1
|
Yang L, Hu M, Shao J. Integration of Gut Mycobiota and Oxidative Stress to Decipher the Roles of C-Type Lectin Receptors in Inflammatory Bowel Diseases. Immunol Invest 2024; 53:1177-1204. [PMID: 39115960 DOI: 10.1080/08820139.2024.2388164] [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] [Indexed: 08/10/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) and Crohn's disease (CD) are two subtypes of inflammatory bowel disease (IBD) with rapidly increased incidence worldwide. Although multiple factors contribute to the occurrence and progression of IBD, the role of intestinal fungal species (gut mycobiota) in regulating the severity of these conditions has been increasingly recognized. C-type lectin receptors (CLRs) on hematopoietic cells, including Dectin-1, Dectin-2, Dectin-3, Mincle and DC-SIGN, are a group of pattern recognition receptors (PRRs) that primarily recognize fungi and mediate defense responses, such as oxidative stress. Recent studies have demonstrated the indispensable role of CLRs in protecting the colon from intestinal inflammation and mucosal damage. METHODS AND RESULTS This review provides a comprehensive overview of the role of CLRs in the pathogenesis of IBD. Given the significant impact of mycobiota and oxidative stress in IBD, this review also discusses recent advancements in understanding how these factors exacerbate or ameliorate IBD. Furthermore, the latest developments in CLR-guided IBD therapy are examined to highlight the modulation of CLRs in fungal recognition and oxidative burst during the IBD process. CONCLUSION This review emphasizes the importance of CLRs in IBD, offering new perspectives on the etiology and therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Liu Yang
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui 230012, P. R. China
| | - Min Hu
- Department of pathology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui, P. R. China
| | - Jing Shao
- Laboratory of Anti-infection and Immunity, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei, Anhui 230012, P. R. China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui, P. R. China
| |
Collapse
|
2
|
Zhou Y, Komnick MR, Sepulveda F, Liu G, Nieves-Ortiz E, Meador K, Ndatabaye O, Fatkhullina A, Wu-Woods NJ, Naydenkov PM, Kent J, Christiansen N, Madariaga ML, Witkowski P, Ismagilov RF, Esterházy D. Inducible, but not constitutive, pancreatic REG/Reg isoforms are regulated by intestinal microbiota and pancreatic diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.619139. [PMID: 39484594 PMCID: PMC11526982 DOI: 10.1101/2024.10.18.619139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The REG / Reg gene locus encodes for a conserved family of potent antimicrobial but also pancreatitis-associated proteins. Here we investigated whether REG/Reg family members differ in their baseline expression levels and abilities to be regulated in the pancreas and gut upon perturbations. We found, in human and mouse, pancreas and gut differed in REG / Reg isoform levels and preferences, with duodenum most resembling the pancreas. Pancreatic acinar cells and intestinal enterocytes were the dominant REG producers. Intestinal symbiotic microbes regulated the expression of the same, select Reg members in gut and pancreas. These Reg members had the most STAT3-binding sites close to the transcription start sites and were partially IL-22 dependent. We thus categorized them as "inducible" and others as "constitutive". Indeed, also in models of pancreatic-ductal adenocarcinoma and pancreatitis, only inducible Reg members were upregulated in pancreas. While intestinal Reg expression remained unchanged upon pancreatic perturbation, pancreatitis altered the microbial composition of the duodenum and feces shortly after disease onset. Our study reveals differential usage and regulation of REG / Reg isoforms as a mechanism for tissue-specific innate immunity, highlights the intimate connection of pancreas and duodenum, and implies a gut-to-pancreas communication axis resulting in a coordinated Reg response.
Collapse
|
3
|
Pan S, Yan H, Zhu J, Ma Y, Wang P, Liu Y, Chen Z. GYY4137, as a slow-releasing H 2S donor, ameliorates sodium deoxycholate-induced chronic intestinal barrier injury and gut microbiota dysbiosis. Front Pharmacol 2024; 15:1476407. [PMID: 39508040 PMCID: PMC11539038 DOI: 10.3389/fphar.2024.1476407] [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: 08/05/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Numerous studies have revealed that a long-term high-fat diet can raise intestinal deoxycholate acid concentration, which can harm intestinal mucosal barrier function in several ways. This study aims to verify the protective effect of GYY4137, as a slow-releasing H2S donor, on microbiome disturbance and the chronic injury of the intestinal mucosal barrier function caused by sodium deoxycholate. Methods Caco-2 monolayer and mouse models were treated with a relatively high concentration of sodium deoxycholate (1.0 mM and 0.2%, respectively) for longer periods (32 h and 12 weeks, respectively) to understand the effects of GYY4137 on sodium deoxycholate-induced chronic intestinal barrier dysfunction and its fundamental mechanisms. Results A relatively long period of sodium deoxycholate treatment can remarkably increase the intestinal barrier permeability, alter the distribution and expression of tight junction proteins and generate the production of pro-inflammatory cytokines (TNF-α and IL-1β) in the Caco-2 monolayers and mouse models. Moreover, it can activate the MLCK-P-MLC2 pathway in the Caco-2 monolayers, which was further confirmed using RNA sequencing. The body weight, intestinal barrier histological score, and TUNEL index of sodium deoxycholate-treated mice worsened. In addition, an induced microbiome imbalance was observed in these mice. The above variations can be reversed with the administration of GYY4137. Conclusion This study demonstrates that GYY4137 ameliorates sodium deoxycholate-induced chronic intestinal barrier injury by restricting the MLCK-P-MLC2 pathway while elevating the expression level of tight junction proteins, anti-apoptosis and maintaining the microbiome's homeostasis.
Collapse
Affiliation(s)
- Shaorong Pan
- Department of Gastrointestinal Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Han Yan
- Department of Gastrointestinal Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Jing Zhu
- Department of Gastrointestinal Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Yuanyuan Ma
- Animal Experiment Center, Peking University First Hospital, Peking University, Beijing, China
| | - Pengyuan Wang
- Department of Gastrointestinal Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Yucun Liu
- Department of Gastrointestinal Surgery, Peking University First Hospital, Peking University, Beijing, China
| | - Zeyang Chen
- Department of Gastrointestinal Surgery, Peking University First Hospital, Peking University, Beijing, China
| |
Collapse
|
4
|
Kumar A, Quraishi MN, Al-Hassi HO, Elasrag M, Segal JP, Jain M, Steed H, Butterworth J, Farmer A, Mclaughlin J, Beggs AD, Brookes MJ. The Effect of Colesevelam on the Microbiome in Postoperative Crohn's Disease. Inflamm Bowel Dis 2024:izae230. [PMID: 39422655 DOI: 10.1093/ibd/izae230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND While surgery plays a pivotal role in the management of ileal Crohn's disease, the risk of endoscopic recurrence following an ileocaecal resection can be greater than 65% within 12 months of surgery. More than 90% of patients with Crohn's disease have a concomitant diagnosis of bile acid diarrhea following an ileal resection. This pilot study aimed to assess whether the use of bile acid sequestrants in patients with Crohn's disease who have undergone a primary terminal ileal resection with concomitant bile acid diarrhea can alter the microbiome and prevent disease recurrence. METHODS Patients with Crohn's disease who underwent a primary terminal ileal resection and had symptoms of diarrhea within 1-3 months of surgery underwent 75SeHCAT testing for bile acid diarrhea. If positive (75SeHCAT ≤ 15%), patients were treated with colesevelam and stool samples were collected at 4 weeks, 8 weeks, and 6-12 months posttreatment. If negative (75SeHCAT > 15%), treatment was not given and were reviewed in the clinic as per local guidelines. All patients underwent a 6-12 month postoperative colonoscopy where further stool samples and mucosal biopsies were taken. Disease activity was established using the endoscopic Rutgeert's score, with disease remission defined as Rutgeert's score RESULTS A total of 14 patients who completed the study, 10 of whom had a 75SeHCAT positive diagnosis of bile acid diarrhea and were started on treatment with colesevelam. Four patients did not require treatment as 3 were asymptomatic and 1 had a negative 75SeHCAT scan. Three of the fourteen patients had disease recurrence at their 6-12 month postoperative colonoscopy assessment, of which 1 patient was taking colesevelam and 2 patients were not taking colesevelam. A total of 44 fecal samples and 44 mucosal biopsies underwent 16S ribosomal RNA gene analysis to assess α/β-diversity and microbial composition. In the colesevelam treated patients there was no significant difference in α/β-diversity pre- and posttreatment. Pretreatment, the 3 most abundant bacterial classes in all patients were Bacteroidia, Clostridia, and Gammaproteobacteria. Following 6-12 months of treatment, out of the 9 patients on colesevelam, 5/9 (55.6%) had a reduction in Bacteroidia, 9/9 (100%) had an increase in Clostridia, and 7/9 (77.8%) had a reduction in Gammaproteobacteria. Of the 2 patients not given colesevelam, one showed a reduction in Bacteroidia, increase in Clostridia and a reduction in Gammaproteobacteria. CONCLUSIONS This small pilot study demonstrated that patients who were given colesevelam, were more likely to be in disease remission at their 6-12 months colonoscopy review compared with those not treated. Furthermore, treatment with colesevelam may have a role in altering the microbiome to help maintain remission states in postoperative Crohn's disease. Larger mechanistic studies are now needed to confirm these findings and demonstrate statistical significance as well as investigate whether this benefit may be present even in those patients with 75SeHCAT negative disease.
Collapse
Affiliation(s)
- Aditi Kumar
- Department of Gastroenterology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Mohammed Nabil Quraishi
- University of Birmingham Microbiome Treatment Centre, Birmingham, UK
- Department of Gastroenterology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Hafid O Al-Hassi
- Research Institute in Healthcare Science, University of Wolverhampton, Wolverhampton, UK
| | - Mohammed Elasrag
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Aston Medical School, College of Health and Life Sciences, Aston University, Birmingham, UK
| | - Jonathan P Segal
- Department of Gastroenterology, Royal Melbourne Hospital, Melbourne, VIC, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC, Australia
| | - Manushri Jain
- Department of Gastroenterology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | - Helen Steed
- Department of Gastroenterology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
- School of Medicine and Clinical Practice, Faculty of Sciences and Engineering, University of Wolverhampton, Wolverhampton, UK
| | | | - Adam Farmer
- Division of Gastroenterology & Hepatology, St Louis University Hospital, St Louis, MO, USA
| | - John Mclaughlin
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Department of Gastroenterology, Salford Royal Foundation Trust, Stott Lane, UK
| | - Andrew D Beggs
- University of Birmingham Microbiome Treatment Centre, Birmingham, UK
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Matthew J Brookes
- Department of Gastroenterology, The Royal Wolverhampton NHS Trust, Wolverhampton, UK
- School of Medicine and Clinical Practice, Faculty of Sciences and Engineering, University of Wolverhampton, Wolverhampton, UK
| |
Collapse
|
5
|
Salem MB, Elzallat M, Mostafa Mohammed D, Hammam OA, Tamim A Abdel-Wareth M, Hassan M. Helix pomatia mucin alleviates DSS-induced colitis in mice: Unraveling the cross talk between microbiota and intestinal chemokine. Heliyon 2024; 10:e37362. [PMID: 39296159 PMCID: PMC11407997 DOI: 10.1016/j.heliyon.2024.e37362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 08/17/2024] [Accepted: 09/02/2024] [Indexed: 09/21/2024] Open
Abstract
Gut microbiota imbalance and alterations in the chemokine-chemokine receptor interactions are pivotal in the initiation and advancement of ulcerative colitis (UC). The current UC treatments are prolonged, exhibit high recurrence rates, and may lead to colorectal cancer. So, this study explores the efficacy of Helix pomatia (H. pomatia) mucin in preventing DSS-induced UC. This research focuses on investigating the underlying mechanisms, such as oxidative stress, inflammation, and alterations in gut microbiota and chemokine-chemokine receptor interactions, to understand the anti-inflammatory and antioxidant characteristics of the mucin. Using 4 % DSS in drinking water, UC was induced in C57BL/6 mice. For seven days, mice were given oral doses of either H. pomatia mucin or sulfasalazine. The study assessed changes in oxidative stress, gut microbiota, and histopathology, along with expression of IL-6, CXCR4, CCR7, CXCL9, and CXCL10. The H. pomatia mucin exhibited unique contents, including high glycolic acid (200 ± 2.08 mg/L), collagen (88 ± 2.52 mg/L), allantoin (20 ± 2 mg/L), and concentrated vitamins and minerals. Treatment with H. pomatia mucin in high dose demonstrated reduction in DAI, an increase in fecal Firmicutes, and elevated expression of colonic CCR7, CXCL9, and CXCL10, accompanied by enhanced CXCR4 (75 %) and diminished IL-6 (1.33 %) immunostaining. It also alleviated oxidative stress, reduced fecal Bacteroidetes, and mitigated inflammation, indicating its potential efficacy against DSS-induced UC. In conclusion, H. pomatia mucin is a promising candidate that could be an effective adjuvant in the management and prophylaxis of UC.
Collapse
Affiliation(s)
- Maha B Salem
- Pharmacology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Mohamed Elzallat
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Dina Mostafa Mohammed
- Nutrition and Food Sciences Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Olfat A Hammam
- Pathology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | | | - Marwa Hassan
- Immunology Department, Theodor Bilharz Research Institute, Giza, Egypt
| |
Collapse
|
6
|
Li Y, Li J, Li S, Zhou S, Yang J, Xu K, Chen Y. Exploring the gut microbiota's crucial role in acute pancreatitis and the novel therapeutic potential of derived extracellular vesicles. Front Pharmacol 2024; 15:1437894. [PMID: 39130638 PMCID: PMC11310017 DOI: 10.3389/fphar.2024.1437894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
During acute pancreatitis, intestinal permeability increases due to intestinal motility dysfunction, microcirculatory disorders, and ischemia-reperfusion injury, and disturbances in the intestinal flora make bacterial translocation easier, which consequently leads to local or systemic complications such as pancreatic and peripancreatic necrotic infections, acute lung injury, systemic inflammatory response syndrome, and multiple organ dysfunction syndrome. Therefore, adjusting intestinal ecosystem balance may be a promising approach to control local and systemic complications of acute pancreatitis. In this paper, we reviewed the causes and manifestations of intestinal flora disorders during acute pancreatitis and their complications, focused on the reduction of acute pancreatitis and its complications by adjusting the intestinal microbial balance, and innovatively proposed the treatment of acute pancreatitis and its complications by gut microbiota-derived extracellular vesicles.
Collapse
Affiliation(s)
- Yijie Li
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jie Li
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Sen Li
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shumin Zhou
- Wenzhou Institute of Shanghai University, Wenzhou, China
| | - Jiahua Yang
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ke Xu
- Wenzhou Institute of Shanghai University, Wenzhou, China
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Yafeng Chen
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
7
|
Hernández-Rocha C, Turpin W, Borowski K, Stempak JM, Sabic K, Gettler K, Tastad C, Chasteau C, Korie U, Hanna M, Khan A, Mengesha E, Bitton A, Schwartz MB, Barrie A, Datta LW, Lazarev M, Brant SR, Rioux JD, McGovern DPB, Duerr RH, Schumm LP, Cho JH, Silverberg MS. After Surgically Induced Remission, Ileal and Colonic Mucosa-Associated Microbiota Predicts Crohn's Disease Recurrence. Clin Gastroenterol Hepatol 2024:S1542-3565(24)00592-5. [PMID: 38969076 DOI: 10.1016/j.cgh.2024.06.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND & AIMS Investigating the tissue-associated microbiota after surgically induced remission may help to understand the mechanisms initiating intestinal inflammation in Crohn's disease. METHODS Patients with Crohn's disease undergoing ileocolic resection were prospectively recruited in 6 academic centers. Biopsy samples from the neoterminal ileum, colon, and rectosigmoid were obtained from colonoscopies performed after surgery. Microbial DNA was extracted for 16S rRNA gene sequencing. Microbial diversity and taxonomic differential relative abundance were analyzed. A random forest model was applied to analyze the performance of clinical and microbial features to predict recurrence. A Rutgeerts score ≥i2 was deemed as endoscopic recurrence. RESULTS A total of 349 postoperative colonoscopies and 944 biopsy samples from 262 patients with Crohn's disease were analyzed. Ileal inflammation accounted for most of the explained variance of the ileal and colonic mucosa-associated microbiota. Samples obtained from 97 patients who were in surgically induced remission at first postoperative colonoscopy who went on to develop endoscopic recurrence at second colonoscopy showed lower diversity and microbial deviations when compared with patients who remained in endoscopic remission. Depletion of genus Anaerostipes and increase of several genera from class Gammaproteobacteria at the 3 biopsy sites increase the risk of further recurrence. Gut microbiome was able to predict future recurrence better than clinical features. CONCLUSIONS Ileal and colonic mucosa-associated microbiome deviations precede development of new-onset ileal inflammation after surgically induced remission and show good predictive performance for future recurrence. These findings suggest that targeted microbial modulation is a plausible modality to prevent postoperative Crohn's disease recurrence.
Collapse
Affiliation(s)
- Cristian Hernández-Rocha
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica of Chile, Santiago, Chile
| | - Williams Turpin
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Krzysztof Borowski
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Joanne M Stempak
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada
| | - Ksenija Sabic
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Kyle Gettler
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Christopher Tastad
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Colleen Chasteau
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ujunwa Korie
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mary Hanna
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Abdul Khan
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Emebet Mengesha
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Alain Bitton
- Division of Gastroenterology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Marc B Schwartz
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Arthur Barrie
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Lisa W Datta
- Harvey M. and Lyn P. Meyerhoff Inflammatory Bowel Disease Center, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mark Lazarev
- Harvey M. and Lyn P. Meyerhoff Inflammatory Bowel Disease Center, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven R Brant
- Harvey M. and Lyn P. Meyerhoff Inflammatory Bowel Disease Center, Division of Gastroenterology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Division of Gastroenterology and Hepatology, Department of Medicine, Rutgers Robert Wood Johnson Medical School and the Crohn's and Colitis Center of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, New Jersey; Department of Genetics and The Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - John D Rioux
- Research Centre, Montreal Heart Institute, Montréal, Quebec, Canada; Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Dermot P B McGovern
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars Sinai Medical Center, Los Angeles, California
| | - Richard H Duerr
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Human Genetics, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
| | - L Phil Schumm
- Biostatistics Laboratory & Research Computing Group, Department of Public Health Sciences, University of Chicago, Chicago, Illinois
| | - Judy H Cho
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark S Silverberg
- Zane Cohen Centre for Digestive Diseases, Mount Sinai Hospital, Toronto, Ontario, Canada; Division of Gastroenterology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Moniaux N, Geoffre N, Deshayes A, Dos Santos A, Job S, Lacoste C, Nguyen TS, Darnaud M, Friedel-Arboleas M, Guettier C, Purhonen J, Kallijärvi J, Amouyal G, Amouyal P, Bréchot C, Vivès RR, Buendia MA, Issad T, Faivre J. Tumor suppressive role of the antimicrobial lectin REG3A targeting the O -GlcNAc glycosylation pathway. Hepatology 2024:01515467-990000000-00943. [PMID: 38975812 DOI: 10.1097/hep.0000000000000993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND AND AIMS Antimicrobial proteins of the regenerating family member 3 alpha (REG3A) family provide a first line of protection against infections and transformed cells. Their expression is inducible by inflammation, which makes their role in cancer biology less clear since an immune-inflammatory context may preexist or coexist with cancer, as occurs in HCC. The aim of this study is to clarify the role of REG3A in liver carcinogenesis and to determine whether its carbohydrate-binding functions are involved. APPROACH AND RESULTS This study provides evidence for a suppressive role of REG3A in HCC by reducing O -GlcNAcylation in 2 mouse models of HCC, in vitro cell studies, and clinical samples. REG3A expression in hepatocytes significantly reduced global O -GlcNAcylation and O -GlcNAcylation of c-MYC in preneoplastic and tumor livers and markedly inhibited HCC development in REG3A-c-MYC double transgenic mice and mice exposed to diethylnitrosamine. REG3A modified O -GlcNAcylation without altering the expression or activity of O-linked N-acetylglucosaminyltransferase, O-linked N-acetylglucosaminyl hydrolase, or glutamine fructose-6-phosphate amidotransferase. Reduced O -GlcNAcylation was consistent with decreased levels of UDP-GlcNAc in precancerous and cancerous livers. This effect was linked to the ability of REG3A to bind glucose and glucose-6 phosphate, suggested by a REG3A mutant unable to bind glucose and glucose-6 phosphate and alter O -GlcNAcylation. Importantly, patients with cirrhosis with high hepatic REG3A expression had lower levels of O -GlcNAcylation and longer cancer-free survival than REG3A-negative cirrhotic livers. CONCLUSIONS REG3A helps fight liver cancer by reducing O -GlcNAcylation. This study suggests a new paradigm for the regulation of O -GlcNAc signaling in cancer-related pathways through interactions with the carbohydrate-binding function of REG3A.
Collapse
Affiliation(s)
- Nicolas Moniaux
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Nicolas Geoffre
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Alice Deshayes
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Alexandre Dos Santos
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Sylvie Job
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Claire Lacoste
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Tung-Son Nguyen
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Marion Darnaud
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | | | - Catherine Guettier
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Saclay, Hôpital Bicêtre, Laboratoire Anatomie Pathologique, Le Kremlin Bicêtre, France
| | - Janne Purhonen
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Jukka Kallijärvi
- Folkhälsan Research Center, Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | | | | | | | - Romain R Vivès
- Université Grenoble Alpes, CNRS, CEA, IBS, Grenoble, France
| | - Marie Annick Buendia
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Tarik Issad
- Institut Cochin, Université de Paris, CNRS, INSERM, Paris, France
| | - Jamila Faivre
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Le Kremlin-Bicêtre, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Medical-University Department (DMU) Biology Genetics, Université Paris-Saclay, Paul-Brousse Hospital, Villejuif, France
| |
Collapse
|
9
|
Rungruangsaphakun J, Ayimbila F, Nakphaichit M, Keawsompong S. Simulated Swine Digestion and Gut Microbiota Fermentation of Hydrolyzed Copra Meal. Animals (Basel) 2024; 14:1677. [PMID: 38891724 PMCID: PMC11171118 DOI: 10.3390/ani14111677] [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: 04/25/2024] [Revised: 05/20/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
This study aimed to compare the effects of hydrolyzed copra meal (HCM) inclusion at 1% on its in vitro digestibility and the microbiota and cecum fermentation using the gut microbiota of weaned swine, targeting microbial community and short-chain fatty acids (SCF). For this reason, three treatments were considered: control (no copra meal), 1% non-hydrolyzed copra meal (CM), and 1% HCM. Non-defatted copra meal was hydrolyzed and analyzed (reducing sugars and total carbohydrates) in our laboratory. For digestion, microbiota identification, and fermentation assays, fresh fecal samples from two weaned pigs (1 month old) were used. Three replicates of each treatment were employed. HCM was more digestible, with approximately 0.68 g of hydrolysate recovered after simulated digestion compared to 0.82 g of hydrolysate recovered from CM. This was shown by Scanning Electron Microscope (SEM) images. Also, the three swine shared the majority of microbial species identified at the phylum and family levels. There were no differences (p > 0.05) between treatments in the microbial community and SCFA during fermentation. However, higher Chao-1 and Shannon indexes were observed in CM and HCM treatments. HCM was also found to be capable of preserving Actinobacterota and Proteobacteria at the phylum level, while at the family level, both treatments may help Lactobacillaceae, Peptostreptococcaceae, Lachnospiraceae, and Ruminococcaceae survive in the long term. Also, there was a potential trend of increasing acetic acid and butyric acid in the CM and HCM treatments. While HCM shows promise in potentially modulating the gut microbiota of weaned swine, additional research is required to investigate the effects of higher doses of HCM on swine performance parameters.
Collapse
Affiliation(s)
- Jurairat Rungruangsaphakun
- Specialized Research Unit: Prebiotics and Probiotics for Health, Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand; (J.R.); (F.A.); (M.N.)
| | - Francis Ayimbila
- Specialized Research Unit: Prebiotics and Probiotics for Health, Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand; (J.R.); (F.A.); (M.N.)
- Center for Advanced Studies for Agriculture and Food, KU Institute of Advanced Studies, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand
| | - Massalin Nakphaichit
- Specialized Research Unit: Prebiotics and Probiotics for Health, Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand; (J.R.); (F.A.); (M.N.)
- Center for Advanced Studies for Agriculture and Food, KU Institute of Advanced Studies, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand
| | - Suttipun Keawsompong
- Specialized Research Unit: Prebiotics and Probiotics for Health, Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand; (J.R.); (F.A.); (M.N.)
- Center for Advanced Studies for Agriculture and Food, KU Institute of Advanced Studies, Kasetsart University (CASAF, NRU-KU), Bangkok 10900, Thailand
| |
Collapse
|
10
|
Liu JL, Xu X, Rixiati Y, Wang CY, Ni HL, Chen WS, Gong HM, Zhang ZL, Li S, Shen T, Li JM. Dysfunctional circadian clock accelerates cancer metastasis by intestinal microbiota triggering accumulation of myeloid-derived suppressor cells. Cell Metab 2024; 36:1320-1334.e9. [PMID: 38838643 DOI: 10.1016/j.cmet.2024.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 02/12/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024]
Abstract
Circadian homeostasis in mammals is a key intrinsic mechanism for responding to the external environment. However, the interplay between circadian rhythms and the tumor microenvironment (TME) and its influence on metastasis are still unclear. Here, in patients with colorectal cancer (CRC), disturbances of circadian rhythm and the accumulation of monocytes and granulocytes were closely related to metastasis. Moreover, dysregulation of circadian rhythm promoted lung metastasis of CRC by inducing the accumulation of myeloid-derived suppressor cells (MDSCs) and dysfunctional CD8+ T cells in the lungs of mice. Also, gut microbiota and its derived metabolite taurocholic acid (TCA) contributed to lung metastasis of CRC by triggering the accumulation of MDSCs in mice. Mechanistically, TCA promoted glycolysis of MDSCs epigenetically by enhancing mono-methylation of H3K4 of target genes and inhibited CHIP-mediated ubiquitination of PDL1. Our study links the biological clock with MDSCs in the TME through gut microbiota/metabolites in controlling the metastatic spread of CRC, uncovering a systemic mechanism for cancer metastasis.
Collapse
Affiliation(s)
- Jing-Lin Liu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xu Xu
- Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China
| | - Youlutuziayi Rixiati
- Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China
| | - Chu-Yi Wang
- Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China
| | - Heng-Li Ni
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Wen-Shu Chen
- Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China
| | - Hui-Min Gong
- Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China
| | - Zi-Long Zhang
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shi Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Tong Shen
- Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China.
| | - Jian-Ming Li
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China; Department of Pathology, Soochow Medical College, Soochow University, Suzhou 215123, China; The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; Department of Pathology and Institute of Molecular Pathology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, China.
| |
Collapse
|
11
|
Yan D, Wei G, Ai Z, Song S, Zhang L, Dong N, Dou X, Shan A. CXCR2, as a key regulatory gene of HDP-PG-1, maintains intestinal mucosal homeostasis. Int J Biol Macromol 2024; 269:132025. [PMID: 38704076 DOI: 10.1016/j.ijbiomac.2024.132025] [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: 03/05/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
The intestine defends against pathogenic microbial invasion via the secretion of host defense peptides (HDPs). Nutritional immunomodulation can stimulate the expression of endogenous HDPs and enhance the body's immune defense, representing a novel non-antibiotic strategy for disease prevention. The project aims to explore the regulatory mechanism of protegrin-1 (PG-1) expression using sodium phenylbutyrate (PBA) by omics sequencing technology and further investigate the role of key regulatory genes on intestinal health. The results showed that PBA promoted PG-1 expression in intestinal epithelial cells based on cell density through epidermal growth factor receptor (EGFR) and G protein-coupled receptor (GPR43). Transcriptome sequencing and microRNA sequencing revealed that C-X-C motif chemokine receptor 2 (CXCR2) exhibited interactions with PG-1. Pre-treatment cells with a CXCR2 inhibitor (SB225002) effectively suppressed the induction of PG-1 by PBA. Furthermore, SB225002 significantly suppressed the gene expression of HDPs in the jejunum of mice without influencing on the morphology, number of goblet cells, and proliferation of the intestine. CXCR2 inhibition significantly reduced the expression of HDPs during E. coli infection, and resulted in the edema of jejunal epithelial cells. The 16S rDNA analysis of cecal contents showed that the E. coli and SB225002 treatments changed gut microbiota diversity and composition at different taxonomic levels. Correlation analysis suggested a potential regulatory relationship between gut microbiota and HDPs. To that end, a gene involved in the HDP expression, CXCR2, has been identified in the study, which contributes to improving intestinal immune function. PBA may be used as a functional additive to regulate intestinal mucosal function, thereby enhancing the health of the intestinal and host.
Collapse
Affiliation(s)
- Di Yan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Guoyang Wei
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Zichun Ai
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Shuang Song
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Na Dong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiujing Dou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
12
|
Yu Y, Rothenberg ME, Ding HT, Brekkan A, Sperinde G, Harder B, Zhang R, Owen R, Kassir N, Lekkerkerker AN. Population pharmacokinetics and pharmacodynamics of efmarodocokin alfa (IL-22Fc). J Pharmacokinet Pharmacodyn 2024; 51:141-153. [PMID: 37864000 DOI: 10.1007/s10928-023-09888-2] [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: 06/16/2023] [Accepted: 09/24/2023] [Indexed: 10/22/2023]
Abstract
Efmarodocokin alfa (IL-22Fc) is a fusion protein of human IL-22 linked to the crystallizable fragment (Fc) of human IgG4. It has been tested in multiple indications including inflammatory bowel disease (IBD). The purposes of the present analyses were to describe the population pharmacokinetics (PK) of efmarodocokin alfa and perform pharmacodynamic (PD) analysis on the longitudinal changes of the PD biomarker REG3A after efmarodocokin alfa treatment as well as identify covariates that affect efmarodocokin alfa PK and REG3A PD. The data used for this analysis included 182 subjects treated with efmarodocokin alfa in two clinical studies. The population PK and PD analyses were conducted sequentially. Efmarodocokin alfa concentration-time data were analyzed using a nonlinear mixed-effects modeling approach, and an indirect response model was adopted to describe the REG3A PD data with efmarodocokin alfa serum concentration linked to the increase in REG3A. The analysis software used were NONMEM and R. A 3-compartment model with linear elimination best described the PK of efmarodocokin alfa. The estimated population-typical value for clearance (CL) was 1.12 L/day, and volume of central compartment was 6.15 L. Efmarodocokin alfa CL increased with higher baseline body weight, C-reactive protein, and CL was 27.6% higher in IBD patients compared to healthy subjects. The indirect response PD model adequately described the longitudinal changes of REG3A after efmarodocokin alfa treatment. A popPK and PD model for efmarodocokin alfa and REG3A was developed and covariates affecting the PK and PD were identified.
Collapse
Affiliation(s)
- Yanke Yu
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA.
| | | | - Han Ting Ding
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | | | | - Brandon Harder
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Rong Zhang
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Ryan Owen
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Nastya Kassir
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, 94080, USA
| | | |
Collapse
|
13
|
Zheng J, Zhou Y, Zhang D, Ma K, Gong Y, Luo X, Liu J, Cui S. Intestinal melatonin levels and gut microbiota homeostasis are independent of the pineal gland in pigs. Front Microbiol 2024; 15:1352586. [PMID: 38596375 PMCID: PMC11003461 DOI: 10.3389/fmicb.2024.1352586] [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: 12/08/2023] [Accepted: 02/28/2024] [Indexed: 04/11/2024] Open
Abstract
Introduction Melatonin (MEL) is a crucial neuroendocrine hormone primarily produced by the pineal gland. Pinealectomy (PINX) has been performed on an endogenous MEL deficiency model to investigate the functions of pineal MEL and its relationship with various diseases. However, the effect of PINX on the gastrointestinal tract (GIT) MEL levels and gut microbiome in pigs has not been previously reported. Methods By using a newly established pig PINX model, we detected the levels of MEL in the GIT by liquid chromatography-tandem mass spectrometry. In addition, we examined the effects of PINX on the expression of MEL synthesis enzymes, intestinal histomorphology, and the intestinal barrier. Furthermore, 16S rRNA sequencing was performed to analyze the colonic microbiome. Results PINX reduced serum MEL levels but did not affect GIT MEL levels. Conversely, MEL supplementation increased MEL levels in the GIT and intestinal contents. Neither PINX nor MEL supplementation had any effect on weight gain, organ coefficient, serum biochemical indexes, or MEL synthetase arylalkylamine N-acetyltransferase (AANAT) expression in the duodenum, ileum, and colon. Furthermore, no significant differences were observed in the intestinal morphology or intestinal mucosal barrier function due to the treatments. Additionally, 16S rRNA sequencing revealed that PINX had no significant impact on the composition of the intestinal microbiota. Nevertheless, MEL supplementation decreased the abundance of Fibrobacterota and increased the abundance of Actinobacteriota, Desulfobacterota, and Chloroflexi. Conclusion We demonstrated that synthesis of MEL in the GIT is independent of the pineal gland. PINX had no influence on intestinal MEL level and microbiota composition in pigs, while exogenous MEL alters the structure of the gut microbiota.
Collapse
Affiliation(s)
- Jiaming Zheng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yewen Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Di Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Kezhe Ma
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yuneng Gong
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xuan Luo
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Jiali Liu
- State Key Laboratory of Animal Biotech Breeding, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Sheng Cui
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- Institute of Reproduction and Metabolism, Yangzhou University, Yangzhou, China
| |
Collapse
|
14
|
Forsyth CB, Shaikh M, Engen PA, Preuss F, Naqib A, Palmen BA, Green SJ, Zhang L, Bogin ZR, Lawrence K, Sharma D, Swanson GR, Bishehsari F, Voigt RM, Keshavarzian A. Evidence that the loss of colonic anti-microbial peptides may promote dysbiotic Gram-negative inflammaging-associated bacteria in aging mice. FRONTIERS IN AGING 2024; 5:1352299. [PMID: 38501032 PMCID: PMC10945560 DOI: 10.3389/fragi.2024.1352299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/02/2024] [Indexed: 03/20/2024]
Abstract
Introduction: Aging studies in humans and mice have played a key role in understanding the intestinal microbiome and an increased abundance of "inflammaging" Gram-negative (Gn) bacteria. The mechanisms underlying this inflammatory profile in the aging microbiome are unknown. We tested the hypothesis that an aging-related decrease in colonic crypt epithelial cell anti-microbial peptide (AMP) gene expression could promote colonic microbiome inflammatory Gn dysbiosis and inflammaging. Methods: As a model of aging, C57BL/6J mice fecal (colonic) microbiota (16S) and isolated colonic crypt epithelial cell gene expression (RNA-seq) were assessed at 2 months (mth) (human: 18 years old; yo), 15 mth (human: 50 yo), and 25 mth (human: 84 yo). Informatics examined aging-related microbial compositions, differential colonic crypt epithelial cell gene expressions, and correlations between colonic bacteria and colonic crypt epithelial cell gene expressions. Results: Fecal microbiota exhibited significantly increased relative abundances of pro-inflammatory Gn bacteria with aging. Colonic crypt epithelial cell gene expression analysis showed significant age-related downregulation of key AMP genes that repress the growth of Gn bacteria. The aging-related decrease in AMP gene expressions is significantly correlated with an increased abundance in Gn bacteria (dysbiosis), loss of colonic barrier gene expression, and senescence- and inflammation-related gene expression. Conclusion: This study supports the proposed model that aging-related loss of colonic crypt epithelial cell AMP gene expression promotes increased relative abundances of Gn inflammaging-associated bacteria and gene expression markers of colonic inflammaging. These data may support new targets for aging-related therapies based on intestinal genes and microbiomes.
Collapse
Affiliation(s)
- Christopher B. Forsyth
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Maliha Shaikh
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Phillip A. Engen
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Fabian Preuss
- Department of Biological Sciences, University of Wisconsin Parkside, Kenosha, WI, United States
| | - Ankur Naqib
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Breanna A. Palmen
- Department of Biological Sciences, University of Wisconsin Parkside, Kenosha, WI, United States
| | - Stefan J. Green
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Genomics and Microbiome Core Facility, Rush University Medical Center, Chicago, IL, United States
| | - Lijuan Zhang
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Zlata R. Bogin
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Kristi Lawrence
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Deepak Sharma
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Garth R. Swanson
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Faraz Bishehsari
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|
15
|
Zhang Z, Huang J, Li C, Zhao Z, Cui Y, Yuan X, Wang X, Liu Y, Zhou Y, Zhu Z. The gut microbiota contributes to the infection of bovine viral diarrhea virus in mice. J Virol 2024; 98:e0203523. [PMID: 38299844 PMCID: PMC10878277 DOI: 10.1128/jvi.02035-23] [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: 01/04/2024] [Accepted: 01/10/2024] [Indexed: 02/02/2024] Open
Abstract
Bovine viral diarrhea virus (BVDV) is prevalent worldwide and causes significant economic losses. Gut microbiota is a large microbial community and has a variety of biological functions. However, whether there is a correlation between gut microbiota and BVDV infection and what kind of relation between them have not been reported. Here, we found that gut microbiota composition changed in normal mice after infecting with BVDV, but mainly the low abundance microbe was affected. Interestingly, BVDV infection significantly reduced the diversity of gut microbiota and changed its composition in gut microbiota-dysbiosis mice. Furthermore, compared with normal mice of BVDV infection, there were more viral loads in the duodenum, jejunum, spleen, and liver of the gut microbiota-dysbiosis mice. However, feces microbiota transplantation (FMT) reversed these effects. The data above indicated that the dysbiosis of gut microbiota was a key factor in the high infection rate of BVDV. It is found that the IFN-I signal was involved by investigating the underlying mechanisms. The inhibition of the proliferation and increase in the apoptosis of peripheral blood lymphocytes (PBL) were also observed. However, FMT treatment reversed these changes by regulating PI3K/Akt, ERK, and Caspase-9/Caspase-3 pathways. Furthermore, the involvement of butyrate in the pathogenesis of BVDV was also further confirmed. Our results showed for the first time that gut microbiota acts as a key endogenous defense mechanism against BVDV infection; moreover, targeting regulation of gut microbiota structure and abundance may serve as a new strategy to prevent and control the disease.IMPORTANCEWhether the high infection rate of BVDV is related to gut microbiota has not been reported. In addition, most studies on BVDV focus on in vitro experiments, which limits the study of its prevention and control strategy and its pathogenic mechanism. In this study, we successfully confirmed the causal relationship between gut microbiota and BVDV infection as well as the potential molecular mechanism based on a mouse model of BVDV infection and a mouse model of gut microbiota dysbiosis. Meanwhile, a mouse model which is more susceptible to BVDV provided in this study lays an important foundation for further research on prevention and control strategy of BVDV and its pathogenesis. In addition, the antiviral effect of butyrate, the metabolites of butyrate-producing bacteria, has been further revealed. Overall, our findings provide a promising prevention and control strategy to treat this infectious disease which is distributed worldwide.
Collapse
Affiliation(s)
- Zecai Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Jiang Huang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Agriculture and Rural Bureau of Sinan County, Sinan County, Guizhou, China
- Animal Health Supervision Institute of Sinan County, Sinan County, Guizhou, China
| | - Chuang Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Zhicheng Zhao
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Yueqi Cui
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Xueying Yuan
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Xue Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
| | - Yu Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Yulong Zhou
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| | - Zhanbo Zhu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
- Key Laboratory of Bovine Disease Control in Northeast China, Ministry of Agriculture and Rural affairs, Daqing, China
- Engineering Research Center for Prevention and Control of Cattle Diseases, Heilongjiang Province, Daqing, China
- Heilongjiang Province Cultivating Collaborative Innovation Center for The Beidahuang Modern Agricultural Industry Technology, Daqing, China
| |
Collapse
|
16
|
Real MVF, Colvin MS, Sheehan MJ, Moeller AH. Major urinary protein ( Mup) gene family deletion drives sex-specific alterations in the house-mouse gut microbiota. Microbiol Spectr 2024; 12:e0356623. [PMID: 38170981 PMCID: PMC10846032 DOI: 10.1128/spectrum.03566-23] [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: 10/04/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
The gut microbiota is shaped by host metabolism. In house mice (Mus musculus), major urinary protein (MUP) pheromone production represents a considerable energy investment, particularly in sexually mature males. Deletion of the Mup gene family shifts mouse metabolism toward an anabolic state, marked by lipogenesis, lipid accumulation, and body mass increases. Given the metabolic implications of MUPs, they may also influence the gut microbiota. Here, we investigated the effect of a deletion of the Mup gene family on the gut microbiota of sexually mature mice. Shotgun metagenomics revealed distinct taxonomic and functional profiles between wild-type and knockout males but not females. Deletion of the Mup gene cluster significantly reduced diversity in microbial families and functions in male mice. Additionally, a species of Ruminococcaceae and several microbial functions, such as transporters involved in vitamin B5 acquisition, were significantly depleted in the microbiota of Mup knockout males. Altogether, these results show that MUPs significantly affect the gut microbiota of house mouse in a sex-specific manner.IMPORTANCEThe community of microorganisms that inhabits the gastrointestinal tract can have profound effects on host phenotypes. The gut microbiota is in turn shaped by host genes, including those involved with host metabolism. In adult male house mice, expression of the major urinary protein (Mup) gene cluster represents a substantial energy investment, and deletion of the Mup gene family leads to fat accumulation and weight gain in males. We show that deleting Mup genes also alters the gut microbiota of male, but not female, mice in terms of both taxonomic and functional compositions. Male mice without Mup genes harbored fewer gut bacterial families and reduced abundance of a species of Ruminococcaceae, a family that has been previously shown to reduce obesity risk. Studying the impact of the Mup gene family on the gut microbiota has the potential to reveal the ways in which these genes affect host phenotypes.
Collapse
Affiliation(s)
- Madalena V. F. Real
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Melanie S. Colvin
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Michael J. Sheehan
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Andrew H. Moeller
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
17
|
Kashyap Y, Wang ZJ. Gut microbiota dysbiosis alters chronic pain behaviors in a humanized transgenic mouse model of sickle cell disease. Pain 2024; 165:423-439. [PMID: 37733476 PMCID: PMC10843763 DOI: 10.1097/j.pain.0000000000003034] [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: 05/02/2023] [Accepted: 05/31/2023] [Indexed: 09/23/2023]
Abstract
ABSTRACT Pain is the most common symptom experienced by patients with sickle cell disease (SCD) throughout their lives and is the main cause of hospitalization. Despite the progress that has been made towards understanding the disease pathophysiology, major gaps remain in the knowledge of SCD pain, the transition to chronic pain, and effective pain management. Recent evidence has demonstrated a vital role of gut microbiota in pathophysiological features of SCD. However, the role of gut microbiota in SCD pain is yet to be explored. We sought to evaluate the compositional differences in the gut microbiota of transgenic mice with SCD and nonsickle control mice and investigate the role of gut microbiota in SCD pain by using antibiotic-mediated gut microbiota depletion and fecal material transplantation (FMT). The antibiotic-mediated gut microbiota depletion did not affect evoked pain but significantly attenuated ongoing spontaneous pain in mice with SCD. Fecal material transplantation from mice with SCD to wild-type mice resulted in tactile allodynia (0.95 ± 0.17 g vs 0.08 ± 0.02 g, von Frey test, P < 0.001), heat hyperalgesia (15.10 ± 0.79 seconds vs 8.68 ± 1.17 seconds, radiant heat, P < 0.01), cold allodynia (2.75 ± 0.26 seconds vs 1.68 ± 0.08 seconds, dry ice test, P < 0.01), and anxiety-like behaviors (Elevated Plus Maze Test, Open Field Test). On the contrary, reshaping gut microbiota of mice with SCD with FMT from WT mice resulted in reduced tactile allodynia (0.05 ± 0.01 g vs 0.25 ± 0.03 g, P < 0.001), heat hyperalgesia (5.89 ± 0.67 seconds vs 12.25 ± 0.76 seconds, P < 0.001), and anxiety-like behaviors. These findings provide insights into the relationship between gut microbiota dysbiosis and pain in SCD, highlighting the importance of gut microbial communities that may serve as potential targets for novel pain interventions.
Collapse
Affiliation(s)
- Yavnika Kashyap
- Departments of Pharmaceutical Sciences and Center for Biomolecular Science, University of Illinois, Chicago, IL, United States
| | - Zaijie Jim Wang
- Departments of Pharmaceutical Sciences and Center for Biomolecular Science, University of Illinois, Chicago, IL, United States
- Department of Neurology & Rehabilitation, and Sickle Cell Center, University of Illinois College of Medicine, Chicago, IL, United States
- Department of Biomedical Engineering, University of Illinois, Chicago, IL 60607, United States
| |
Collapse
|
18
|
Kim S, Jang SH, Kim MJ, Lee JJ, Kim KM, Kim YH, Lee JH, Jung SK. Hybrid nutraceutical of 2-ketoglutaric acid in improving inflammatory bowel disease: Role of prebiotics and TAK1 inhibitor. Biomed Pharmacother 2024; 171:116126. [PMID: 38219386 DOI: 10.1016/j.biopha.2024.116126] [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: 09/26/2023] [Revised: 12/19/2023] [Accepted: 01/02/2024] [Indexed: 01/16/2024] Open
Abstract
The main cause of inflammatory bowel disease (IBD) is abnormal intestinal permeability due to the disruption of the tight junction of the intestinal barrier through a pathogen-mediated inflammatory mechanism and an imbalance of the gut microbiota. This study aimed to evaluate whether 2-ketoglutaric acid alleviated permeability dysfunction with tight junction localization, activated the transforming growth factor beta-activated kinase 1 (TAK1) inflammation pathway, and regulated the homeostasis of the intestinal microbiome in vitro and in vivo IBD model. Our findings revealed that 2-ketoglutaric acid significantly suppressed abnormal intestinal permeability, delocalization of tight junction proteins from the intestinal cell, expression of inflammatory cytokines, such as TNF-α, both in vitro and in vivo. 2-Ketoglutaric acid was found to directly bind to TAK1 and inhibit the TNF receptor-associated factor 6 (TRAF6)-TAK1 interaction, which is related to the activation of nuclear factor kappa B (NF-κB) pathways, thereby regulating the expression of mitogen-activated protein kinase. Dietary 2-ketoglutaric acid also alleviated gut microbiota dysbiosis and IBD symptoms, as demonstrated by improvements in the intestine length and the abundance of Ligilactobacillus, Coriobacteriaceae_UCG_002, and Ruminococcaceae_unclassified in mice with colitis. This study indicated that 2-ketoglutaric acid binds to TAK1 for activity inhibition which is related to the NF-κB pathway and alleviates abnormal permeability by regulating tight junction localization and gut microbiome homeostasis. Therefore, 2-ketoglutaric acid is an effective nutraceutical agent and prebiotic for the treatment of IBD.
Collapse
Affiliation(s)
- San Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Se Hyeon Jang
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Min Jeong Kim
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jeong Jae Lee
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Kyung-Min Kim
- Division of Plant Biosciences, School of Applied Biosciences, College of Agriculture and Life Science, Kyungpook National University, Daegu 41566, Korea; Coastal Agriculture Research Institute, Kyungpook National University, Daegu 41566, Korea
| | - Young Hoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Ju-Hoon Lee
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea; Department of Food and Animal Biotechnology and Center for Food and Bioconvergence, Seoul National University, Seoul 08826, Republic of Korea
| | - Sung Keun Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea; Research Institute of Tailored Food Technology, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
19
|
Krigul KL, Feeney RH, Wongkuna S, Aasmets O, Holmberg SM, Andreson R, Puértolas-Balint F, Pantiukh K, Sootak L, Org T, Tenson T, Org E, Schroeder BO. A history of repeated antibiotic usage leads to microbiota-dependent mucus defects. Gut Microbes 2024; 16:2377570. [PMID: 39034613 PMCID: PMC11529412 DOI: 10.1080/19490976.2024.2377570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 07/03/2024] [Indexed: 07/23/2024] Open
Abstract
Recent evidence indicates that repeated antibiotic usage lowers microbial diversity and ultimately changes the gut microbiota community. However, the physiological effects of repeated - but not recent - antibiotic usage on microbiota-mediated mucosal barrier function are largely unknown. By selecting human individuals from the deeply phenotyped Estonian Microbiome Cohort (EstMB), we here utilized human-to-mouse fecal microbiota transplantation to explore long-term impacts of repeated antibiotic use on intestinal mucus function. While a healthy mucus layer protects the intestinal epithelium against infection and inflammation, using ex vivo mucus function analyses of viable colonic tissue explants, we show that microbiota from humans with a history of repeated antibiotic use causes reduced mucus growth rate and increased mucus penetrability compared to healthy controls in the transplanted mice. Moreover, shotgun metagenomic sequencing identified a significantly altered microbiota composition in the antibiotic-shaped microbial community, with known mucus-utilizing bacteria, including Akkermansia muciniphila and Bacteroides fragilis, dominating in the gut. The altered microbiota composition was further characterized by a distinct metabolite profile, which may be caused by differential mucus degradation capacity. Consequently, our proof-of-concept study suggests that long-term antibiotic use in humans can result in an altered microbial community that has reduced capacity to maintain proper mucus function in the gut.
Collapse
Affiliation(s)
- Kertu Liis Krigul
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Rachel H. Feeney
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Supapit Wongkuna
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Oliver Aasmets
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Sandra M. Holmberg
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Reidar Andreson
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Fabiola Puértolas-Balint
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| | - Kateryna Pantiukh
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Linda Sootak
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Tõnis Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Tanel Tenson
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Elin Org
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Bjoern O. Schroeder
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Shah T, Wang Y, Wang Y, Li Q, Zhou J, Hou Y, Wang B, Xia X. A Comparative Analysis of the Stomach, Gut, and Lung Microbiomes in Rattus norvegicus. Microorganisms 2023; 11:2359. [PMID: 37764203 PMCID: PMC10534326 DOI: 10.3390/microorganisms11092359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Urban rats serve as reservoirs for several zoonotic pathogens that seriously endanger public health, destroy stored food, and damage infrastructure due to their close interaction with humans and domestic animals. Here, we characterize the core microbiomes of R. norvegicus's stomach, gut, and lung using 16S rRNA next-generation Illumina HiSeq sequencing. The USEARCH software (v11) assigned the dataset to operational taxonomic units (OTUs). The alpha diversity index was calculated using QIIME1, while the beta diversity index was determined using the Bray-Curtis and Euclidean distances between groups. Principal component analyses visualized variation across samples based on the OTU information using the R package. Linear discriminant analysis, effect sizes (LEfSe), and phylogenetic investigation were used to identify differentially abundant taxa among groups. We reported an abundance of microbiota in the stomach, and they shared some of them with the gut and lung microbiota. A close look at the microbial family level reveals abundant Lactobacillaceae and Bifidobacteriaceae in the stomach, whereas Lactobacillaceae and Erysipelotrichaceae were more abundant in the gut; in contrast, Alcaligenaceae were abundant in the lungs. At the species level, some beneficial bacteria, particularly Lactobacillus reuteri and Lactobacillus johnsonii, and some potential pathogens, such as Bordetella hinzii, Streptococcus parauberis, Porphyromonas pogonae, Clostridium perfringens, etc., were identified in stomach, gut, and lung samples. Moreover, the alpha and beta diversity indexes revealed significant differences between the groups. Further analysis revealed abundant differential taxonomic biomarkers, i.e., increased Prevotellaceae and Clostridia in the lungs, whereas Campylobacteria and Lachnospirales were richest in the stomachs. In conclusion, we identified many beneficial, opportunistic, and highly pathogenic bacteria, confirming the importance of urban rats for public health. This study recommends a routine survey program to monitor rodent distribution and the pathogens they carry and transmit to humans and other domestic mammals.
Collapse
Affiliation(s)
- Taif Shah
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (T.S.)
| | - Yuhan Wang
- Department of Biodiversity Conservation, Southwest Forestry University, Kunming 650500, China
- Research Institute of Forest Protection, Yunnan Academy of Forestry and Grassland, Kunming 650500, China
| | - Yixuan Wang
- Research Institute of Forest Protection, Yunnan Academy of Forestry and Grassland, Kunming 650500, China
| | - Qian Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (T.S.)
| | - Jiuxuan Zhou
- Research Institute of Forest Protection, Yunnan Academy of Forestry and Grassland, Kunming 650500, China
| | - Yutong Hou
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (T.S.)
| | - Binghui Wang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (T.S.)
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; (T.S.)
- School of Public Health, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
21
|
Jang KK, Heaney T, London M, Ding Y, Putzel G, Yeung F, Ercelen D, Chen YH, Axelrad J, Gurunathan S, Zhou C, Podkowik M, Arguelles N, Srivastava A, Shopsin B, Torres VJ, Keestra-Gounder AM, Pironti A, Griffin ME, Hang HC, Cadwell K. Antimicrobial overproduction sustains intestinal inflammation by inhibiting Enterococcus colonization. Cell Host Microbe 2023; 31:1450-1468.e8. [PMID: 37652008 PMCID: PMC10502928 DOI: 10.1016/j.chom.2023.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 07/02/2023] [Accepted: 08/07/2023] [Indexed: 09/02/2023]
Abstract
Loss of antimicrobial proteins such as REG3 family members compromises the integrity of the intestinal barrier. Here, we demonstrate that overproduction of REG3 proteins can also be detrimental by reducing a protective species in the microbiota. Patients with inflammatory bowel disease (IBD) experiencing flares displayed heightened levels of secreted REG3 proteins that mediated depletion of Enterococcus faecium (Efm) from the gut microbiota. Efm inoculation of mice ameliorated intestinal inflammation through activation of the innate immune receptor NOD2, which was associated with the bacterial DL-endopeptidase SagA that generates NOD2-stimulating muropeptides. NOD2 activation in myeloid cells induced interleukin-1β (IL-1β) secretion to increase the proportion of IL-22-producing CD4+ T helper cells and innate lymphoid cells that promote tissue repair. Finally, Efm was unable to protect mice carrying a NOD2 gene variant commonly found in IBD patients. Our findings demonstrate that inflammation self-perpetuates by causing aberrant antimicrobial activity that disrupts symbiotic relationships with gut microbes.
Collapse
Affiliation(s)
- Kyung Ku Jang
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Thomas Heaney
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Mariya London
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yi Ding
- Department of Laboratory Medicine, Geisinger Health, Danville, PA 17822, USA
| | - Gregory Putzel
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Frank Yeung
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Defne Ercelen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ying-Han Chen
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jordan Axelrad
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sakteesh Gurunathan
- Division of Gastroenterology and Hepatology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Chaoting Zhou
- Cell and Molecular Biology Graduate Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Magdalena Podkowik
- Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA; Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Natalia Arguelles
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Anusha Srivastava
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Bo Shopsin
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA; Division of Infectious Diseases and Immunology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Victor J Torres
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - A Marijke Keestra-Gounder
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Alejandro Pironti
- Department of Microbiology, New York University Grossman School of Medicine, New York, NY 10016, USA; Antimicrobial-Resistant Pathogens Program, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Matthew E Griffin
- Department of Immunology and Microbiology, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA; Department of Chemistry, Scripps Research, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Ken Cadwell
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
He X, Zhou H. Decoding the IBD paradox: A triadic interplay between REG3, enterococci, and NOD2. Cell Host Microbe 2023; 31:1425-1427. [PMID: 37708849 DOI: 10.1016/j.chom.2023.08.008] [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: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
In this issue of Cell Host & Microbe, Jang et al. decode an intriguing paradox: excessive antimicrobial peptide-regenerating family member 3 (REG3) expression in inflammatory bowel disease (IBD) patients depletes protective enterococci. This depletion impairs the NOD2 anti-inflammatory pathway and perpetuates inflammation, thus providing insight into IBD pathogenesis.
Collapse
Affiliation(s)
- Xiaolong He
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510655, China
| | - Hongwei Zhou
- Microbiome Medicine Center, Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510655, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou, Guangdong, 510655, China.
| |
Collapse
|
23
|
He Z, Liu J, Liu Y. Daphnetin attenuates intestinal inflammation, oxidative stress, and apoptosis in ulcerative colitis via inhibiting REG3A-dependent JAK2/STAT3 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:2132-2142. [PMID: 37209277 DOI: 10.1002/tox.23837] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/22/2023]
Abstract
Daphnetin is a natural coumarin compound with anti-inflammatory, anti-oxidant, and anti-apoptotic effects, which has been previously demonstrated to ameliorate DSS-induced ulcerative colitis (UC). However, the molecular mechanism involved in the daphnetin-mediated pathological process of UC remains unclarified. The current study used DSS-induced mice and LPS-challenged Caco-2 cells as UC models. Bodyweight, disease activity index (DAI) score, and colon length were used to evaluate the severity of colitis. The histological changes in colon tissues were observed using H&E and PAS staining. Protein levels were detected by western blot. The malondialdehyde (MDA) and superoxide dismutase (SOD) activities were used to assess oxidative stress. Inflammatory responses were evaluated by detecting the levels of inflammatory cytokines (IFN-r, IL-1β, IL-6, and TNF-α) using flow cytometry. CCK-8 and TUNEL assay were employed to determine cell growth and cell death, respectively. The results showed that daphnetin could ameliorate the severity of colitis and attenuate the damage to intestinal structure in DSS-induced mice. Compared with the DSS group, the expression of ZO-1, occludin, and anti-apoptotic protein (BCL-2) was increased while the level of pro-apoptotic proteins (Bax and cleaved caspase 3) was decreased in DSS + daphnetin group. The activity of MDA and SOD, as well as the levels of inflammatory cytokines were substantially suppressed by daphnetin. In consistency, in vitro assays indicated that daphnetin protected Caco-2 cells from LPS-stimulated viability impairment, apoptosis, oxidative stress, and inflammation. Furthermore, daphnetin suppressed the activity of JAK2/STAT signaling in LPS-induced Caco-2 cells in a REG3A-dependent manner. REG3A overexpression abated the ameliorative effects of daphnetin while JAK2/STAT signaling inhibition functioned synergically with daphnetin in LPS-stimulated Caco-2 cells. Collectively, this study deepened the understanding of the therapeutic effects of daphnetin on UC and uncovered for the first time that daphnetin functioned through REG3A-activated JAK2/STAT3 signaling in UC, which may provide novel insights for the treatment of UC.
Collapse
Affiliation(s)
- Zhi He
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Jingjing Liu
- Center of Clinical Laboratory Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Yang Liu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
24
|
Shin JH, Bozadjieva-Kramer N, Seeley RJ. Reg3γ: current understanding and future therapeutic opportunities in metabolic disease. Exp Mol Med 2023; 55:1672-1677. [PMID: 37524871 PMCID: PMC10474034 DOI: 10.1038/s12276-023-01054-5] [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] [Received: 04/11/2023] [Accepted: 05/01/2023] [Indexed: 08/02/2023] Open
Abstract
Regenerating family member gamma, Reg3γ (the mouse homolog of human REG3A), belonging to the antimicrobial peptides (AMPs), functions as a part of the host immune system to maintain spatial segregation between the gut bacteria and the host in the intestine via bactericidal activity. There is emerging evidence that gut manipulations such as bariatric surgery, dietary supplementation or drug treatment to produce metabolic benefits alter the gut microbiome. In addition to changes in a wide range of gut hormones, these gut manipulations also induce the expression of Reg3γ in the intestine. Studies over the past decades have revealed that Reg3γ not only plays a role in the gut lumen but can also contribute to host physiology through interaction with the gut microbiota. Herein, we discuss the current knowledge regarding the biology of Reg3γ, its role in various metabolic functions, and new opportunities for therapeutic strategies to treat metabolic disorders.
Collapse
Affiliation(s)
- Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | | | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Real MVF, Colvin MS, Sheehan MJ, Moeller AH. Major urinary protein ( Mup) gene family deletion drives sex-specific alterations on the house mouse gut microbiota. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.01.551491. [PMID: 37577672 PMCID: PMC10418228 DOI: 10.1101/2023.08.01.551491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
The gut microbiota is shaped by host metabolism. In house mice (Mus musculus), major urinary protein (MUP) pheromone production represents a considerable energy investment, particularly in sexually mature males. Deletion of the Mup gene family shifts mouse metabolism towards an anabolic state, marked by lipogenesis, lipid accumulation, and body mass increases. Given the metabolic implications of MUPs, they may also influence the gut microbiota. Here, we investigated the effect of deletion of the Mup gene family on the gut microbiota of sexually mature mice. Shotgun metagenomics revealed distinct taxonomic and functional profiles between wildtype and knockout males, but not females. Deletion of the Mup gene cluster significantly reduced diversity in microbial families and functions in male mice. Additionally, specific taxa of the Ruminococcaceae family, which is associated with gut health and reduced risk of developing metabolic syndrome, and several microbial functions, such as transporters involved in vitamin B5 acquisition, were significantly depleted in the microbiota of Mup-knockout males. Altogether these results show that major urinary proteins significantly affect the gut microbiota of house mouse in a sex-specific manner.
Collapse
Affiliation(s)
- Madalena V. F. Real
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| | - Melanie S. Colvin
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Michael J. Sheehan
- Department of Neurobiology and Behavior, Cornell University, Ithaca, New York, USA
| | - Andrew H. Moeller
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
| |
Collapse
|
26
|
Chen H, Li Y, Wang J, Zheng T, Wu C, Cui M, Feng Y, Ye H, Dong Z, Dang Y. Plant Polyphenols Attenuate DSS-induced Ulcerative Colitis in Mice via Antioxidation, Anti-inflammation and Microbiota Regulation. Int J Mol Sci 2023; 24:10828. [PMID: 37446006 DOI: 10.3390/ijms241310828] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
The pathogenesis of ulcerative colitis (UC) is associated with inflammation, oxidative stress, and gut microbiota imbalance. Although most researchers have demonstrated the antioxidant bioactivity of the phenolic compounds in plants, their UC-curing ability and underlying mechanisms still need to be further and adequately explored. Herein, we studied the antioxidation-structure relationship of several common polyphenols in plants including gallic acid, proanthocyanidin, ellagic acid, and tannic acid. Furthermore, the in vivo effects of the plant polyphenols on C57BL/6 mice with dextran-sulfate-sodium-induced UC were evaluated and the action mechanisms were explored. Moreover, the interplay of several mechanisms was determined. The higher the number of phenolic hydroxyl groups, the stronger the antioxidant activity. All polyphenols markedly ameliorated the symptoms and pathological progression of UC in mice. Furthermore, inflammatory cytokine levels were decreased and the intestinal barrier was repaired. The process was regulated by the antioxidant-signaling pathway of nuclear-erythroid 2-related factor 2. Moreover, the diversity of the intestinal microbiota, Firmicutes-to-Bacteroides ratio, and relative abundance of beneficial bacteria were increased. An interplay was observed between microbiota regulation and oxidative stress, immunity, and inflammatory response. Furthermore, intestinal barrier repair was found to be correlated with inflammatory responses. Our study results can form a basis for comprehensively developing plant-polyphenol-related medicinal products.
Collapse
Affiliation(s)
- Huan Chen
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Ying Li
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100700, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100700, China
| | - Jinrui Wang
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Tingting Zheng
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Chenyang Wu
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Mengyao Cui
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Yifan Feng
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Hanyi Ye
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
| | - Zhengqi Dong
- Drug Delivery Research Center, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100193, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100094, China
- Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Beijing 100700, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing 100700, China
| | - Yunjie Dang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| |
Collapse
|
27
|
Liu T, Sun Z, Yang Z, Qiao X. Microbiota-derived short-chain fatty acids and modulation of host-derived peptides formation: Focused on host defense peptides. Biomed Pharmacother 2023; 162:114586. [PMID: 36989711 DOI: 10.1016/j.biopha.2023.114586] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/12/2023] [Accepted: 03/21/2023] [Indexed: 03/29/2023] Open
Abstract
The byproducts of bacterial fermentation known as short-chain fatty acids (SCFAs) are chemically comprised of a carboxylic acid component and a short hydrocarbon chain. Recent investigations have demonstrated that SCFAs can affect intestinal immunity by inducing endogenous host defense peptides (HDPs) and their beneficial effects on barrier integrity, gut health, energy supply, and inflammation. HDPs, which include defensins, cathelicidins, and C-type lectins, perform a significant function in innate immunity in gastrointestinal mucosal membranes. SCFAs have been demonstrated to stimulate HDP synthesis by intestinal epithelial cells via interactions with G protein-coupled receptor 43 (GPR43), activation of the Jun N-terminal kinase (JNK) and Mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinase (ERK) pathways, and the cell growth pathways. Furthermore, SCFA butyrate has been demonstrated to enhance the number of HDPs released from macrophages. SCFAs promote monocyte-to-macrophage development and stimulate HDP synthesis in macrophages by inhibiting histone deacetylase (HDAC). Understanding the etiology of many common disorders might be facilitated by studies into the function of microbial metabolites, such as SCFAs, in the molecular regulatory processes of immune responses (e.g., HDP production). This review will focus on the current knowledge of the role and mechanism of microbiota-derived SCFAs in influencing the synthesis of host-derived peptides, particularly HDPs.
Collapse
|
28
|
Zhang M, Zhang M, Kou G, Li Y. The relationship between gut microbiota and inflammatory response, learning and memory in mice by sleep deprivation. Front Cell Infect Microbiol 2023; 13:1159771. [PMID: 37293204 PMCID: PMC10244646 DOI: 10.3389/fcimb.2023.1159771] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 06/10/2023] Open
Abstract
Objective Sleep deprivation has developed into a common phenomenon, which can lead to inflammatory responses and cognitive impairment, but the underlying mechanism is ambiguous. Emerging evidence shows that gut microbiota plays a crucial role in theoccurrence and development of inflammatory and psychiatric diseases, possibly through neuroinflammation and the brain-gut axis. The current study investigated the influence of sleep deprivation on gut microbiota composition, pro-inflammatory cytokines, learning and memory in mice. Further, it explored whether changes in gut microbiota increase pro-inflammatory cytokine and induce learning and memory impairment. Methods Healthy 8-week-old male C57BL/6J mice were randomly divided into the regular control group (RC), environmental control group (EC), and sleep deprivation group (SD). The sleep deprivation model was established by the Modified Multiple Platform Method. The experimental mice were subjected to sleep deprivation for 6h/d (8:00 am∼14:00 pm) in a sleep deprivation chamber, and the duration of sleep deprivation was 8 weeks. Morris water maze test to assess learning and memory in mice. Enzyme-Linked Immunosorbent Assay determined the concentrations of inflammatory cytokines. The changes in gut microbiota in mice were analyzed by 16S rRNA sequencing. Results We found that SD mice had elevated latency of exploration to reach the hidden platform (p>0.05) and significantly decreased traversing times, swimming distance, and swimming time in the target zone when the hidden platform was removed (p<0.05). Sleep deprivation caused dysregulated expression in serum IL-1β, IL-6, and TNF-α in mice, and the difference was significant (all p<0.001). Tannerellaceae, Rhodospirillales, Alistipes, and Parabacteroides were significantly increased in SD mice. Correlation analysis showed IL-1β was positively correlated with the abundance of Muribaculaceae (r=0.497, p<0.05) and negatively correlated with the abundance of Lachnospiraceae (r=-0.583, p<0.05). The TNF-α was positively correlated with the abundances of Erysipelotrichaceae, Burkholderiaceae, and Tannerellaceae (r=0.492, r=0.646, r=0.726, all p<0.05). Conclusion Sleep deprivation can increase pro-inflammatory cytokine responses and learning and memory impairment in mice and may be caused by the disorder of the microbiota. These findings of this study may open avenues for potential interventions that can relieve the detrimental consequences of sleep loss.
Collapse
Affiliation(s)
- Mengjie Zhang
- School of Physical Education and Sport Science, Fujian Normal University, Fuzhou, China
- Zhengzhou University, Zhengzhou, China
| | - Mengying Zhang
- Zhengzhou University, Zhengzhou, China
- Synergetic Innovation Center of Kinesis and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, China
| | - Guangning Kou
- Centre of Sport Nutrition and Health, School of Physical Education, Zhengzhou University, Zhengzhou, China
| | - Yan Li
- Zhengzhou University, Zhengzhou, China
- Synergetic Innovation Center of Kinesis and Health, School of Physical Education (Main Campus), Zhengzhou University, Zhengzhou, China
| |
Collapse
|
29
|
Demir S, Yet I, Sardan Ekiz M, Sag E, Bilginer Y, Celikbicak O, Lay I, Ozen S. Plasma Proteomic Analysis Reveals the Potential Role of Lectin and Alternative Complement Pathways in IgA Vasculitis Pathogenesis. Diagnostics (Basel) 2023; 13:diagnostics13101729. [PMID: 37238213 DOI: 10.3390/diagnostics13101729] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND IgA vasculitis (IgAV) is the most common form of childhood vasculitis. A better understanding of its pathophysiology is required to identify new potential biomarkers and treatment targets. OBJECTIVE to assess the underlying molecular mechanisms in the pathogenesis of IgAV using an untargeted proteomics approach. METHODS Thirty-seven IgAV patients and five healthy controls were enrolled. Plasma samples were collected on the day of diagnosis before any treatment was initiated. We used nano-liquid chromatography-tandem mass spectrometry (nLC-MS/MS) to investigate the alterations in plasma proteomic profiles. For the bioinformatics analyses, databases including Uniprot, PANTHER, KEGG, Reactome, Cytoscape, and IntAct were used. RESULTS Among the 418 proteins identified in the nLC-MS/MS analysis, 20 had significantly different expressions in IgAV patients. Among them, 15 were upregulated and 5 were downregulated. According to the KEGG pathway and function classification analysis, complement and coagulation cascades were the most enriched pathways. GO analyses showed that the differentially expressed proteins were mainly involved in defense/immunity proteins and the metabolite interconversion enzyme family. We also investigated molecular interactions in the identified 20 proteins of IgAV patients. We extracted 493 interactions from the IntAct database for the 20 proteins and used Cytoscape for the network analyses. CONCLUSION Our results clearly suggest the role of the lectin and alternate complement pathways in IgAV. The proteins defined in the pathways of cell adhesion may serve as biomarkers. Further functional studies may lead the way to better understanding of the disease and new therapeutic options for IgAV treatment.
Collapse
Affiliation(s)
- Selcan Demir
- Department of Pediatric Rheumatology, Hacettepe University Medical Faculty, 06230 Ankara, Turkey
| | - Idil Yet
- Department of Bioinformatics, Graduate School of Health Sciences, Hacettepe University, 06230 Ankara, Turkey
| | - Melis Sardan Ekiz
- Advanced Technologies Application and Research Center (HUNITEK), Hacettepe University, 06230 Ankara, Turkey
| | - Erdal Sag
- Department of Pediatric Rheumatology, Hacettepe University Medical Faculty, 06230 Ankara, Turkey
| | - Yelda Bilginer
- Department of Pediatric Rheumatology, Hacettepe University Medical Faculty, 06230 Ankara, Turkey
| | - Omur Celikbicak
- Department of Chemistry, Hacettepe University, 06230 Ankara, Turkey
| | - Incilay Lay
- Department of Biochemistry, Hacettepe University Medical Faculty, 06230 Ankara, Turkey
| | - Seza Ozen
- Department of Pediatric Rheumatology, Hacettepe University Medical Faculty, 06230 Ankara, Turkey
| |
Collapse
|
30
|
Tian H, Li J, Chen X, Ren Z, Pan X, Huang W, Bhatia M, Pan LL, Sun J. Oral Delivery of Mouse β-Defensin 14 (mBD14)-Producing Lactococcus lactis NZ9000 Attenuates Experimental Colitis in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:5185-5194. [PMID: 36943701 DOI: 10.1021/acs.jafc.2c07098] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Antimicrobial peptides (AMPs) play essential roles in maintaining intestinal health and have been suggested as possible therapeutic strategies against inflammatory bowel disease (IBD). However, the instability of AMPs in the process of transmission in vivo limits their application in the treatment of IBD. In this study, we constructed the mBD14-producing Lactococcus lactis NZ9000 (L. lactis/mBD14) to achieve enteric delivery of mBD14 and evaluated its protective effect on dextran sodium sulfate (DSS)-induced colitis. Mice treated with L. lactis/mBD14 exhibited milder symptoms of colitis (P < 0.01). Additionally, L. lactis/mBD14 treatment reversed DSS-induced epithelial dysfunction and reduced the production of pro-inflammatory cytokines in colon (P < 0.01). Mechanistically, L. lactis/mBD14 significantly inhibited NOD-like receptor pyrin domain containing three inflammasome-mediated pro-inflammatory response (P < 0.05) and regulated microbiota homeostasis by promoting the abundance of probiotic bacteria Akkermansia muciniphila and Faecalibacterium prausnitzii and decreasing the pathogenic Escherichia coli (P < 0.01). Taken together, this study demonstrates the protective effect of L. lactis/mBD14 in DSS-induced colitis, and suggests that oral administration of L. lactis/mBD14 may represent a potential therapeutic strategy for IBD.
Collapse
Affiliation(s)
- Haizhi Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Xiaopei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Zhengnan Ren
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Xiaohua Pan
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Weining Huang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 9016, New Zealand
| | - Li-Long Pan
- School of Medicine, Jiangnan University, Wuxi 214126, China
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214126, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214126, China
| |
Collapse
|
31
|
Bao K, Wang M, Liu L, Zhang D, Jin C, Zhang J, Shi L. Jinhong decoction protects sepsis-associated acute lung injury by reducing intestinal bacterial translocation and improving gut microbial homeostasis. Front Pharmacol 2023; 14:1079482. [PMID: 37081964 PMCID: PMC10110981 DOI: 10.3389/fphar.2023.1079482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 03/27/2023] [Indexed: 04/07/2023] Open
Abstract
Background: Currently no specific treatments are available for sepsis and the associated syndromes including acute lung injury (ALI). Jinhong Decoction (JHD) is a traditional Chinese prescription, and it has been applied clinically as an efficient and safe treatment for sepsis, but the underlying mechanism remains unknown. The aim of the study was to explore the potential mechanisms of JHD ameliorating sepsis and concurrent ALI.Methods: The cecum ligation puncture (CLP)- induced murine sepsis model was established for determining the efficacy of JHD protecting CLP and ALI. The role of gut microbiota involved in the efficacy of JHD was evaluated by 16S rRNA sequencing and fecal microbiota transplantation (FMT). Translocation of intestinal Escherichia coli (E. coli) to lungs after CLP was verified by qPCR and in vivo-imaging. Intestinal permeability was analyzed by detecting FITC-dextran leakness. Junction proteins were evaluated by Western blotting and immunofluorescence.Results: JHD treatment remarkably increased survival rate of septic mice and alleviated sepsis-associated lung inflammation and injury. FMT suggested that the protective role for JHD was mediated through the regulation of gut microbiota. We further revealed that JHD administration partially restored the diversity and configuration of microbiome that was distorted by CLP operation. Of interest, the intestinal bacteria, E. coli particularly, was found to translocate into the lungs upon CLP via disrupting the intestinal mucosal barrier, leading to the inflammatory response and tissue damage in lungs. JHD impeded the migration and hence lung accumulation of intestinal E. coli, and thereby prevented severe ALI associated with sepsis. This effect is causatively related with the ability of JHD to restore intestinal barrier by up-regulating tight junctions.Conclusion: Our study unveils a mechanism whereby the migration of gut bacteria leads to sepsis-associated ALI, and we demonstrate the potential of JHD as an effective strategy to block this bacterial migration for treating sepsis and the associated immunopathology in the distal organs.
Collapse
Affiliation(s)
- Kaifan Bao
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Meiling Wang
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Li Liu
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Dongya Zhang
- Department of Medical Microbiology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Cuiyuan Jin
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Junfeng Zhang
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Liyun Shi
- Department of Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, Zhejiang, China
- *Correspondence: Liyun Shi,
| |
Collapse
|
32
|
Gonzalez P, Dos Santos A, Darnaud M, Moniaux N, Rapoud D, Lacoste C, Nguyen TS, Moullé VS, Deshayes A, Amouyal G, Amouyal P, Bréchot C, Cruciani-Guglielmacci C, Andréelli F, Magnan C, Faivre J. Antimicrobial protein REG3A regulates glucose homeostasis and insulin resistance in obese diabetic mice. Commun Biol 2023; 6:269. [PMID: 36918710 PMCID: PMC10015038 DOI: 10.1038/s42003-023-04616-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Innate immune mediators of pathogen clearance, including the secreted C-type lectins REG3 of the antimicrobial peptide (AMP) family, are known to be involved in the regulation of tissue repair and homeostasis. Their role in metabolic homeostasis remains unknown. Here we show that an increase in human REG3A improves glucose and lipid homeostasis in nutritional and genetic mouse models of obesity and type 2 diabetes. Mice overexpressing REG3A in the liver show improved glucose homeostasis, which is reflected in better insulin sensitivity in normal weight and obese states. Delivery of recombinant REG3A protein to leptin-deficient ob/ob mice or wild-type mice on a high-fat diet also improves glucose homeostasis. This is accompanied by reduced oxidative protein damage, increased AMPK phosphorylation and insulin-stimulated glucose uptake in skeletal muscle tissue. Oxidative damage in differentiated C2C12 myotubes is greatly attenuated by REG3A, as is the increase in gp130-mediated AMPK activation. In contrast, Akt-mediated insulin action, which is impaired by oxidative stress, is not restored by REG3A. These data highlight the importance of REG3A in controlling oxidative protein damage involved in energy and metabolic pathways during obesity and diabetes, and provide additional insight into the dual function of host-immune defense and metabolic regulation for AMP.
Collapse
Affiliation(s)
- Patrick Gonzalez
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Alexandre Dos Santos
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Marion Darnaud
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Nicolas Moniaux
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Delphine Rapoud
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Claire Lacoste
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Tung-Son Nguyen
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | - Valentine S Moullé
- Université of Paris, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, 75013, France
| | - Alice Deshayes
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France
| | | | | | | | | | - Fabrizio Andréelli
- Sorbonne Université, INSERM, NutriOmics team, Institute of Cardiometabolism and Nutrition (ICAN), Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Paris, 75013, France
| | - Christophe Magnan
- Université of Paris, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, 75013, France
| | - Jamila Faivre
- INSERM, U1193, Paul-Brousse University Hospital, Hepatobiliary Centre, Villejuif, 94800, France.
- Université Paris-Saclay, Faculté de Médecine Le Kremlin-Bicêtre, Le Kremlin-Bicêtre, 94270, France.
- Assistance Publique-Hôpitaux de Paris (AP-HP). Université Paris Saclay, Medical-University Department (DMU) Biology, Genetics, Pharmacy, Paul-Brousse Hospital, Villejuif, 94800, France.
| |
Collapse
|
33
|
Kim YM, Choi JO, Cho YJ, Hong BK, Shon HJ, Kim BJ, Park JH, Kim WU, Kim D. Mycobacterium potentiates protection from colorectal cancer by gut microbial alterations. Immunology 2023; 168:493-510. [PMID: 36183156 DOI: 10.1111/imm.13586] [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: 01/31/2022] [Accepted: 09/26/2022] [Indexed: 11/29/2022] Open
Abstract
Not only are many Mycobacteria pathogens, but they can act as strong non-specific immunopotentiators, generating beneficial effects on the pathogenesis of some diseases. However, there has been no direct evidence of the effect of mycobacterial species on colorectal cancer (CRC). Herein, we showed that there may be a meaningful inverse correlation between the incidence of tuberculosis and CRC based on global statistics and that heat-killed Mycobacterial tuberculosis and live Mycobacterium bovis (Bacillus Calmette-Guérin strain) could ameliorate CRC development. In particular, using a faecal microbiota transplantation and a comparison between separate housing and cohousing, we demonstrated that the gut microbiota is involved in the protective effects. The microbial alterations can be elucidated by the modulation of antimicrobial activities including those of the Reg3 family genes. Furthermore, interleukin-22 production by T helper cells contributed to the anti-inflammatory activity of Mycobacteria. Our results revealed a novel role of Mycobacteria involving gut microbial alterations in dampening inflammation-associated CRC and an immunological mechanism underlying the interaction between microbes and host immunity.
Collapse
Affiliation(s)
- Yu-Mi Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin-Ouk Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong-Joon Cho
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.,Institute for Basic Science, Seoul, Republic of Korea
| | - Bong-Ki Hong
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hoh-Jeong Shon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bum-Joon Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Joo-Hong Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, Republic of Korea.,Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Donghyun Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Endemic Diseases, Seoul National University Medical Research Center, Seoul, Republic of Korea
| |
Collapse
|
34
|
Ju Y, Huang L, Luo H, Huang Y, Huang X, Chen G, Gui J, Liu Z, Yang L, Liu X. Passion fruit peel and its zymolyte enhance gut function in Sanhuang broilers by improving antioxidation and short-chain fatty acids and decreasing inflammatory cytokines. Poult Sci 2023; 102:102672. [PMID: 37104904 PMCID: PMC10160589 DOI: 10.1016/j.psj.2023.102672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 03/31/2023] Open
Abstract
The passion fruit peel (PFP) is the by-product of juice processing and is rich in phenolic compounds and dietary fibers. As the high ADF content in PFP (34.20%), we proceeded to treat PFP with cellulase. The ADF decreased to 16.70% after enzymatic processing, and we supposed that enzymolytic passion fruit peel (EPF) should have a greater growth performance than PFP to broilers. Two trials were conducted to evaluate the effects of dietary PFP or EPF supplementation on growth performance, serum biochemical indices, meat quality, and cecal short-chain fatty acids, microbiota, and metabolites in broilers. In Exp. 1, 180 1-day-old Sanhuang broilers (male, 36.17 ± 2.47 g) were randomly allocated into 3 treatments, with 6 replicates in each treatment. The 3 experimental diets included 1 basal diet (control) and 2 PFP-added diets supplemented with 1 and 2% PFP, respectively. The trial lasted for 42 d. In Exp. 2, 144 Sanhuang broilers (male, 112-day-old, 1.62 ± 0.21 kg) were randomly allocated to 3 treatments. Each treatment was distributed among 6 pens, and each pen contained 8 broilers. The 3 treatment diets included: a control diet, a positive control diet supplementing 75 mg/kg chlortetracycline, and the experimental diet supplementing 3% EPF. The trial lasted for 56 d. Results showed that dietary 1 and 2% PFP addition did not affect growth performance in Exp. 1, and the 3% EPF supplementation had a negative effect on ADFI (P < 0.05) in Exp. 2. A decreased serum triglyceride (P < 0.05) in broilers was observed in Exp. 1. Broilers fed EPF had a higher glutathione peroxidase (GSH-Px) (P < 0.05), and lower levels of tumor necrosis factor-α (TNF-α) (P < 0.05) and glucose (P < 0.05) in Exp. 2. We also found that broilers from PFP or EPF-treated treatments had an increased butyrate content and higher microbial diversity in the cecum. The effects of antioxidation, anti-inflammatory function, and elevated SCFAs were confirmed after the microbe and untargeted metabolomic analysis. Dietary EPF supplementation significantly increased the SCFA-generating bacteria, anti-inflammatory-related bacteria, the antioxidant-related and anti-inflammatory-related metabolites. Moreover, dietary 3% EPF addition positively affects the biosynthesis of phenylpropanoids, which strongly correlate with the antioxidant and anti-inflammatory properties. In conclusion, the proper addition level did not affect the growth performance, and the PFP and EPF could improve the antioxidation state, anti-inflammatory activity, and intestinal functions of Sanhuang broilers to some extent.
Collapse
|
35
|
Jang KK, Heaney T, London M, Ding Y, Yeung F, Ercelen D, Chen YH, Axelrad J, Gurunathan S, Marijke Keestra-Gounder A, Griffin ME, Hang HC, Cadwell K. Antimicrobial overproduction sustains intestinal inflammation by inhibiting Enterococcus colonization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.29.526128. [PMID: 36778381 PMCID: PMC9915521 DOI: 10.1101/2023.01.29.526128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Loss of antimicrobial proteins such as REG3 family members compromises the integrity of the intestinal barrier. Here, we demonstrate that overproduction of REG3 proteins can also be detrimental by reducing a protective species in the microbiota. Patients with inflammatory bowel disease (IBD) experiencing flares displayed heightened levels of secreted REG3 proteins that mediated depletion of Enterococcus faecium ( Efm ) from the gut microbiota. Efm inoculation of mice ameliorated intestinal inflammation through activation of the innate immune receptor NOD2, which was associated with the bacterial DL-endopeptidase SagA. Microbiota sensing by NOD2 in myeloid cells mediated IL-1β secretion and increased the proportion of IL-22-producing CD4 + T helper cells and innate lymphoid cells. Finally, Efm was unable to protect mice carrying a NOD2 gene variant commonly found in IBD patients. Our findings demonstrate that inflammation self-perpetuates by causing aberrant antimicrobial activity that disrupts symbiotic relationships with gut microbes.
Collapse
|
36
|
Alam MZ, Maslanka JR, Abt MC. Immunological consequences of microbiome-based therapeutics. Front Immunol 2023; 13:1046472. [PMID: 36713364 PMCID: PMC9878555 DOI: 10.3389/fimmu.2022.1046472] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
The complex network of microscopic organisms living on and within humans, collectively referred to as the microbiome, produce wide array of biologically active molecules that shape our health. Disruption of the microbiome is associated with susceptibility to a range of diseases such as cancer, diabetes, allergy, obesity, and infection. A new series of next-generation microbiome-based therapies are being developed to treat these diseases by transplanting bacteria or bacterial-derived byproducts into a diseased individual to reset the recipient's microbiome and restore health. Microbiome transplantation therapy is still in its early stages of being a routine treatment option and, with a few notable exceptions, has had limited success in clinical trials. In this review, we highlight the successes and challenges of implementing these therapies to treat disease with a focus on interactions between the immune system and microbiome-based therapeutics. The immune activation status of the microbiome transplant recipient prior to transplantation has an important role in supporting bacterial engraftment. Following engraftment, microbiome transplant derived signals can modulate immune function to ameliorate disease. As novel microbiome-based therapeutics are developed, consideration of how the transplants will interact with the immune system will be a key factor in determining whether the microbiome-based transplant elicits its intended therapeutic effect.
Collapse
Affiliation(s)
| | | | - Michael C. Abt
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
37
|
Ouyang J, Yan J, Zhou X, Isnard S, Harypursat V, Cui H, Routy JP, Chen Y. Relevance of biomarkers indicating gut damage and microbial translocation in people living with HIV. Front Immunol 2023; 14:1173956. [PMID: 37153621 PMCID: PMC10160480 DOI: 10.3389/fimmu.2023.1173956] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 04/10/2023] [Indexed: 05/10/2023] Open
Abstract
The intestinal barrier has the daunting task of allowing nutrient absorption while limiting the entry of microbial products into the systemic circulation. HIV infection disrupts the intestinal barrier and increases intestinal permeability, leading to microbial product translocation. Convergent evidence has shown that gut damage and an enhanced level of microbial translocation contribute to the enhanced immune activation, the risk of non-AIDS comorbidity, and mortality in people living with HIV (PLWH). Gut biopsy procedures are invasive, and are not appropriate or feasible in large populations, even though they are the gold standard for intestinal barrier investigation. Thus, validated biomarkers that measure the degree of intestinal barrier damage and microbial translocation are needed in PLWH. Hematological biomarkers represent an objective indication of specific medical conditions and/or their severity, and should be able to be measured accurately and reproducibly via easily available and standardized blood tests. Several plasma biomarkers of intestinal damage, i.e., intestinal fatty acid-binding protein (I-FABP), zonulin, and regenerating islet-derived protein-3α (REG3α), and biomarkers of microbial translocation, such as lipopolysaccharide (LPS) and (1,3)-β-D-Glucan (BDG) have been used as markers of risk for developing non-AIDS comorbidities in cross sectional analyses and clinical trials, including those aiming at repair of gut damage. In this review, we critically discuss the value of different biomarkers for the estimation of gut permeability levels, paving the way towards developing validated diagnostic and therapeutic strategies to repair gut epithelial damage and to improve overall disease outcomes in PLWH.
Collapse
Affiliation(s)
- Jing Ouyang
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Jiangyu Yan
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Xin Zhou
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Stéphane Isnard
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
- Canadian HIV Trials Network, Canadian Institutes for Health Research, Vancouver, BC, Canada
| | - Vijay Harypursat
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Jean-Pierre Routy
- Infectious Diseases and Immunity in Global Health Program, Research Institute, McGill University Health Centre, Montréal, QC, Canada
- Chronic Viral Illness Service, McGill University Health Centre, Montréal, QC, Canada
- Division of Hematology, McGill University Health Centre, Montréal, QC, Canada
- *Correspondence: Jean-Pierre Routy, ; Yaokai Chen,
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
- *Correspondence: Jean-Pierre Routy, ; Yaokai Chen,
| |
Collapse
|
38
|
Xiao W, Zhao X, Li C, Huang Q, He A, Liu G. The efficacy of probiotics on the prevention of pouchitis for patients after ileal pouch-anal anastomosis: A meta-analysis. Technol Health Care 2023; 31:401-415. [PMID: 36278367 DOI: 10.3233/thc-220402] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
BACKGROUND To date, a few studies indicated that probiotics are beneficial to pouchitis, but no meta-analyses summarized the outcomes of probiotics in pouchitis in detail. OBJECTIVE This meta-analysis discusses probiotics in the prevention of pouchitis for patients after ileal pouch-anal anastomosis (IPAA) and the relationship between probiotics preventive effect and the duration of therapy and history. METHODS PubMed, EMBASE and Cochrane Library databases were searched from inception until February 2022. Risk ratio (RR), mean difference (MD) and their 95% confidence interval (CI) were analyzed by Review Manager 5.3. The subgroup analysis was also performed to explore the agent for influencing outcomes. RESULTS A total of 8 studies were included in this meta-analysis. The incidence of pouchitis in probiotics was significantly lower than that in the control (RR = 0.19, 95%CI [0.12, 0.32], Pï¼ 0.00001), and the PDAI (pouchitis disease activity index) in probiotics was also significantly lower (MD =-5.65, 95%CI [-9.48, -1.83]). After the subgroup analysis, we found that probiotics work better in the short-term (RR = 0.12, 95%CI [0.04, 0.40], P= 0.0004), but may not achieve the desired effect in the long-term (RR = 1.20, 95%CI [0.40, 3.60], P= 0.75). CONCLUSIONS Probiotics are beneficial in the prevention of pouchitis after IPAA, especially in the short-term.
Collapse
Affiliation(s)
- Wanyi Xiao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinyu Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunqiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Qianpeng Huang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Anqi He
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| | - Gang Liu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin, China
| |
Collapse
|
39
|
Gao N, Yang Y, Liu S, Fang C, Dou X, Zhang L, Shan A. Gut-Derived Metabolites from Dietary Tryptophan Supplementation Quench Intestinal Inflammation through the AMPK-SIRT1-Autophagy Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:16080-16095. [PMID: 36521060 DOI: 10.1021/acs.jafc.2c05381] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Tryptophan has drawn wide attention due to its involvement in improving intestinal immune defense directly and indirectly by regulating metabolic pathways. The study aims to elucidate the potential modulating roles of tryptophan to protect against intestinal inflammation and elucidate the underlying molecular mechanisms. The protective effects of tryptophan against intestinal inflammation are examined in the lipopolysaccharide (LPS)-induced inflammatory model. We first found that tryptophan markedly (p < 0.01) inhibited proinflammatory cytokines production and nuclear factor κB (NF-κB) pathway activation upon LPS challenge. Next, we demonstrated that tryptophan (p < 0.05) attenuated LPS-caused intestinal mucosal barrier damage by increasing the number of goblet cells, mucins, and antimicrobial peptides (AMPs) in the ileum of mice. In addition, tryptophan (p < 0.05) inhibited LPS-induced autophagic flux through the AMP-activated protein kinase (AMPK)-sirtuin 1 (SIRT1) pathway in the intestinal systems to maintain autophagy homeostasis. Meanwhile, tryptophan also reshaped the gut microbiota composition in LPS-challenge mice by increasing the abundance of short-chain fatty acid (SCFA)-producing bacteria such as Acetivibrio (0.053 ± 0.017 to 0.21 ± 0.0041%). Notably, dietary tryptophan resulted in the activation of metabolic pathways during the inflammatory response. Furthermore, exogenous treatment of tryptophan metabolites kynurenine (Kyn) and 5-HT in porcine intestinal epithelial cells (IPEC-J2 cells) reproduced similar protective effects as tryptophan to attenuate LPS-induced intestinal inflammation through regulating the AMPK-SIRT1-autophagy. Taken together, the present study indicates that tryptophan exhibits intestinal protective and immunoregulatory effects resulting from the activation of metabolic pathways, maintenance of gut mucosal barrier integrity, microbiota composition, and AMPK-SIRT1-autophagy level.
Collapse
Affiliation(s)
- Nan Gao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Yang Yang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Siqi Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Chunyang Fang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Xiujing Dou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Licong Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| | - Anshan Shan
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
40
|
Shin JH, Bozadjieva-Kramer N, Shao Y, Lyons-Abbott S, Rupp AC, Sandoval DA, Seeley RJ. The gut peptide Reg3g links the small intestine microbiome to the regulation of energy balance, glucose levels, and gut function. Cell Metab 2022; 34:1765-1778.e6. [PMID: 36240758 PMCID: PMC9633559 DOI: 10.1016/j.cmet.2022.09.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/12/2022] [Accepted: 09/26/2022] [Indexed: 02/08/2023]
Abstract
Changing composition of the gut microbiome is an important component of the gut adaptation to various environments, which have been implicated in various metabolic diseases including obesity and type 2 diabetes, but the mechanisms by which the microbiota influence host physiology remain contentious. Here we find that both diets high in the fermentable fiber inulin and vertical sleeve gastrectomy increase intestinal expression and circulating levels of the anti-microbial peptide Reg3g. Moreover, a number of beneficial effects of these manipulations on gut function, energy balance, and glucose regulation are absent in Reg3g knockout mice. Peripheral administration of various preparations of Reg3g improves glucose tolerance, and this effect is dependent on the putative receptor Extl3 in the pancreas. These data suggest Reg3g acts both within the lumen and as a gut hormone to link the intestinal microbiome to various aspects of host physiology that may be leveraged for novel treatment strategies.
Collapse
Affiliation(s)
- Jae Hoon Shin
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Veterans Affairs Ann Arbor Healthcare System, Research Service, Ann Arbor, MI, USA
| | - Yikai Shao
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Center for Obesity and Metabolic Surgery, Huashan Hospital of Fudan University, Shanghai, China
| | | | - Alan C Rupp
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Darleen A Sandoval
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA; Department of Pediatrics Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, USA
| | - Randy J Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
41
|
Jo JK, Lee G, Nguyen CD, Park SE, Kim EJ, Kim HW, Seo SH, Cho KM, Kwon SJ, Kim JH, Son HS. Effects of Donepezil Treatment on Brain Metabolites, Gut Microbiota, and Gut Metabolites in an Amyloid Beta-Induced Cognitive Impairment Mouse Pilot Model. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196591. [PMID: 36235127 PMCID: PMC9572896 DOI: 10.3390/molecules27196591] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 12/12/2022]
Abstract
Accumulated clinical and biomedical evidence indicates that the gut microbiota and their metabolites affect brain function and behavior in various central nervous system disorders. This study was performed to investigate the changes in brain metabolites and composition of the fecal microbial community following injection of amyloid β (Aβ) and donepezil treatment of Aβ-injected mice using metataxonomics and metabolomics. Aβ treatment caused cognitive dysfunction, while donepezil resulted in the successful recovery of memory impairment. The Aβ + donepezil group showed a significantly higher relative abundance of Verrucomicrobia than the Aβ group. The relative abundance of 12 taxa, including Blautia and Akkermansia, differed significantly between the groups. The Aβ + donepezil group had higher levels of oxalate, glycerol, xylose, and palmitoleate in feces and oxalate, pyroglutamic acid, hypoxanthine, and inosine in brain tissues than the Aβ group. The levels of pyroglutamic acid, glutamic acid, and phenylalanine showed similar changes in vivo and in vitro using HT-22 cells. The major metabolic pathways in the brain tissues and gut microbiota affected by Aβ or donepezil treatment of Aβ-injected mice were related to amino acid pathways and sugar metabolism, respectively. These findings suggest that alterations in the gut microbiota might influence the induction and amelioration of Aβ-induced cognitive dysfunction via the gut–brain axis. This study could provide basic data on the effects of Aβ and donepezil on gut microbiota and metabolites in an Aβ-induced cognitive impairment mouse model.
Collapse
Affiliation(s)
- Jae-Kwon Jo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Gihyun Lee
- Department of Korean Medicine, Dongshin University, Naju 58245, Korea
| | - Cong Duc Nguyen
- Department of Korean Medicine, Dongshin University, Naju 58245, Korea
| | - Seong-Eun Park
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Eun-Ju Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | - Hyun-Woo Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
| | | | | | | | - Jae-Hong Kim
- Department of Acupuncture and Moxibustion Medicine, College of Korean Medicine, Dongshin University, Naju 58245, Korea
- Correspondence: (J.-H.K.); (H.-S.S.); Tel.: +82-62-350-7209 (J.-H.K.); +82-2-3290-3053 (H.-S.S.)
| | - Hong-Seok Son
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Korea
- Correspondence: (J.-H.K.); (H.-S.S.); Tel.: +82-62-350-7209 (J.-H.K.); +82-2-3290-3053 (H.-S.S.)
| |
Collapse
|
42
|
Ma Y, Liu T, Li X, Kong A, Xiao R, Xie R, Gao J, Wang Z, Cai Y, Zou J, Yang L, Wang L, Zhao J, Xu H, Margaret W, Xu X, Gustafsson JA, Fan X. Estrogen receptor β deficiency impairs gut microbiota: a possible mechanism of IBD-induced anxiety-like behavior. MICROBIOME 2022; 10:160. [PMID: 36175956 PMCID: PMC9520828 DOI: 10.1186/s40168-022-01356-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/24/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Although the lack of estrogen receptor β (ERβ) is a risk factor for the development of inflammatory bowel disease (IBD) and psychiatric disorders, the underlying cellular and molecular mechanisms are not fully understood. Herein, we revealed the role of gut microbiota in the development of IBD and related anxiety-like behavior in ERβ-deficient mice. RESULTS In response to dextran sodium sulfate (DSS) insult, the ERβ knockout mice displayed significant shift in α and β diversity in the fecal microbiota composition and demonstrated worsening of colitis and anxiety-like behaviors. In addition, DSS-induced colitis also induced hypothalamic-pituitary-adrenal (HPA) axis hyperactivity in ERβ-deficient mice, which was associated with colitis and anxiety-like behaviors. In addition, RNA sequencing data suggested that ErbB4 might be the target of ERβ that is involved in regulating the HPA axis hyperactivity caused by DSS insult. Gut microbiota remodeling by co-housing showed that both the colitis and anxiety-like behaviors were aggravated in co-housed wild-type mice compared to single-housed wild-type mice. These findings suggest that gut microbiota play a critical role in mediating colitis disease activity and anxiety-like behaviors via aberrant neural processing within the gut-brain axis. CONCLUSIONS ERβ has the potential to inhibit colitis development and anxiety-like behaviors via remodeling of the gut microbiota, which suggests that ERβ is a promising therapeutic target for the treatment of IBD and related anxiety-like behaviors. Video Abstract.
Collapse
Affiliation(s)
- Yuanyuan Ma
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Xin Li
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Anqi Kong
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Rui Xiao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Ruxin Xie
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Junwei Gao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Zhongke Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Yun Cai
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Jiao Zou
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Ling Yang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Lian Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Jinghui Zhao
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China
| | - Haiwei Xu
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Warner Margaret
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden
| | - Xingshun Xu
- Institute of Neuroscience, Soochow University, Suzhou, China.
| | - Jan-Ake Gustafsson
- Center for Innovative Medicine, Department of Biosciences and Nutrition, Karolinska Institute, Stockholm, Sweden.
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, USA.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, China.
| |
Collapse
|
43
|
Huangshan Maofeng Green Tea Extracts Prevent Obesity-Associated Metabolic Disorders by Maintaining Homeostasis of Gut Microbiota and Hepatic Lipid Classes in Leptin Receptor Knockout Rats. Foods 2022; 11:foods11192939. [PMID: 36230016 PMCID: PMC9562686 DOI: 10.3390/foods11192939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
Huangshan Maofeng green tea (HMGT) is one of the most well-known green teas consumed for a thousand years in China. Research has demonstrated that consumption of green tea effectively improves metabolic disorders. However, the underlying mechanisms of obesity prevention are still not well understood. This study investigated the preventive effect and mechanism of long-term intervention of Huangshan Maofeng green tea water extract (HTE) on obesity-associated metabolic disorders in leptin receptor knockout (Lepr−/−) rats by using gut microbiota and hepatic lipidomics data. The Lepr−/− rats were administered with 700 mg/kg HTE for 24 weeks. Our results showed that HTE supplementation remarkably reduced excessive fat accumulation, as well as ameliorated hyperlipidemia and hepatic steatosis in Lepr−/− rats. In addition, HTE increased gut microbiota diversity and restored the relative abundance of the microbiota responsible for producing short chain fatty acids, including Ruminococcaceae, Faecalibaculum, Veillonellaceae, etc. Hepatic lipidomics analysis found that HTE significantly recovered glycerolipid and glycerophospholipid classes in the liver of Lepr−/− rats. Furthermore, nineteen lipid species, mainly from phosphatidylcholines (PCs), phosphatidylethanolamines (PEs), and triglycerides (TGs), were significantly restored increases, while nine lipid species from TGs and diglycerides (DGs) were remarkably recovered decreases by HTE in the liver of Lepr−/− rats. Our results indicated that prevention of obesity complication by HTE may be possible through maintaining homeostasis of gut microbiota and certain hepatic lipid classes.
Collapse
|
44
|
Azmy AM, Abd Elbaki BT, Ali MA, Mahmoud AA. Effect of ozone versus naringin on testicular injury in experimentally induced ulcerative colitis in adult male albino rats. Ultrastruct Pathol 2022; 46:439-461. [DOI: 10.1080/01913123.2022.2132337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Abeer M. Azmy
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Bassant T. Abd Elbaki
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mohammed A. Ali
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abeer A Mahmoud
- Medical Histology and Cell Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
45
|
Negi S, Hashimoto-Hill S, Alenghat T. Neonatal microbiota-epithelial interactions that impact infection. Front Microbiol 2022; 13:955051. [PMID: 36090061 PMCID: PMC9453604 DOI: 10.3389/fmicb.2022.955051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/31/2022] [Indexed: 11/13/2022] Open
Abstract
Despite modern therapeutic developments and prophylactic use of antibiotics during birth or in the first few months of life, enteric infections continue to be a major cause of neonatal mortality and morbidity globally. The neonatal period is characterized by initial intestinal colonization with microbiota and concurrent immune system development. It is also a sensitive window during which perturbations to the environment or host can significantly impact colonization by commensal microbes. Extensive research has demonstrated that these early life alterations to the microbiota can lead to enhanced susceptibility to enteric infections and increased systemic dissemination in newborns. Various contributing factors continue to pose challenges in prevention and control of neonatal enteric infections. These include alterations in the gut microbiota composition, impaired immune response, and effects of maternal factors. In addition, there remains limited understanding for how commensal microbes impact host-pathogen interactions in newborns. In this review, we discuss the recent recognition of initial microbiota-epithelial interactions that occur in neonates and can regulate susceptibility to intestinal infection. These studies suggest the development of neonatal prophylactic or therapeutic regimens that include boosting epithelial defense through microbiota-directed interventions.
Collapse
|
46
|
Fu Q, Song T, Ma X, Cui J. Research progress on the relationship between intestinal microecology and intestinal bowel disease. Animal Model Exp Med 2022; 5:297-310. [PMID: 35962562 PMCID: PMC9434592 DOI: 10.1002/ame2.12262] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 06/21/2022] [Indexed: 12/02/2022] Open
Abstract
Intestinal microecology is the main component of human microecology. Intestinal microecology consists of intestinal microbiota, intestinal epithelial cells, and intestinal mucosal immune system. These components are interdependent and establish a complex interaction network that restricts each other. According to the impact on the human body, there are three categories of symbiotic bacteria, opportunistic pathogens, and pathogenic bacteria. The intestinal microecology participates in digestion and absorption, and material metabolism, and inhibits the growth of pathogenic microorganisms. It also acts as the body's natural immune barrier, regulates the innate immunity of the intestine, controls the mucosal barrier function, and also participates in the intestinal epithelial cells' physiological activities such as hyperplasia or apoptosis. When the steady‐state balance of the intestinal microecology is disturbed, the existing core intestinal microbiota network changes and leads to obesity, diabetes, and many other diseases, especially irritable bowel syndrome, inflammatory bowel disease (IBD), and colorectal malignancy. Intestinal diseases, including tumors, are particularly closely related to intestinal microecology. This article systematically discusses the research progress on the relationship between IBD and intestinal microecology from the pathogenesis, treatment methods of IBD, and the changes in intestinal microbiota.
Collapse
Affiliation(s)
- Qianhui Fu
- School of Pharmacy, Minzu University of China, Beijing, China.,Ministry of Education, Key Laboratory of Ethnomedicine, Minzu University of China, Beijing, China
| | - Tianyuan Song
- School of Pharmacy, Minzu University of China, Beijing, China.,Ministry of Education, Key Laboratory of Ethnomedicine, Minzu University of China, Beijing, China
| | - Xiaoqin Ma
- School of Pharmacy, Minzu University of China, Beijing, China.,Ministry of Education, Key Laboratory of Ethnomedicine, Minzu University of China, Beijing, China
| | - Jian Cui
- School of Pharmacy, Minzu University of China, Beijing, China.,Ministry of Education, Key Laboratory of Ethnomedicine, Minzu University of China, Beijing, China
| |
Collapse
|
47
|
Le Lay A, Philippe E, Roth F, Sanchez-Archidona AR, Mehl F, Denom J, Prasad R, Asplund O, Hansson O, Ibberson M, Andreelli F, Santoro L, Amouyal P, Amouyal G, Brechot C, Jamot L, Cruciani-Guglielmacci C, Magnan C. Regenerating islet-derived protein 3α: A promising therapy for diabetes. Preliminary data in rodents and in humans. Heliyon 2022; 8:e09944. [PMID: 35874080 PMCID: PMC9304733 DOI: 10.1016/j.heliyon.2022.e09944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/13/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
The aim of our study was to test the hypothesis that administration of Regenerating islet-derived protein 3α (Reg3α), a protein described as having protective effects against oxidative stress and anti-inflammatory activity, could participate in the control of glucose homeostasis and potentially be a new target of interest in the treatment of type 2 diabetes. To that end the recombinant human Reg3α protein was administered for one month in insulin-resistant mice fed high fat diet. We performed glucose and insulin tolerance tests, assayed circulating chemokines in plasma and measured glucose uptake in insulin sensitive tissues. We evidenced an increase in insulin sensitivity during an oral glucose tolerance test in ALF-5755 treated mice vs controls and decreased the pro-inflammatory cytokine C-X-C Motif Chemokine Ligand 5 (CXCL5). We also demonstrated an increase in glucose uptake in skeletal muscle. Finally, correlation studies using human and mouse muscle biopsies showed negative correlation between intramuscular Reg3α mRNA expression (or its murine isoform Reg3γ) and insulin resistance. Thus, we have established the proof of concept that Reg3α could be a novel molecule of interest in the treatment of T2D by increasing insulin sensitivity via a skeletal muscle effect.
Collapse
Affiliation(s)
- Aurélie Le Lay
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | - Erwann Philippe
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | - Fanny Roth
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | | | - Florence Mehl
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | - Jessica Denom
- Université de Paris, BFA, UMR 8251, CNRS, F-75013 Paris, France
| | - Rashmi Prasad
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Olof Asplund
- Department of Clinical Sciences, Lund University, Malmö, Sweden.,Institute for Molecular Medicine Finland (FIMM), Helsinki University, Helsinki, Finland
| | - Ola Hansson
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute for Bioinformatics, 1015 Lausanne, Switzerland
| | - Fabrizio Andreelli
- Nutrition and Obesities; Systemic Approaches (NutriOmics), Sorbonne Université, INSERM; Pitié-Salpêtrière Hospital, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Lyse Santoro
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | - Paul Amouyal
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | - Gilles Amouyal
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | - Christian Brechot
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France.,University of South Florida, Tampa, FL 33612, USA
| | - Laure Jamot
- The Healthy Aging Company, Incubateur Paris Biotech Santé, F-75014 Paris, France
| | | | | |
Collapse
|
48
|
Longitudinal 16S rRNA Sequencing Reveals Relationships among Alterations of Gut Microbiota and Nonalcoholic Fatty Liver Disease Progression in Mice. Microbiol Spectr 2022; 10:e0004722. [PMID: 35647690 PMCID: PMC9241867 DOI: 10.1128/spectrum.00047-22] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a prevalent and progressive disease spectrum ranging from nonalcoholic fatty liver (NAFL) to nonalcoholic steatohepatitis (NASH), yet there is no effective treatment and efficient noninvasive diagnostic method for NASH. The present study investigated the longitudinal alternations of gut microbiota in the Western diet (WD) induced murine NAFLD model using 16S rRNA sequencing. Evident steatosis and inflammation were detected in the liver at the 8th and 12th week, while prompted hepatic oxidative injury and fibrosis were found at the 16th week. In this progressive process, impaired bile acid (BA) metabolism plays a vital part. Long-term WD intervention alters microbial richness and composition in the intestine, shaping characteristic microbial feature correspondence to each NAFLD stage. Descending abundances of Clostridia and Ruminococcaceae were found in NAFLD progression, while inflammation-related microbes [Eubacterium]_fissicatena_group, Romboutsia, and Erysipelatoclostridium were verified to identify borderline NASH at 8th and 12th week, and BA-associated taxa Dubosiella, Bosea, Helicobacter, and Alistipes were recognized as special symbols reflecting the state of oxidative damage and fibrosis in NASH at 16th week. Further, feces and colon abundances of Akkermansia were verified to be depleted in the process of borderline NASH progressed to NASH, and exhibited substantial correlations with NAFLD indexes ALT, AST, TC, and TBA. These characteristic taxa were effective to identify NAFLD and NASH, and microbiota-derived predictive models for NAFLD and NASH exhibited great potential (AUC 0.983 and 0.784). These findings demonstrate that a core set of gut microbiome especially BA-related taxa may be adopted as a noninvasive diagnostic tool for NAFLD and NASH. IMPORTANCE This study concentrates on longitudinal alternations of gut microbiota in NAFLD progression and discovers the interrelationships between them. These findings may uncover the role of gut microbiota in NAFLD progression and identify novel noninvasive diagnostic tools for NAFLD based on microbial biomarkers.
Collapse
|
49
|
Liu S, Cao Y, Ma L, Sun J, Ramos-Mucci L, Ma Y, Yang X, Zhu Z, Zhang J, Xiao B. Oral antimicrobial peptide-EGCG nanomedicines for synergistic treatment of ulcerative colitis. J Control Release 2022; 347:544-560. [PMID: 35580812 DOI: 10.1016/j.jconrel.2022.05.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 04/30/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022]
Abstract
The pathogenesis of ulcerative colitis (UC) is associated with severe inflammation, damaged colonic barriers, increased oxidative stress, and intestinal dysbiosis. The majority of current medications strive to alleviate inflammation but fail to target additional disease pathologies. Addressing multiple symptoms using a single 'magic bullet' remains a challenge. To overcome this, a smart epigallocatechin-3-gallate (EGCG)-loaded silk fibroin-based nanoparticle (NP) with the surface functionalization of antimicrobial peptides (Cathelicidin-BF, CBF) was constructed, which were internalized by Colon-26 cells and RAW 264.7 macrophages with high efficiencies. These CBF-EGCG-NPs efficiently restored colonic epithelial barriers by relieving oxidative stress and promoting epithelium migration. They also alleviated immune responses through the downregulation of pro-inflammatory factors, upregulation of anti-inflammatory factors, M2 macrophage polarization, and lipopolysaccharide (LPS) elimination. Interestingly, oral administration of hydrogel (chitosan/alginate)-embedding CBF-EGCG-NPs could not only retard progression and treat UC, but also modulate intestinal microbiota by increasing their overall diversity and richness and augmenting the abundance of beneficial bacteria (e.g., Firmicutes and Lactobacillaceae). Our work provides a "many birds with one stone" strategy for addressing UC symptoms using a single NP-based oral platform that targets immune microenvironment modulation, LPS clearance, and microbial remodeling.
Collapse
Affiliation(s)
- Shengsheng Liu
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Yingui Cao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Lingli Ma
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Jianfeng Sun
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7LD, UK
| | - Lorenzo Ramos-Mucci
- Botnar Research Centre, Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7LD, UK
| | - Ya Ma
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Beibei, Chongqing 400715, China
| | - Xiao Yang
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Shapingba, Chongqing 400038, China
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China; Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
50
|
Methamphetamine induces intestinal injury by altering gut microbiota and promoting inflammation in mice. Toxicol Appl Pharmacol 2022; 443:116011. [PMID: 35390362 DOI: 10.1016/j.taap.2022.116011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/06/2023]
Abstract
Methamphetamine (METH) is a psychostimulant abused worldwide. Its abuse induces intestinal toxicity. Moreover, the gut microbiota is altered by drugs, which induces intestinal injury. Whether gut microbiota mediates METH-induced intestinal toxicity remains to be validated. In the present study, wild-type and TLR4-/- mice were treated with METH. Gut microbiota was determined using 16S rRNA gene sequencing. Transcriptomics of the intestinal mucosa was performed by RNA-Sequencing. Blood levels of pro-inflammatory cytokines and lipopolysaccharide (LPS), the intestinal barrier, and inflammation were also assessed. METH treatment weakened the intestinal barrier and increased pro-inflammatory cytokines and LPS levels in the blood. Moreover, METH treatment significantly decreased the diversity of probiotics but increased the abundance of pathogenic gut microbiota, contributing to the over-production of LPS and disruption of intestinal barrier. Inflammatory pathways were enriched in the intestinal mucosa of METH-treated mice by KEGG analysis. Consistently, activation of the TLR4 pathway was determined in METH-treated mice, which confirmed intestinal inflammation. However, pretreatment with antibiotics or Tlr4 silencing significantly alleviated METH-induced gut microbiota dysbiosis, LPS over-production, intestinal inflammation, and disruption of the intestinal barrier. These findings suggested that the gut microbiota and LPS-mediated inflammation took an important role in METH-induced intestinal injury. Taken together, these findings suggest that METH-induced intestinal injury is mediated by gut microbiota dysbiosis and LPS-associated inflammation.
Collapse
|