecheng Yao
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Rui Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Mengqing Sun
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Jichang Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China
| | - Zhiyong Wu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Xiangfang District, Harbin 150030, PR China; Institute of Chinese Materia Medica, Heilongjiang Academy of Chinese Medicine Sciences, Harbin 150036, PR China.
| |
Collapse
|
3
|
Rahmati M, Moghtaderi H, Mohammadi S, Al-Harrasi A. Aryl hydrocarbon receptor dynamics in esophageal squamous cell carcinoma: From immune modulation to therapeutic opportunities. World J Exp Med 2024; 14:96269. [PMID: 39312702 PMCID: PMC11372732 DOI: 10.5493/wjem.v14.i3.96269] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/26/2024] [Accepted: 06/14/2024] [Indexed: 08/29/2024] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a substantial global health burden. Immune escape mechanisms are important in ESCC progression, enabling cancer cells to escape the surveillance of the host immune system. One key player in this process is the Aryl Hydrocarbon Receptor (AhR), which influences multiple cellular processes, including proliferation, differentiation, metabolism, and immune regulation. Dysregulated AhR signaling participates in ESCC development by stimulating carcinogenesis, epithelial-mesenchymal transition, and immune escape. Targeting AhR signaling is a potential therapeutic approach for ESCC, with AhR ligands showing efficacy in preclinical studies. Additionally, modification of AhR ligands and combination therapies present new opportunities for therapeutic intervention. This review aims to address the knowledge gap related to the role of AhR signaling in ESCC pathogenesis and immune escape.
Collapse
Affiliation(s)
- Mina Rahmati
- Biotechnology Research Center, Pasteur Institute of Iran, Tehran 1316943551, Tehran, Iran
| | - Hassan Moghtaderi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| | - Saeed Mohammadi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Nizwa 616, Ad Dakhiliyah, Oman
| |
Collapse
|
4
|
Devereaux J, Robinson AM, Stavely R, Davidson M, Dargahi N, Ephraim R, Kiatos D, Apostolopoulos V, Nurgali K. Alterations in tryptophan metabolism and de novo NAD + biosynthesis within the microbiota-gut-brain axis in chronic intestinal inflammation. Front Med (Lausanne) 2024; 11:1379335. [PMID: 39015786 PMCID: PMC11250461 DOI: 10.3389/fmed.2024.1379335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 06/17/2024] [Indexed: 07/18/2024] Open
Abstract
Background Inflammatory bowel disease is an incurable and idiopathic disease characterized by recurrent gastrointestinal tract inflammation. Tryptophan metabolism in mammalian cells and some gut microbes comprise intricate chemical networks facilitated by catalytic enzymes that affect the downstream metabolic pathways of de novo nicotinamide adenine dinucleotide (NAD+) synthesis. It is hypothesized that a correlation exists between tryptophan de novo NAD+ synthesis and chronic intestinal inflammation. Methods Transcriptome analysis was performed using high-throughput sequencing of mRNA extracted from the distal colon and brain tissue of Winnie mice with spontaneous chronic colitis and C57BL/6 littermates. Metabolites were assessed using ultra-fast liquid chromatography to determine differences in concentrations of tryptophan metabolites. To evaluate the relative abundance of gut microbial genera involved in tryptophan and nicotinamide metabolism, we performed 16S rRNA gene amplicon sequencing of fecal samples from C57BL/6 and Winnie mice. Results Tryptophan and nicotinamide metabolism-associated gene expression was altered in distal colons and brains of Winnie mice with chronic intestinal inflammation. Changes in these metabolic pathways were reflected by increases in colon tryptophan metabolites and decreases in brain tryptophan metabolites in Winnie mice. Furthermore, dysbiosis of gut microbiota involved in tryptophan and nicotinamide metabolism was evident in fecal samples from Winnie mice. Our findings shed light on the physiological alterations in tryptophan metabolism, specifically, its diversion from the serotonergic pathway toward the kynurenine pathway and consequential effects on de novo NAD+ synthesis in chronic intestinal inflammation. Conclusion The results of this study reveal differential expression of tryptophan and nicotinamide metabolism-associated genes in the distal colon and brain in Winnie mice with chronic intestinal inflammation. These data provide evidence supporting the role of tryptophan metabolism and de novo NAD+ synthesis in IBD pathophysiology.
Collapse
Affiliation(s)
- Jeannie Devereaux
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ainsley M. Robinson
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- School of Rural Health, La Trobe University, Melbourne, VIC, Australia
- Department of Medicine, Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
| | - Rhian Stavely
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Pediatric Surgery, Pediatric Surgery Research Laboratories, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Majid Davidson
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Narges Dargahi
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Ramya Ephraim
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Dimitros Kiatos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Medicine, Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Immunology Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Department of Medicine, Western Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Regenerative Medicine and Stem Cells Program, Australian Institute of Musculoskeletal Science (AIMSS), Melbourne, VIC, Australia
| |
Collapse
|
5
|
Ding Y, Yanagi K, Yang F, Callaway E, Cheng C, Hensel ME, Menon R, Alaniz RC, Lee K, Jayaraman A. Oral supplementation of gut microbial metabolite indole-3-acetate alleviates diet-induced steatosis and inflammation in mice. eLife 2024; 12:RP87458. [PMID: 38412016 PMCID: PMC10942630 DOI: 10.7554/elife.87458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in Western countries. There is growing evidence that dysbiosis of the intestinal microbiota and disruption of microbiota-host interactions contribute to the pathology of NAFLD. We previously demonstrated that gut microbiota-derived tryptophan metabolite indole-3-acetate (I3A) was decreased in both cecum and liver of high-fat diet-fed mice and attenuated the expression of inflammatory cytokines in macrophages and Tnfa and fatty acid-induced inflammatory responses in an aryl-hydrocarbon receptor (AhR)-dependent manner in hepatocytes. In this study, we investigated the effect of orally administered I3A in a mouse model of diet-induced NAFLD. Western diet (WD)-fed mice given sugar water (SW) with I3A showed dramatically decreased serum ALT, hepatic triglycerides (TG), liver steatosis, hepatocyte ballooning, lobular inflammation, and hepatic production of inflammatory cytokines, compared to WD-fed mice given only SW. Metagenomic analysis show that I3A administration did not significantly modify the intestinal microbiome, suggesting that I3A's beneficial effects likely reflect the metabolite's direct actions on the liver. Administration of I3A partially reversed WD-induced alterations of liver metabolome and proteome, notably, decreasing expression of several enzymes in hepatic lipogenesis and β-oxidation. Mechanistically, we also show that AMP-activated protein kinase (AMPK) mediates the anti-inflammatory effects of I3A in macrophages. The potency of I3A in alleviating liver steatosis and inflammation clearly demonstrates its potential as a therapeutic modality for preventing the progression of steatosis to non-alcoholic steatohepatitis (NASH).
Collapse
Affiliation(s)
- Yufang Ding
- Artie McFerrin Department of Chemical Engineering, Texas A&M UniversityCollege StationUnited States
| | - Karin Yanagi
- Department of Chemical and Biological Engineering, Tufts UniversityMedfordUnited States
| | - Fang Yang
- Artie McFerrin Department of Chemical Engineering, Texas A&M UniversityCollege StationUnited States
| | - Evelyn Callaway
- Artie McFerrin Department of Chemical Engineering, Texas A&M UniversityCollege StationUnited States
| | - Clint Cheng
- Artie McFerrin Department of Chemical Engineering, Texas A&M UniversityCollege StationUnited States
| | - Martha E Hensel
- Department of Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M UniversityCollege StationUnited States
| | - Rani Menon
- Artie McFerrin Department of Chemical Engineering, Texas A&M UniversityCollege StationUnited States
| | - Robert C Alaniz
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas Health Science Center, Texas A&M UniversityBryanUnited States
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts UniversityMedfordUnited States
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering, Texas A&M UniversityCollege StationUnited States
- Department of Microbial Pathogenesis and Immunology, College of Medicine, Texas Health Science Center, Texas A&M UniversityBryanUnited States
| |
Collapse
|
6
|
Kang C, Zhang J, Xue M, Li X, Ding D, Wang Y, Jiang S, Chu FF, Gao Q, Zhang M. Metabolomics analyses of cancer tissue from patients with colorectal cancer. Mol Med Rep 2023; 28:219. [PMID: 37772396 PMCID: PMC10568249 DOI: 10.3892/mmr.2023.13106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 07/31/2023] [Indexed: 09/30/2023] Open
Abstract
The alteration of metabolism is essential for the initiation and progression of numerous types of cancer, including colorectal cancer (CRC). Metabolomics has been used to study CRC. At present, the reprogramming of the metabolism in CRC remains to be fully elucidated. In the present study, comprehensive untargeted metabolomics analysis was performed on the paired CRC tissues and adjacent normal tissues from patients with CRC (n=35) using ultra‑high‑performance liquid chromatography‑mass spectrometry. Subsequently, bioinformatic analysis was performed on the differentially expressed metabolites. The changes in these differential metabolites were compared among groups of patients based on sex, anatomical tumor location, grade of tumor differentiation and stage of disease. A total of 927 metabolites were detected in the tissue samples, and 24 metabolites in the CRC tissue were significantly different compared with the adjacent normal tissue. The present study revealed that the levels of three amino acid metabolites were increased in the CRC tissue, specifically, N‑α‑acetyl‑ε‑(2‑propenal)‑Lys, cyclo(Glu‑Glu) and cyclo(Phe‑Glu). The metabolites with decreased levels in the CRC tissue included quinaldic acid (also referred to as quinoline‑2‑carboxilic acid), 17α‑ and 17β‑estradiol, which are associated with tumor suppression activities, as well as other metabolites such as, anhydro‑β‑glucose, Asp‑Arg, lysophosphatidylcholine, lysophosphatidylethanolamine (lysoPE), lysophosphatidylinositol, carnitine, 5'‑deoxy‑5'‑(methylthio) adenosine, 2'‑deoxyinosine‑5'‑monophosphate and thiamine monophosphate. There was no difference in the levels of the differential metabolites between male and female patients. The differentiation of CRC also showed no impact on the levels of the differential metabolites. The levels of lysoPE were increased in the right side of the colon compared with the left side of the colon and rectum. Analysis of the different tumor stages indicated that 2‑aminobenzenesulfonic acid, P‑sulfanilic acid and quinoline‑4‑carboxylic acid were decreased in stage I CRC tissue compared with stage II, III and IV CRC tissue. The levels of N‑α‑acetyl‑ε‑(2‑propenal)‑Lys, methylcysteine and 5'‑deoxy‑5'‑(methylthio) adenosine varied at different stages of tumorigenesis. These differential metabolites were implicated in multiple metabolism pathways, including carbohydrate, amino acid, lipid, nucleotide and hormone. In conclusion, the present study demonstrated that CRC tumors had altered metabolites compared with normal tissue. The data from the metabolic profile of CRC tissues in the present study provided supportive evidence to understand tumorigenesis.
Collapse
Affiliation(s)
- Chunbo Kang
- Department of Surgery, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Jie Zhang
- Department of Surgery, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Mei Xue
- Department of Gastroenterology and Hepatology, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Xiaowei Li
- Department of Surgery, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Danyang Ding
- Department of Surgery, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Ye Wang
- Department of Surgery, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Shujing Jiang
- Department of Acute Medicine, Queen Elizabeth Hospital, London SE18 4QH, UK
| | - Fong-Fong Chu
- Department of Cancer Genetics and Epigenetics, Beckman Research Institute of The City of Hope, Duarte, CA 91010, USA
| | - Qiang Gao
- Department of Gastroenterology and Hepatology, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| | - Mengqiao Zhang
- Department of Gastroenterology and Hepatology, Center of Gastrointestinal Rehabilitation, Beijing Rehabilitation Hospital, Capital Medical University, Beijing 100144, P.R. China
| |
Collapse
|
7
|
Wang AJ, Song D, Hong YM, Liu NN. Multi-omics insights into the interplay between gut microbiota and colorectal cancer in the "microworld" age. Mol Omics 2023; 19:283-296. [PMID: 36916422 DOI: 10.1039/d2mo00288d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Colorectal cancer (CRC) is a multifactorial heterogeneous disease largely due to both genetic predisposition and environmental factors including the gut microbiota, a dynamic microbial ecosystem inhabiting the gastrointestinal tract. Elucidation of the molecular mechanisms by which the gut microbiota interacts with the host may contribute to the pathogenesis, diagnosis, and promotion of CRC. However, deciphering the influence of genetic variants and interactions with the gut microbial ecosystem is rather challenging. Despite recent advancements in single omics analysis, the application of multi-omics approaches to integrate multiple layers of information in the microbiome and host to introduce effective prevention, diagnosis, and treatment strategies is still in its infancy. Here, we integrate host- and microbe-based multi-omics studies, respectively, to provide a strategy to explore potential causal relationships between gut microbiota and colorectal cancer. Specifically, we summarize the recent multi-omics studies such as metagenomics combined with metabolomics and metagenomics combined with genomics. Meanwhile, the sample size and sample types commonly used in multi-omics research, as well as the methods of data analysis, were also generalized. We highlight multiple layers of information from multi-omics that need to be verified by different types of models. Together, this review provides new insights into the clinical diagnosis and treatment of colorectal cancer patients.
Collapse
Affiliation(s)
- An-Jun Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Dingka Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Yue-Mei Hong
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| |
Collapse
|
8
|
Gamage HKAH, Robinson KJ, Luu L, Paulsen IT, Laird AS. Machado Joseph disease severity is linked with gut microbiota alterations in transgenic mice. Neurobiol Dis 2023; 179:106051. [PMID: 36822548 DOI: 10.1016/j.nbd.2023.106051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/06/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023] Open
Abstract
Emerging evidence suggests the presence of bidirectional interactions between the central nervous system and gut microbiota that may contribute to the pathogenesis of neurodegenerative diseases. However, the potential role of gut microbes in forms of spinocerebellar ataxia, such as the fatal neurodegenerative disease Machado Joseph disease (MJD), remains unexplored. Here, we examined whether gut microbiota alterations may be an early disease phenotype of MJD. We profiled the gut microbiota of male and female transgenic MJD mice (CMVMJD135) expressing human ATXN3 with expanded CAG repeats (133-143 CAG) at pre-symptomatic, symptomatic and well-established stages of the disease (7, 11 and 15 weeks of age, respectively). We compared these profiles with the gut microbiota of male and female wild-type (WT) littermate control mice at same ages. Correlation network analyses were employed to explore the relevance of microbiota changes to disease progression. The results demontrated distinct sex-dependent effects in disease development whereby male MJD mice displayed earlier motor impairments than female MJD mice. The gut microbiota community structure and composition also demonstrated sex-specific differences between MJD and WT mice. In both male and female MJD mice, the shifts in the microbiota were present by 7 weeks, before the onset of any symptoms. These pre-symptomatic microbial changes correlated with the severity of neurological impairments present at later stages of the disease. Previous efforts towards developing treatments for MJD have failed to yield meaningful outcomes. Our study reports a novel relationship between the gut microbiota and MJD development and severity. Elucidating how gut microbes are involved in MJD pathogenesis may offer new and efficacious treatment strategies for this currently untreatable disease.
Collapse
Affiliation(s)
- Hasinika K A H Gamage
- School of Natural Sciences, Macquarie University, NSW 2109, Australia; ARC Training Centre for Facilitated Advancement of Australia's Bioactives, Macquarie University, NSW 2109, Australia
| | - Katherine J Robinson
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Luan Luu
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia
| | - Ian T Paulsen
- School of Natural Sciences, Macquarie University, NSW 2109, Australia; ARC Training Centre for Facilitated Advancement of Australia's Bioactives, Macquarie University, NSW 2109, Australia; ARC Centre of Excellence in Synthetic Biology, Macquarie University, NSW 2109, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW 2109, Australia.
| |
Collapse
|
9
|
Madison CA, Hillbrick L, Kuempel J, Albrecht GL, Landrock KK, Safe S, Chapkin RS, Eitan S. Intestinal epithelium aryl hydrocarbon receptor is involved in stress sensitivity and maintaining depressive symptoms. Behav Brain Res 2023; 440:114256. [PMID: 36528169 PMCID: PMC9839636 DOI: 10.1016/j.bbr.2022.114256] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/03/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a key regulator in the microbiome-gut-brain axis, and AhR-active microbial metabolites modulate multiple neuronal responses. We recently demonstrated that 3,3'-diindolylmethane (DIM) and 1,4-dihydroxy-2-naphthoic acid (DHNA), two selective AhR modulators (SAhRMs), act as antidepressants in female mice. Thus, to examine the role of intestinal AhR in depression, anxiety, and spatial learning, this study employed transgenic mice in which the AhR was knockout only in the intestinal epithelium (AhRΔIEC). Additionally, this study examined whether the antidepressant effects of dietary DIM and DHNA is mediated by intestinal AhR. AhRΔIEC and WT female mice were fed daily with vehicle, 20 mg/kg DIM or DHNA for three weeks prior to four weeks of unpredictable chronic mild stress (UCMS). Mice were examined for weight gain, anhedonia-like behavior (sucrose preference test), anxiety levels (open field, light/dark, elevated plus maze, novelty-induced hypophagia, and marble burying tests), and spatial learning (Morris water maze). UCMS reduced weight gain in AhRΔIECs, but not WTs. Moreover, UCMS initially reduced sucrose preference in both AhRΔIECs and WTs, but over 4 weeks of UCMS, AhRΔIECs develop resilience to UCMS-induced anhedonia. Additionally, AhRΔIECs exhibit slightly reduced anxiety in certain tests and faster spatial learning. DIM and DHNA acted as antidepressants in both AhRΔIECs and WTs. Thus, this study suggests that intestinal AhR plays differential roles, mitigating stress effects on weight gain, and increasing stress effects on mood. However, the site of antidepressant action of SAhRMs, such as DIM and DHNA, is not dependent on the expression of intestinal AhR.
Collapse
Affiliation(s)
- Caitlin A Madison
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Lauren Hillbrick
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Jacob Kuempel
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Georgia Lee Albrecht
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA
| | - Kerstin K Landrock
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, 4466 TAMU, College Station, TX 77843-4466, USA
| | - Robert S Chapkin
- Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
| | - Shoshana Eitan
- Behavioral and Cellular Neuroscience, Department of Psychological and Brain Sciences, Texas A&M University, 4235 TAMU, College Station, TX 77843, USA.
| |
Collapse
|
10
|
Alvik K, Shao P, Hutin D, Baglole C, Grant DM, Matthews J. Increased sensitivity to chemically induced colitis in mice harboring a DNA-binding deficient aryl hydrocarbon receptor. Toxicol Sci 2023; 191:321-331. [PMID: 36519841 PMCID: PMC9936212 DOI: 10.1093/toxsci/kfac132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The aryl hydrocarbon receptor (AHR), a transcription factor best known for mediating toxic responses of environmental pollutants, also integrates metabolic signals to promote anti-inflammatory responses, intestinal homeostasis, and maintain barrier integrity. AHR regulates its target genes through direct DNA-binding to aryl hydrocarbon response elements (AHREs) but also through tethering to other transcription factors in a DNA-binding independent manner. However, it is not known if AHR's anti-inflammatory role in the gut requires its ability to bind to AHREs. To test this, we determined the sensitivity of Ahrdbd/dbd mice, a genetically modified mouse line that express an AHR protein incapable of binding to AHREs, to dextran sulfate sodium (DSS)-induced colitis. Ahrdbd/dbd mice exhibited more severe symptoms of intestinal inflammation than Ahr+/+ mice. None of the Ahrdbd/dbd mice survived after the 5-day DSS followed by 7-day washout period. By day 6, the Ahrdbd/dbd mice had severe body weight loss, shortening of the colon, higher disease index scores, enlarged spleens, and increased expression of several inflammation genes, including interleukin 1b (Il-1b), Il-6, Il-17, C-x-c motif chemokine ligand 1 (Cxcl1), Cxcl2, Prostaglandin-endoperoxide synthase (Ptgs2), and lipocalin-2. Our findings show that AHR's DNA-binding domain and ability to bind to AHREs are required to reduce inflammation, maintain a healthy intestinal environment, and protect against DSS-induced colitis.
Collapse
Affiliation(s)
- Karoline Alvik
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Peng Shao
- Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S1A8, Canada
| | - David Hutin
- Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S1A8, Canada
| | - Carolyn Baglole
- Department of Medicine, McGill University, Montreal H4A3J1, Canada.,Department of Pathology, McGill University, Montreal H4A3J1, Canada.,Department of Pharmacology and Therapeutics, McGill University, Montreal H3G1Y6, Canada
| | - Denis M Grant
- Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S1A8, Canada
| | - Jason Matthews
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.,Department of Pharmacology and Toxicology, University of Toronto, Toronto M5S1A8, Canada
| |
Collapse
|
11
|
Merchak AR, Cahill HJ, Brown LC, Brown RM, Rivet-Noor C, Beiter RM, Slogar ER, Olgun DG, Gaultier A. The activity of the aryl hydrocarbon receptor in T cells tunes the gut microenvironment to sustain autoimmunity and neuroinflammation. PLoS Biol 2023; 21:e3002000. [PMID: 36787309 PMCID: PMC9928083 DOI: 10.1371/journal.pbio.3002000] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 01/13/2023] [Indexed: 02/15/2023] Open
Abstract
Multiple sclerosis (MS) is a T cell-driven autoimmune disease that attacks the myelin of the central nervous system (CNS) and currently has no cure. MS etiology is linked to both the gut flora and external environmental factors but this connection is not well understood. One immune system regulator responsive to nonpathogenic external stimuli is the aryl hydrocarbon receptor (AHR). The AHR, which binds diverse molecules present in the environment in barrier tissues, is a therapeutic target for MS. However, AHR's precise function in T lymphocytes, the orchestrators of MS, has not been described. Here, we show that in a mouse model of MS, T cell-specific Ahr knockout leads to recovery driven by a decrease in T cell fitness. At the mechanistic level, we demonstrate that the absence of AHR changes the gut microenvironment composition to generate metabolites that impact T cell viability, such as bile salts and short chain fatty acids. Our study demonstrates a newly emerging role for AHR in mediating the interdependence between T lymphocytes and the microbiota, while simultaneously identifying new potential molecular targets for the treatment of MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Andrea R. Merchak
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- Neuroscience Graduate Program, University of Virginia, Charlottesville Virginia, United States of America
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, United States of America
| | - Hannah J. Cahill
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Lucille C. Brown
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Ryan M. Brown
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- Neuroscience Graduate Program, University of Virginia, Charlottesville Virginia, United States of America
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, United States of America
| | - Courtney Rivet-Noor
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- Neuroscience Graduate Program, University of Virginia, Charlottesville Virginia, United States of America
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, United States of America
| | - Rebecca M. Beiter
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- Neuroscience Graduate Program, University of Virginia, Charlottesville Virginia, United States of America
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, United States of America
| | - Erica R. Slogar
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Deniz G. Olgun
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
| | - Alban Gaultier
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, United States of America
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
12
|
Jiang S, Miao Z. High-fat diet induces intestinal mucosal barrier dysfunction in ulcerative colitis: emerging mechanisms and dietary intervention perspective. Am J Transl Res 2023; 15:653-677. [PMID: 36915785 PMCID: PMC10006746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 01/09/2023] [Indexed: 03/16/2023]
Abstract
The incidence of ulcerative colitis (UC) is increasing worldwide, but its pathogenesis remains largely unclear. The intestinal mucosa is a barrier that maintains the stability of the body's internal environment, and dysfunction of this barrier leads to the occurrence and aggravation of UC. A high-fat diet (HFD) contains more animal fat and low fiber, and accumulating evidence has shown that long-term intake of an HFD is associated with UC. The mechanism linking an HFD with intestinal mucosal barrier disruption is multifactorial, and it typically involves microbiota dysbiosis and altered metabolism of fatty acids, bile acids, and tryptophan. Dysbiosis-induced metabolic changes can enhance intestinal permeability through multiple pathways. These changes modulate the programmed death of intestinal epithelial cells, inhibit the secretion of goblet cells and Paneth cells, and impair intercellular interactions. Gut metabolites can also induce intestinal immune imbalance by stimulating multiple proinflammatory signaling pathways and decreasing the effect of anti-inflammatory immune cells. In this review, we critically analyze the molecular mechanisms by which an HFD disrupts the intestinal mucosal barrier (IMB) and contributes to the development of UC. We also discuss the application and future direction of dietary intervention in the treatment of the IMB and prevention of UC.
Collapse
Affiliation(s)
- Shijing Jiang
- First Clinical Medical College, Nanjing University of Chinese Medicine Nanjing, Jiangsu, China
| | - Zhiwei Miao
- Department of Gastroenterology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine Zhangjiagang, Suzhou, Jiangsu, China
| |
Collapse
|
13
|
Davis EW, Wong CP, Arnold HK, Kasschau K, Gaulke CA, Sharpton TJ, Ho E. Age and micronutrient effects on the microbiome in a mouse model of zinc depletion and supplementation. PLoS One 2022; 17:e0275352. [PMID: 36534653 PMCID: PMC9762596 DOI: 10.1371/journal.pone.0275352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022] Open
Abstract
Older adult populations are at risk for zinc deficiency, which may predispose them to immune dysfunction and age-related chronic inflammation that drives myriad diseases and disorders. Recent work also implicates the gut microbiome in the onset and severity of age-related inflammation, indicating that dietary zinc status and the gut microbiome may interact to impact age-related host immunity. We hypothesize that age-related alterations in the gut microbiome contribute to the demonstrated zinc deficits in host zinc levels and increased inflammation. We tested this hypothesis with a multifactor two-part study design in a C57BL/6 mouse model. The two studies included young (2 month old) and aged (24 month old) mice fed either (1) a zinc adequate or zinc supplemented diet, or (2) a zinc adequate or marginal zinc deficient diet, respectively. Overall microbiome composition did not significantly change with zinc status; beta diversity was driven almost exclusively by age effects. Microbiome differences due to age are evident at all taxonomic levels, with more than half of all taxonomic units significantly different. Furthermore, we found 150 out of 186 genera were significantly different between the two age groups, with Bacteriodes and Parabacteroides being the primary taxa of young and old mice, respectively. These data suggest that modulating individual micronutrient concentrations does not lead to comprehensive microbiome shifts, but rather affects specific components of the gut microbiome. However, a phylogenetic agglomeration technique (ClaaTU) revealed phylogenetic clades that respond to modulation of dietary zinc status and inflammation state in an age-dependent manner. Collectively, these results suggest that a complex interplay exists between host age, gut microbiome composition, and dietary zinc status.
Collapse
Affiliation(s)
- Edward W. Davis
- Center for Quantitative Life Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Carmen P. Wong
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, United States of America
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, United States of America
| | - Holly K. Arnold
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
| | - Kristin Kasschau
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
| | - Christopher A. Gaulke
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
| | - Thomas J. Sharpton
- Department of Microbiology, Oregon State University, Corvallis, Oregon, United States of America
- Department of Statistics, Oregon State University, Corvallis, Oregon, United States of America
| | - Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon, United States of America
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, Oregon, United States of America
- * E-mail:
| |
Collapse
|
14
|
Han Y, Li L, Wang B. Role of Akkermansia muciniphila in the development of nonalcoholic fatty liver disease: current knowledge and perspectives. Front Med 2022; 16:667-685. [PMID: 36318353 DOI: 10.1007/s11684-022-0960-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/06/2022] [Indexed: 11/19/2022]
|
15
|
Letsinger AC, Yang F, Menon R, Little-Letsinger SE, Granados JZ, Breidenbach B, Iyer AR, Padovani TC, Nagel EC, Jayaraman A, Lightfoot JT. Reduced Wheel Running via a High-Fat Diet Is Reversed by a Chow Diet with No Added Benefit from Fecal Microbial Transplants. Med Sci Sports Exerc 2022; 54:1437-1447. [PMID: 35969165 DOI: 10.1249/mss.0000000000002941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE Chronic overfeeding via a high-fat/high-sugar (HFHS) diet decreases wheel running and substantially alters the gut metabolome of C57BL/6J mice. In this study, we tested the hypothesis that fecal microbial transplants can modulate the effect of diet on wheel running. METHODS Singly housed, 6-wk-old male C57BL/6J mice were fed either a grain-based diet (CHOW) or HFHS diet and provided a running wheel for 13 wk. Low-active, HFHS-exposed mice were then either switched to a CHOW diet and given an oral fecal microbial transplant from mice fed the CHOW diet, switched to a CHOW diet and given a sham transplant, or remained on the HFHS diet and given a fecal microbial transplant from mice fed the CHOW diet. Total wheel running, nutrient intake, body composition, fecal microbial composition, fecal metabolite composition, and liver steatosis were measured at various times throughout the study. RESULTS We found that an HFHS diet decreases wheel running activity, increases body fat, and decreases microbial alpha diversity compared with a CHOW diet. Improvements in wheel running, body composition, and microbial alpha diversity were accomplished within 2 wk for mice switched from an HFHS diet to a CHOW diet with no clear evidence of an added benefit from fecal transplants. A fecal transplant from mice fed a CHOW diet without altering diet did not improve wheel running or body composition. Wheel running, body composition, fecal microbial composition, fecal metabolite composition, and liver steatosis percentage were primarily determined by diet. CONCLUSIONS Our results suggest that diet is a primary mediator of wheel running with no clear effect from fecal microbial transplants.
Collapse
Affiliation(s)
- Ayland C Letsinger
- The Department of Health Kinesiology, Texas A&M University, College Station, TX
| | - Fang Yang
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX
| | - Rani Menon
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX
| | | | - Jorge Z Granados
- The Department of Health Kinesiology, Texas A&M University, College Station, TX
| | - Brianne Breidenbach
- The Department of Health Kinesiology, Texas A&M University, College Station, TX
| | - Anjushree R Iyer
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX
| | | | - Edward C Nagel
- The Department of Health Kinesiology, Texas A&M University, College Station, TX
| | - Arul Jayaraman
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX
| | - J Timothy Lightfoot
- The Department of Health Kinesiology, Texas A&M University, College Station, TX
| |
Collapse
|
16
|
Squire N, Lux C, Tolbert K, Lidbury J, Sun X, Suchodolski JS. Characterization of the Fecal Microbiome in Dogs Receiving Medical Management for Congenital Portosystemic Shunts. Front Vet Sci 2022; 9:897760. [PMID: 35968011 PMCID: PMC9366551 DOI: 10.3389/fvets.2022.897760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
Background The GI microbiome has not been characterized in dogs being medically managed for congenital portosystemic shunts (CPSS). Objectives To characterize the fecal microbiome in a population of dogs being medically managed for CPSS. Animals 27 client-owned dogs. Methods Prospective cohort study enrollment of fecal samples was performed with follow-up data collected retrospectively. The overall fecal dysbiosis index (DI) and individual bacterial abundances were determined using real-time qPCR. Medical management, clinical findings, clinicopathologic, and outcome variables were collected, and logistic regression analyses were performed to evaluate associations between these variables and overall DI and bacterial abundances. Numerical variables were evaluated with general linear models for normality and equal variance using Shapiro-Wilk test and Levene's test, respectively. Results All dogs were administered a hepatic diet and lactulose, while antibiotics were used in 22 (81.5%) and acid suppressants in 7 (25.9%). Seventeen dogs (63.0%) had a DI >2. The median DI in this population was 3.02 (range 4.23–8.42), and the median DI in dogs receiving and not receiving antibiotics was 4.3 (range −4.23–8.42) and 1.52 (range −1.62–5.43), respectively. No significant association between any of the analyzed variables and the DI was identified. There was a significant association between the use of metronidazole and a larger abundance of E. coli (p = 0.024). Conclusions and Clinical Importance Dysbiosis appears to be common in dogs that are being medically managed for CPSS, though the clinical significance remains unclear.
Collapse
Affiliation(s)
- Nathan Squire
- Department of Small Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Cassie Lux
- Department of Small Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
- *Correspondence: Cassie Lux
| | - Katie Tolbert
- Department of Veterinary Small Animal Clinical Sciences, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX, United States
| | - Jonathan Lidbury
- Department of Veterinary Small Animal Clinical Sciences, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX, United States
| | - Xiaocun Sun
- Office of Information and Technology, University of Tennessee Knoxville, Knoxville, TN, United States
| | - Jan S. Suchodolski
- Department of Veterinary Small Animal Clinical Sciences, Texas A&M College of Veterinary Medicine and Biomedical Sciences, College Station, TX, United States
| |
Collapse
|
17
|
Li J, Zhang AH, Wu FF, Wang XJ. Alterations in the Gut Microbiota and Their Metabolites in Colorectal Cancer: Recent Progress and Future Prospects. Front Oncol 2022; 12:841552. [PMID: 35223525 PMCID: PMC8875205 DOI: 10.3389/fonc.2022.841552] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/18/2022] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer morbidity and mortality worldwide. The etiology and pathogenesis of CRC remain unclear. A growing body of evidence suggests dysbiosis of gut bacteria can contribute to the occurrence and development of CRC by generating harmful metabolites and changing host physiological processes. Metabolomics, a systems biology method, will systematically study the changes in metabolites in the physiological processes of the body, eventually playing a significant role in the detection of metabolic biomarkers and improving disease diagnosis and treatment. Metabolomics, in particular, has been highly beneficial in tracking microbially derived metabolites, which has substantially advanced our comprehension of host-microbiota metabolic interactions in CRC. This paper has briefly compiled recent research progress of the alterations of intestinal flora and its metabolites associated with CRC and the application of association analysis of metabolomics and gut microbiome in the diagnosis, prevention, and treatment of CRC; furthermore, we discuss the prospects for the problems and development direction of this association analysis in the study of CRC. Gut microbiota and their metabolites influence the progression and causation of CRC, and the association analysis of metabolomics and gut microbiome will provide novel strategies for the prevention, diagnosis, and therapy of CRC.
Collapse
Affiliation(s)
- Jing Li
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
- National Chinmedomics Research Center, National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ai-hua Zhang
- National Chinmedomics Research Center, National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fang-fang Wu
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
| | - Xi-jun Wang
- National Engineering Laboratory for the Development of Southwestern Endangered Medicinal Materials, Guangxi Botanical Garden of Medicinal Plant, Nanning, China
- National Chinmedomics Research Center, National Traditional Chinese Medicine (TCM) Key Laboratory of Serum Pharmacochemistry, Functional Metabolomics Laboratory, Department of Pharmaceutical Analysis, Heilongjiang University of Chinese Medicine, Harbin, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
| |
Collapse
|
18
|
Qi M, Cao Z, Shang P, Zhang H, Hussain R, Mehmood K, Chang Z, Wu Q, Dong H. Comparative analysis of fecal microbiota composition diversity in Tibetan piglets suffering from diarrheagenic Escherichia coli (DEC). Microb Pathog 2021; 158:105106. [PMID: 34311015 DOI: 10.1016/j.micpath.2021.105106] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/29/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023]
Abstract
This study was ascertained to investigate the adverse effects of pathogenic E. coli on gut microbiota of Tibetan piglets with history of yellow and white dysentery. For this purpose, a total of 18 fecal samples were collected from infected and healthy Tibetan piglets for 16S rRNA gene amplification and sequencing of V3-V4 region. Results showed that Firmicutes, Bacteroidia Fusobacteriota, Proteobacteria and Actinobacteriota were the predominant bacteria in Tibetan piglets at the level of phylum classification. Results on classification at family level showed that Lactobacillus, Bacteroidota, Fusobacteriota and Enterobacteriaceae were the dominant bacteria. Results on classification of bacteria at phylum level compared with normal piglets indicated that Bacteroidota, Actinobacteriota, Euryarchaota and Spirochaetota in fecal microbial community in Tibetan piglets showing yellow dysenteric and diarrhea group were significantly decreased (P ≤ 0.05). Compared with the feces of healthy Tibetan piglets, the abundance of Escherichia-Shigella, Lactobacillus and Enterococcus increased significantly in feces of Tibetan piglets having yellow dysentery and white dysentery. Moreover, results exhibited that the Proteobacteria and Fusobacteriota were significantly increased (P ≤ 0.05) suggesting dominant microbial community. Results revealed that E. coli induced different pathological alterations in intestine including damage to intestinal epithelial cells, infiltration of inflammatory cells, presence of red blood cells in spaces of tissues, hemorrhages and necrosis of intestinal villi in piglets with history of yellow dysentery. This study for the first time reported the composition, characteristics, and differences of the fecal microflora diversity of Tibetan piglets with yellow and white dysentery in Qinghai-Tibet Plateau, which can provide a suitable support for effective control of diarrhoeal disease in these animals.
Collapse
Affiliation(s)
- Ming Qi
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Zhipeng Cao
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Peng Shang
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Riaz Hussain
- Department of Pathology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Khalid Mehmood
- Department of Clinical Medicine and Surgery, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Zhenyu Chang
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Qingxia Wu
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Hailong Dong
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China.
| |
Collapse
|
19
|
Yin J, Song Y, Hu Y, Wang Y, Zhang B, Wang J, Ji X, Wang S. Dose-Dependent Beneficial Effects of Tryptophan and Its Derived Metabolites on Akkermansia In Vitro: A Preliminary Prospective Study. Microorganisms 2021; 9:microorganisms9071511. [PMID: 34361945 PMCID: PMC8305782 DOI: 10.3390/microorganisms9071511] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/14/2022] Open
Abstract
Akkermansia muciniphila, a potential probiotic, has been proven to lessen the effects of several diseases. As established, the relative abundance of Akkermansia is positively correlated with tryptophan metabolism. However, the reciprocal interaction between tryptophan and Akkemansia is still unclear. Herein, for the first time, the possible effects of tryptophan and its derived metabolites on A. muciniphila were preliminarily investigated, including growth, physiological function, and metabolism. Obtained results suggested that 0.4 g/L of tryptophan treatment could significantly promote the growth of A. muciniphila. Notably, when grown in BHI with 0.8 g/L of tryptophan, the hydrophobicity and adhesion of A. muciniphila were significantly improved, potentially due to the increase in the rate of cell division. Furthermore, A. muciniphila metabolized tryptophan to indole, indole-3-acetic acid, indole-3-carboxaldehyde, and indole-3-lactic acid. Indoles produced by gut microbiota could significantly promote the growth of A. muciniphila. These results could provide a valuable reference for future research on the relationship between tryptophan metabolism and A. muciniphila.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Shuo Wang
- Correspondence: ; Tel.: +86-22-85358445
| |
Collapse
|
20
|
Duong HT, Wu Y, Sutor A, Burek BO, Hollmann F, Bloh JZ. Intensification of Photobiocatalytic Decarboxylation of Fatty Acids for the Production of Biodiesel. CHEMSUSCHEM 2021; 14:1053-1056. [PMID: 33528107 PMCID: PMC7986711 DOI: 10.1002/cssc.202002957] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/21/2021] [Indexed: 05/16/2023]
Abstract
Light-driven biocatalytic processes are notoriously hampered by poor penetration of light into the turbid reaction media. In this study, wirelessly powered light-emitting diodes are found to represent an efficient and scalable approach for process intensification of the photobiosynthetic production of diesel alkanes from renewable fatty acids.
Collapse
Affiliation(s)
- Hong T. Duong
- Chemical Technology GroupDECHEMA Research InstituteTheodor-Heuss-Allee 2560486Frankfurt am MainGermany
| | - Yinqi Wu
- Department of BiotechnologyDelft University of Technologyvan der Maasweg 92629HXDelftThe Netherlands
| | - Alexander Sutor
- Department Institute of Measurement and Sensor TechnologyUMIT – University for Health Sciences, Medical Informatics and Technology GmbHEduard-Wallnöfer-Zentrum 1, 16060Hall in TirolAustria
| | - Bastien O. Burek
- Chemical Technology GroupDECHEMA Research InstituteTheodor-Heuss-Allee 2560486Frankfurt am MainGermany
| | - Frank Hollmann
- Department of BiotechnologyDelft University of Technologyvan der Maasweg 92629HXDelftThe Netherlands
| | - Jonathan Z. Bloh
- Chemical Technology GroupDECHEMA Research InstituteTheodor-Heuss-Allee 2560486Frankfurt am MainGermany
| |
Collapse
|
21
|
Dong H, Liu B, Li A, Iqbal M, Mehmood K, Jamil T, Chang YF, Zhang H, Wu Q. Microbiome Analysis Reveals the Attenuation Effect of Lactobacillus From Yaks on Diarrhea via Modulation of Gut Microbiota. Front Cell Infect Microbiol 2021; 10:610781. [PMID: 33665171 PMCID: PMC7920975 DOI: 10.3389/fcimb.2020.610781] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Domestic yaks (Bos grunniens) are indigenous to the Tibetan Plateau and display a high diarrhea rate due to poor habitat and husbandry conditions. Lactobacillus has been shown to exert beneficial effects as antimicrobial, growth promotion, and gut microbiota in humans and/or murine models, but the relevant data regarding Lactobacillus isolated from yaks was unavailable. Therefore, this study aimed to investigate the effects of Lactobacillus from yaks on the intestinal microbial community in a mouse model and determine whether Lactobacillus supplementation contributed in alleviating diarrhea by modulating gut microbiota. A total of 12 ileac samples from four groups were collected for 16S rRNA gene amplicon sequencing of V3-V4 region. Results revealed that although Lactobacillus supplementation did not change the diversity of gut microbiota in mice, the proportion of some intestinal microbiota significantly changed. Specifically, the proportion of Lactobacillus and Sphingomonas in the Lactobacillus treated-group (L-group) were increased as compared to control group (C-group), whereas Pantoea, Cutibacterium, Glutamicibacter, Turicibacter, Globicatella, Microbacterium, Facklamia, unidentified_Corynebacteriaceae, Brachybacterium, and Staphylococcus were significantly decreased in the L-group. In contrast, Escherichia coli (E. coli) infection significantly decreased the proportion of beneficial bacteria such as Globicatella, Acinetobacter, Aerococcus, and Comamonas, while loads of pathogenic bacteria significantly increased including Roseburia and Megasphaera. Interestingly, Lactobacillus administration could ameliorate the microbial community structure of E. coli-induced diarrheal mice by reducing the relative abundance of pathogenic bacteria such as Paenibacillus, Aerococcus, Comamonas, Acinetobacter, Corynebacterium, Facklamia, and Globicatella. Results in this study revealed that Lactobacillus supplementation not only improved the gut microbiota but also alleviated diarrhea in mice, which may be mediated by modulating the composition and function of gut microbiota. Moreover, this study is expected to provide a new theoretical basis for the establishment of a preventive and treatment system for diarrhea in yaks.
Collapse
Affiliation(s)
- Hailong Dong
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| | - Bingxian Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Aoyun Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - Mudassar Iqbal
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Khalid Mehmood
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Tariq Jamil
- Institute of Bacterial Infections and Zoonoses, Friedrich-Loeffler-Institut, Jena, Germany
| | - Yung-Fu Chang
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, United States
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Qingxia Wu
- Animal Science College, Tibet Agriculture & Animal Husbandry University, Linzhi, China
| |
Collapse
|