1
|
Smail SW, Albarzinji N, Salih RH, Taha KO, Hirmiz SM, Ismael HM, Noori MF, Azeez SS, Janson C. Microbiome dysbiosis in SARS-CoV-2 infection: implication for pathophysiology and management strategies of COVID-19. Front Cell Infect Microbiol 2025; 15:1537456. [PMID: 40330025 PMCID: PMC12052750 DOI: 10.3389/fcimb.2025.1537456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/27/2025] [Indexed: 05/08/2025] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of coronavirus disease 2019 (COVID-19), in late 2019 initiated a global health crisis marked by widespread infection, significant mortality, and long-term health implications. While SARS-CoV-2 primarily targets the respiratory system, recent findings indicate that it also significantly disrupts the human microbiome, particularly the gut microbiota, contributing to disease severity, systemic inflammation, immune dysregulation, and increased susceptibility to secondary infections and chronic conditions. Dysbiosis, or microbial imbalance, exacerbates the clinical outcomes of COVID-19 and has been linked to long-COVID, a condition affecting a significant proportion of survivors and manifesting with over 200 symptoms across multiple organ systems. Despite the growing recognition of microbiome alterations in COVID-19, the precise mechanisms by which SARS-CoV-2 interacts with the microbiome and influences disease progression remain poorly understood. This narrative review investigates the impact of SARS-CoV-2 on host-microbiota dynamics and evaluates its implications in disease severity and for developing personalized therapeutic strategies for COVID-19. Furthermore, it highlights the dual role of the microbiome in modulating disease progression, and as a promising target for advancing diagnostic, prognostic, and therapeutic approaches in managing COVID-19.
Collapse
Affiliation(s)
- Shukur Wasman Smail
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | | | - Rebaz Hamza Salih
- Department of Respiratory Medicine, PAR Private Hospital, Erbil, Kurdistan Region, Iraq
| | - Kalthum Othman Taha
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Sarah Mousa Hirmiz
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Hero M. Ismael
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Marwa Fateh Noori
- College of Pharmacy, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq
| | - Sarkar Sardar Azeez
- Department of Medical Laboratory Technology, Soran Technical College, Erbil Polytechnic University, Erbil, Kurdistan Region, Iraq
| | - Christer Janson
- Department of Medical Science, Respiratory Medicine, and Allergology, Uppsala University and University Hospital, Uppsala, Sweden
| |
Collapse
|
2
|
Xie Y, Tao M, Yan X, Fan X, Bayoude A, Lu Y, Zhao S, Yu B, Li R. Acidic polysaccharide CP-2 from Dioscoreae Rhizoma ameliorated acute alcoholic liver injury through the gut-liver axis and AMPK/PPAR pathway. Int J Biol Macromol 2025; 310:143145. [PMID: 40233909 DOI: 10.1016/j.ijbiomac.2025.143145] [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: 01/20/2025] [Revised: 03/31/2025] [Accepted: 04/12/2025] [Indexed: 04/17/2025]
Abstract
Dioscoreae Rhizoma polysaccharides exhibit gastrointestinal protective properties, yet their efficacy against acute alcoholic liver injury (AALI) remains unexplored. This study identifies a novel acidic heteropolysaccharide (CP-2, Mw = 8.4 × 103 kDa) with galactose/galacturonic acid dominance and delineates its multimodal hepatoprotective mechanisms. In AALI mice, CP-2 attenuated liver injury by enhancing ADH and ALDH activities while restoring redox balance via SOD/CAT activation and MDA reduction, and suppressed inflammation by inhibiting IL-1β, IL-6, and TNF-α levels. Gut-liver axis modulation was achieved through intestinal barrier reinforcement (ZO-1, Occludin, Claudin-1) and microbiota rebalancing. CP-2 could reduce gram-negative bacteria ([Ruminococcus]_ torques_ group and Escherichia- Shigella) and Proteobacteria abundance while enriching Bacteroides and Akkermansia abundance, which collectively suppressed serum LPS level. In addition, CP-2 could activate the AMPK/PPAR signaling pathway to reduce the production of fatty acids and promote their degradation in AALI. CP-2 can improve AALI by adjusting the composition of gut microbiota, repairing intestinal barrier function, decreasing systemic inflammation and oxidative reactions, and regulating the AMPK/PPAR pathway. Our findings unveil CP-2 as a prebiotic candidate for AALI intervention and may advance functional food development for alcohol-related hepatopathies.
Collapse
Affiliation(s)
- Yujun Xie
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mingxing Tao
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaodong Yan
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Xinxin Fan
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 8128582, Japan
| | - Alamusi Bayoude
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Lu
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 8128582, Japan
| | - Shuangli Zhao
- Division of Pharmaceutical Cell Biology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 8128582, Japan
| | - Boyang Yu
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Renshi Li
- Jiangsu Provincial Key Laboratory of TCM Evaluation and Translational Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
3
|
Wang J, Hou Y, Mu L, Yang M, Ai X. Gut microbiota contributes to the intestinal and extraintestinal immune homeostasis by balancing Th17/Treg cells. Int Immunopharmacol 2024; 143:113570. [PMID: 39547012 DOI: 10.1016/j.intimp.2024.113570] [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: 08/09/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/17/2024]
Abstract
Gut microbiota is generally considered to play an important role in host health due to its extensive immunomodulatory activities. Th17 and Treg cells are two important CD4+ T cell subsets involved in immune regulation, and their imbalance is closely tied to many immune diseases. Recently, abundant researches have highlighted the importance of gut microbiota in supporting intestinal and extraintestinal immunity through the balance of Th17 and Treg cells. Here, we presented a comprehensive review of these findings. This review first provided an overview of gut microbiota, along with Th17/Treg cell differentiation and cytokine production. Subsequently, the review summarized the regulatory effects of gut microbiota (in terms of species, components, and metabolites) on the Th17/Treg cell balance in the local intestines and extraintestinal organs, such as lung, liver, brain, kidney, and bone. Specifically, the Th17 and Treg cells that can be modulated by gut microbiota originate not only from the gut and extraintestinal organs, but also from peripheral blood and spleen. Then, the microbial therapeutics, including probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT), were also reviewed because of their therapeutic potentials in addressing intestinal and extraintestinal diseases via the Th17/Treg axis. Finally, the review discussed the clinical applications and future study prospects of microbial therapeutics by targeting the Th17/Treg cell balance. In conclusion, this review focused on elucidating the regulatory effects of gut microbiota in balancing Th17/Treg cells to maintain intestinal and extraintestinal immune homeostasis, contributing to the further development and promotion of microbial therapeutics.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Yaqin Hou
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Lifeng Mu
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China
| | - Ming Yang
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| | - Xiaopeng Ai
- Department of Pharmacy, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China; Department of Pharmacy, North Sichuan Medical College, Nanchong 637000, China.
| |
Collapse
|
4
|
Johansen SU, Goll R, Nordborg A, Vernstad K, Jensen EPH, Florholmen JR, Hansen T. Plasma Levels of Organic Acids Associated with the Gut Microbiome Display Significant Alterations in Neuroendocrine Tumor Patients. Neuroendocrinology 2024; 115:283-294. [PMID: 39715594 DOI: 10.1159/000543247] [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: 09/10/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION The gut microbiome, allegedly involved in both healthy homeostasis and development of disease, is found to be associated with several types of cancer. Short-chain fatty acids (SCFAs), important metabolites derived from the gut microbiota, are described to carry both protective and promoting features in cancer development. Limited research exists on neuroendocrine tumors (NETs) and their association with microbiota-derived SCFAs. The aim of this study was to investigate possible alterations in plasma SCFAs/organic acids in NET patients compared to healthy controls. METHODS We quantified 11 organic acids, including SCFAs, in plasma from 109 NET patients (49 curatively operated patients and 60 patients with distant metastasis) as well as 20 healthy controls. Acids were quantified using liquid chromatography tandem mass spectrometry. RESULTS We found that levels of 3OH-propionic acid, 3OH-butyric acid, lactic acid, formic acid, acetic acid, glyoxylic acid, and glycolic acid were significantly altered in NET patients with metastatic disease, as well as curatively operated NET patients, compared to healthy controls (p < 0.05). In addition, a trend displaying increased acid level alterations from healthy controls in curatively operated patients with future recurrence, compared to patients with no documented recurrent disease, was detected. CONCLUSION Our results demonstrating significantly altered levels of multiple organic acids in NET patients represents a novel finding implicating further research on their role in NET pathophysiology.
Collapse
Affiliation(s)
- Silje Udjus Johansen
- Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway
- Medical Gastroenterology, Division of Internal medicine, University Hospital of North Norway, Tromsø, Norway
| | - Rasmus Goll
- Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway
- Medical Gastroenterology, Division of Internal medicine, University Hospital of North Norway, Tromsø, Norway
| | - Anna Nordborg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Kai Vernstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | | | - Jon Ragnar Florholmen
- Department of Clinical Medicine, UiT the Arctic University of Norway, Tromsø, Norway
- Medical Gastroenterology, Division of Internal medicine, University Hospital of North Norway, Tromsø, Norway
| | - Terkel Hansen
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
- Department of Pharmacy, UiT the Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
5
|
Min M, Nadora D, Chakkalakal M, Afzal N, Subramanyam C, Gahoonia N, Pan A, Thacker S, Nong Y, Chambers CJ, Sivamani RK. An Oral Botanical Supplement Improves Small Intestinal Bacterial Overgrowth (SIBO) and Facial Redness: Results of an Open-Label Clinical Study. Nutrients 2024; 16:3149. [PMID: 39339748 PMCID: PMC11435404 DOI: 10.3390/nu16183149] [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: 07/25/2024] [Revised: 08/31/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Small intestinal bacterial overgrowth (SIBO) is a common, yet underdiagnosed, gut condition caused by gut dysbiosis. A previous study has shown the potential of herbal therapy, providing equivalent results to rifaximin. OBJECTIVES The objective of this study was to assess how the use of an oral botanical regimen may modulate the gut microbiome, facial erythema, and intestinal permeability in those with SIBO. METHODS This was an open-label prospective study of adults that had lactulose breath test-confirmed SIBO. Participants received a 10-week oral supplementation of a Biocidin liquid tincture and GI Detox+. If participants were found to be non-responsive to treatment after 10 weeks with a persistently positive lactulose breath test, a third oral supplement, Olivirex, was administered for an additional 4 weeks. Lactulose breath tests were administered at baseline, weeks 6, 10, and 14 to assess for SIBO status. A high-resolution photographic analysis system was utilized to analyze changes in facial erythema. Stool sample collections and venipuncture were performed to analyze the gut microbiome and intestinal permeability. RESULTS A total of 33 subjects were screened with breath testing, and 19 subjects were found to have SIBO. Three of the subjects withdrew during the screening period prior to baseline, and sixteen subjects enrolled. Four subjects dropped out after baseline. Hydrogen-dominant SIBO was the most common subtype of SIBO, followed by methane and hydrogen sulfide. The botanical regimen was most effective for hydrogen- and hydrogen sulfide-dominant SIBO, leading to negative breath test results at week 10 in 42.8% and 66.7% of participants, respectively. Compared to baseline, supplementation with the botanical regimen led to positive shifts in short-chain fatty acid-producing bacteria such as A. muciniphila, F. prausnitzii, C. eutectus, and R. faecis by 31.4%, 35.4%, 24.8%, and 48.7% percent at week 10, respectively. The mean abundance of Firmicutes decreased by 20.2%, Bacteroides increased by 30%, and the F/B ratio decreased by 25.4% at week 10 compared to baseline. At week 10, there was a trending 116% increase in plasma LPS/IgG (p = 0.08). There were no significant changes in plasma zonulin, DAO, histamine, DAO/histamine, LPS/IgG, LPS/IgA, or LPS/IgM. Facial erythema was not statistically different at week 6, but at week 10, there was a 20% decrease (p = 0.001) in redness intensity. Among the patients that extended to week 14, there was no statistical change in erythema. CONCLUSIONS Supplementation with an antimicrobial botanical supplemental regimen may have therapeutic potential in hydrogen and hydrogen-sulfide subtypes of SIBO. Furthermore, the botanical supplemental regimen may reduce facial erythema, increase SCFA-producing bacteria, decrease the F/B ratio, and modulate markers of intestinal permeability.
Collapse
Affiliation(s)
- Mildred Min
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Dawnica Nadora
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | | | - Nasima Afzal
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
| | - Chaitra Subramanyam
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Osteopathic Medicine, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Nimrit Gahoonia
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Osteopathic Medicine, Touro University, Vallejo, CA 94592, USA
| | - Adrianne Pan
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
| | - Shivani Thacker
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
| | - Yvonne Nong
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Cindy J. Chambers
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- Pacific Skin Institute, Sacramento, CA 95815, USA
| | - Raja K. Sivamani
- Integrative Skin Science and Research, Sacramento, CA 95815, USA
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
- Pacific Skin Institute, Sacramento, CA 95815, USA
- Department of Dermatology, University of California-Davis, Sacramento, CA 95816, USA
| |
Collapse
|
6
|
Lutsiv T, Hussan H, Thompson HJ. Ecosystemic Approach to Understanding Gut Microbiome-Mediated Prevention of Colorectal Cancer. Cancer J 2024; 30:329-344. [PMID: 39312453 DOI: 10.1097/ppo.0000000000000743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Humans and their associated microorganisms coexist in complex symbiotic relationships. Continuously advancing research is demonstrating the crucial role of host-associated microbiota in the pathophysiology and etiology of disease and in mediating the prevention thereof. As an exemplar, the gut microbiota, especially colonic bacteria, have been extensively studied in colorectal cancer (CRC), and the growing body of evidence establishes new oncomicrobes and their oncometabolites associated with the initiation and promotion of carcinogenesis. Herein, we discuss the importance of approaching the gut microbiome as an ecosystem rather than an assortment of individual factors, especially in the context of cancer prevention. Furthermore, we argue that a dietary pattern effectively drives multiple nodes of the gut microbial ecosystem toward disease- or health-promoting qualities. In the modern circumstances of excessive consumption of ultraprocessed and animal-based foods and concomitant escalation of chronic disease burden worldwide, we focus on whole food-derived dietary fiber as a key to establishing a health-promoting eubiosis in the gut.
Collapse
|
7
|
Lee H, Song J, Lee B, Cha J, Lee H. Food carbohydrates in the gut: structural diversity, microbial utilization, and analytical strategies. Food Sci Biotechnol 2024; 33:2123-2140. [PMID: 39130670 PMCID: PMC11315866 DOI: 10.1007/s10068-024-01648-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 08/13/2024] Open
Abstract
Carbohydrates, which are a vital dietary component, undergo digestion and gut fermentation through microbial enzymes to produce beneficial short-chain fatty acids. Certain carbohydrates selectively modulate the gut microbiota, impacting host health. Carbohydrate-active enzymes within the gut microbiota significantly contribute to carbohydrate utilization and microbial diversity. Despite their importance, the structural complexity of carbohydrates poses analytical challenges. However, recent advancements, notably, mass spectrometry, have allowed for their characterization and functional analysis. This review examines the intricate relationship between dietary carbohydrates and the gut microbiota, highlighting the crucial role of advanced analytical techniques in understanding their diversity and implications. These advancements provide valuable insights into carbohydrate bioactivity. Integrating high-throughput analysis with next-generation sequencing provides deeper insights into gut microbial interactions, potentially revealing which carbohydrate structures are beneficial for gut health.
Collapse
Affiliation(s)
- HyunJi Lee
- Department of Applied Chemistry, Food Science and Technology, Dong-eui University, Busan, 47340 Republic of Korea
| | - JaeHui Song
- Department of Applied Chemistry, Food Science and Technology, Dong-eui University, Busan, 47340 Republic of Korea
| | - Bokyung Lee
- Department of Food Science and Nutrition, Dong-A University, Busan, 49315 Republic of Korea
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, 49315 Republic of Korea
| | - Jaeho Cha
- Department of Microbiology, Pusan National University, Busan, 46241 Republic of Korea
- Microbiological Resources Research Institute, Pusan National University, Busan, 46241 Republic of Korea
| | - Hyeyoung Lee
- Department of Applied Chemistry, Food Science and Technology, Dong-eui University, Busan, 47340 Republic of Korea
| |
Collapse
|
8
|
Doan TNM, Maruyama D, Tian X, Prakash A. Sequential Blood Collection from Inferior Vena Cava Followed by Portal Vein to Evaluate Gut Microbial Metabolites in Mice. J Vis Exp 2024:10.3791/66673. [PMID: 38975757 PMCID: PMC11563659 DOI: 10.3791/66673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Abstract
Gut microbial products are known to act both locally within the intestine and get absorbed into circulation, where their effects can extend to numerous distant organ systems. Short-chain fatty acids (SCFA) are one class of metabolites produced by gut microbes during the fermentation of indigestible dietary fiber. They are now recognized as important contributors to how the gut microbiome influences extra-intestinal organ systems via the gut-lung, gut-brain, and other gut-organ axes throughout the host. SCFAs are absorbed from the colon, through intestinal tissue, into the portal vein (PV). They then pass through the liver, and are consumed in various organs such as the brain, muscle, adipose tissue, and lungs. SCFAs are most easily measured in the expelled fecal material however, more accurate measurements have been obtained from intra-colonic fecal contents. Here we propose that sampling PV and systemic circulating plasma of a single subject may be preferable for studying the absorption, transport, and systemic levels of SCFAs in mice. We present a new technique for efficient blood sampling from the PV and inferior vena cava (IVC) that allows for the collection of relatively large volumes of blood from the portal and systemic circulations. This is accomplished by ligating the PV, thereby allowing for the dilation or enlargement of the PV as it backfills from the mesenteric veins that drain into it. Using this method, we were able to improve the rate of successful collection as well as the total amount of blood collected (up to 0.3 mL from IVC and 0.5 mL from PV).
Collapse
Affiliation(s)
- Thien N M Doan
- Department of Anesthesia and Perioperative Care, University of California San Francisco and San Francisco General Hospital
| | - Daisuke Maruyama
- Department of Anesthesia and Perioperative Care, University of California San Francisco and San Francisco General Hospital;
| | - Xiaoli Tian
- Department of Anesthesia and Perioperative Care, University of California San Francisco and San Francisco General Hospital
| | - Arun Prakash
- Department of Anesthesia and Perioperative Care, University of California San Francisco and San Francisco General Hospital;
| |
Collapse
|
9
|
Corrêa PS, Fernandes MA, Jimenez CR, Mendes LW, Lima PDMT, Abdalla AL, Louvandini H. Interaction between methanotrophy and gastrointestinal nematodes infection on the rumen microbiome of lambs. FEMS Microbiol Ecol 2024; 100:fiae083. [PMID: 38821514 PMCID: PMC11165275 DOI: 10.1093/femsec/fiae083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/21/2024] [Accepted: 05/29/2024] [Indexed: 06/02/2024] Open
Abstract
Complex cross-talk occurs between gastrointestinal nematodes and gut symbiotic microbiota, with consequences for animal metabolism. To investigate the connection between methane production and endoparasites, this study evaluated the effect of mixed infection with Haemonchus contortus and Trichostrongylus colubriformis on methanogenic and methanotrophic community in rumen microbiota of lambs using shotgun metagenomic and real-time quantitative PCR (qPCR). The rumen content was collected from six Santa Inês lambs, (7 months old) before and after 42 days infection by esophageal tube. The metagenomic analysis showed that the infection affected the microbial community structure leading to decreased abundance of methanotrophs bacteria, i.e. α-proteobacteria and β-proteobacteria, anaerobic methanotrophic archaea (ANME), protozoa, sulfate-reducing bacteria, syntrophic bacteria with methanogens, geobacter, and genes related to pyruvate, fatty acid, nitrogen, and sulfur metabolisms, ribulose monophosphate cycle, and Entner-Doudoroff Pathway. Additionally, the abundance of methanogenic archaea and the mcrA gene did not change. The co-occurrence networks enabled us to identify the interactions between each taxon in microbial communities and to determine the reshaping of rumen microbiome associations by gastrointestinal nematode infection. Besides, the correlation between ANMEs was lower in the animal's postinfection. Our findings suggest that gastrointestinal parasites potentially lead to decreased methanotrophic metabolism-related microorganisms and genes.
Collapse
Affiliation(s)
- Patricia Spoto Corrêa
- Laboratory of Animal Nutrition, Center for Nuclear Energy in Agriculture, University of São Paulo, 303 Centenario Avenue, Piracicaba, SP 13416-000, Brazil
| | - Murilo Antonio Fernandes
- Laboratory of Animal Nutrition, Center for Nuclear Energy in Agriculture, University of São Paulo, 303 Centenario Avenue, Piracicaba, SP 13416-000, Brazil
| | - Carolina Rodriguez Jimenez
- Laboratory of Animal Nutrition, Center for Nuclear Energy in Agriculture, University of São Paulo, 303 Centenario Avenue, Piracicaba, SP 13416-000, Brazil
| | - Lucas William Mendes
- Laboratory of Molecular Cell Biology, Center for Nuclear Energy in Agriculture, University of São Paulo, 303 Centenario Avenue, Piracicaba, SP 13416-000, Brazil
| | - Paulo de Mello Tavares Lima
- Department of Animal Science, University of Wyoming, 1000 East University Avenue, Laramie, WY 82071, United States
| | - Adibe Luiz Abdalla
- Laboratory of Animal Nutrition, Center for Nuclear Energy in Agriculture, University of São Paulo, 303 Centenario Avenue, Piracicaba, SP 13416-000, Brazil
| | - Helder Louvandini
- Laboratory of Animal Nutrition, Center for Nuclear Energy in Agriculture, University of São Paulo, 303 Centenario Avenue, Piracicaba, SP 13416-000, Brazil
| |
Collapse
|
10
|
Cardoneanu A, Rezus II, Burlui AM, Richter P, Bratoiu I, Mihai IR, Macovei LA, Rezus E. Autoimmunity and Autoinflammation: Relapsing Polychondritis and VEXAS Syndrome Challenge. Int J Mol Sci 2024; 25:2261. [PMID: 38396936 PMCID: PMC10889424 DOI: 10.3390/ijms25042261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Relapsing polychondritis is a chronic autoimmune inflammatory condition characterized by recurrent episodes of inflammation at the level of cartilaginous structures and tissues rich in proteoglycans. The pathogenesis of the disease is complex and still incompletely elucidated. The data support the important role of a particular genetic predisposition, with HLA-DR4 being considered an allele that confers a major risk of disease occurrence. Environmental factors, mechanical, chemical or infectious, act as triggers in the development of clinical manifestations, causing the degradation of proteins and the release of cryptic cartilage antigens. Both humoral and cellular immunity play essential roles in the occurrence and perpetuation of autoimmunity and inflammation. Autoantibodies anti-type II, IX and XI collagens, anti-matrilin-1 and anti-COMPs (cartilage oligomeric matrix proteins) have been highlighted in increased titers, being correlated with disease activity and considered prognostic factors. Innate immunity cells, neutrophils, monocytes, macrophages, natural killer lymphocytes and eosinophils have been found in the perichondrium and cartilage, together with activated antigen-presenting cells, C3 deposits and immunoglobulins. Also, T cells play a decisive role in the pathogenesis of the disease, with relapsing polychondritis being considered a TH1-mediated condition. Thus, increased secretions of interferon γ, interleukin (IL)-12 and IL-2 have been highlighted. The "inflammatory storm" formed by a complex network of pro-inflammatory cytokines and chemokines actively modulates the recruitment and infiltration of various cells, with cartilage being a source of antigens. Along with RP, VEXAS syndrome, another systemic autoimmune disease with genetic determinism, has an etiopathogenesis that is still incompletely known, and it involves the activation of the innate immune system through different pathways and the appearance of the cytokine storm. The clinical manifestations of VEXAS syndrome include an inflammatory phenotype often similar to that of RP, which raises diagnostic problems. The management of RP and VEXAS syndrome includes common immunosuppressive therapies whose main goal is to control systemic inflammatory manifestations. The objective of this paper is to detail the main etiopathogenetic mechanisms of a rare disease, summarizing the latest data and presenting the distinct features of these mechanisms.
Collapse
Affiliation(s)
- Anca Cardoneanu
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Irina Rezus
- Discipline of Radiology, Surgery Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania;
| | - Alexandra Maria Burlui
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Patricia Richter
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Bratoiu
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Ioana Ruxandra Mihai
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Luana Andreea Macovei
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| | - Elena Rezus
- Discipline of Rheumatology, Medical Department II, University of Medicine and Pharmacy “Grigore T Popa”, 700115 Iasi, Romania; (A.C.); (A.M.B.); (P.R.); (I.B.); (I.R.M.); (L.A.M.)
- Clinical Rehabilitation Hospital, 700661 Iasi, Romania
| |
Collapse
|
11
|
Chang YH, Yanckello LM, Chlipala GE, Green SJ, Aware C, Runge A, Xing X, Chen A, Wenger K, Flemister A, Wan C, Lin AL. Prebiotic inulin enhances gut microbial metabolism and anti-inflammation in apolipoprotein E4 mice with sex-specific implications. Sci Rep 2023; 13:15116. [PMID: 37704738 PMCID: PMC10499887 DOI: 10.1038/s41598-023-42381-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/09/2023] [Indexed: 09/15/2023] Open
Abstract
Gut dysbiosis has been identified as a crucial factor of Alzheimer's disease (AD) development for apolipoprotein E4 (APOE4) carriers. Inulin has shown the potential to mitigate dysbiosis. However, it remains unclear whether the dietary response varies depending on sex. In the study, we fed 4-month-old APOE4 mice with inulin for 16 weeks and performed shotgun metagenomic sequencing to determine changes in microbiome diversity, taxonomy, and functional gene pathways. We also formed the same experiments with APOE3 mice to identify whether there are APOE-genotype dependent responses to inulin. We found that APOE4 female mice fed with inulin had restored alpha diversity, significantly reduced Escherichia coli and inflammation-associated pathway responses. However, compared with APOE4 male mice, they had less metabolic responses, including the levels of short-chain fatty acids-producing bacteria and the associated kinases, especially those related to acetate and Erysipelotrichaceae. These diet- and sex- effects were less pronounced in the APOE3 mice, indicating that different APOE variants also play a significant role. The findings provide insights into the higher susceptibility of APOE4 females to AD, potentially due to inefficient energy production, and imply the importance of considering precision nutrition for mitigating dysbiosis and AD risk in the future.
Collapse
Affiliation(s)
- Ya-Hsuan Chang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, 65212, USA
| | - Lucille M Yanckello
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - George E Chlipala
- Research Informatics Core, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Stefan J Green
- Genomics and Microbiome Core Facility, Rush University, Chicago, IL, 60612, USA
| | - Chetan Aware
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, 65212, USA
| | - Amelia Runge
- Department of Biological Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Xin Xing
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, 65212, USA
- Department of Computer Science, University of Kentucky, Lexington, KY, 40506, USA
| | - Anna Chen
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Kathryn Wenger
- Department of Biochemistry, University of Missouri, Columbia, MO, 65211, USA
| | - Abeoseh Flemister
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA
- NextGen Precision Health, University of Missouri, Columbia, MO, 65212, USA
| | - Caixia Wan
- Department of Biological and Biomedical Engineering, University of Missouri, Columbia, MO, 65211, USA
| | - Ai-Ling Lin
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, 40536, USA.
- Sanders Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.
- Department of Radiology, University of Missouri, Columbia, MO, 65212, USA.
- NextGen Precision Health, University of Missouri, Columbia, MO, 65212, USA.
- Department of Biological Sciences, University of Missouri, Columbia, MO, 65211, USA.
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO, 65211, USA.
| |
Collapse
|
12
|
Liang X, Li Y, Cheng L, Wu Y, Wu T, Wen J, Huang D, Liao Z, Tan C, Luo Y, Liu Y. Gut microbiota dysbiosis characterized by abnormal elevation of Lactobacillus in patients with immune-mediated necrotizing myopathy. Front Cell Infect Microbiol 2023; 13:1243512. [PMID: 37692165 PMCID: PMC10486907 DOI: 10.3389/fcimb.2023.1243512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Aim The gut microbiota plays an important role in human health. In this study, we aimed to investigate whether and how gut microbiota communities are altered in patients with immune-mediated necrotizing myopathy (IMNM) and provide new ideas to further explore the pathogenesis of IMNM or screen for its clinical therapeutic targets in the future. Methods The gut microbiota collected from 19 IMNM patients and 23 healthy controls (HCs) were examined by using 16S rRNA gene sequencing. Alpha and beta-diversity analyses were applied to examine the bacterial diversity and community structure. Welch's t test was performed to identify the significantly abundant taxa of bacteria between the two groups. Spearman correlation analysis was performed to analyze the correlation between gut microbiota and clinical indicators. A receiver operator characteristic (ROC) curve was used to reflect the sensitivity and specificity of microbial biomarker prediction of IMNM disease. P < 0.05 was considered statistically significant. Results Nineteen IMNM patients and 23 HCs were included in the analysis. Among IMNM patients, 94.74% (18/19) of them used glucocorticoids, while 57.89% (11/19) of them used disease-modifying antirheumatic drugs (DMARDs), and the disease was accessed by MITAX (18.26 ± 8.62) and MYOACT (20.68 ± 8.65) scores. Participants in the groups were matched for gender and age. The diversity of the gut microbiota of IMNM patients differed and decreased compared to that of HCs (Chao1, Shannon, and Simpson indexes: p < 0.05). In IMNM patients, the relative abundances of Bacteroides, Roseburia, and Coprococcus were decreased, while that of Lactobacillus and Streptococcus were relatively increased. Furthermore, in IMNM patients, Lactobacillus was positively correlated with the levels of anti-signal recognition particle (SRP) antibodies, anti-Ro52 antibodies, and erythrocyte sedimentation rate (ESR), while Streptococcus was positively correlated with anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) antibodies and C-reactive protein (CRP). Roseburia was negatively correlated with myoglobin (MYO), cardiac troponin T (cTnT), ESR, CRP, and the occurrence of interstitial lung disease (ILD). Bacteroides was negatively correlated with ESR and CRP, and Coprococcus was negatively correlated with ESR. Finally, the prediction model was built using the top five differential genera, which was verified using a ROC curve (area under the curve (AUC): 87%, 95% confidence interval: 73%-100%). Conclusion We observed a characteristic compositional change in the gut microbiota with an abnormal elevation of Lactobacillus in IMNM patients, which was accompanied by changes in clinical indicators. This suggests that gut microbiota dysbiosis occurs in IMNM patients and is correlated with systemic autoimmune features.
Collapse
Affiliation(s)
- Xiuping Liang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Yanhong Li
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Lu Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Yinlan Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Tong Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Ji Wen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Deying Huang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Zehui Liao
- Department of Rheumatolopy and Immunolopy, Meishan People’s Hospital, Meishan, Sichuan, China
| | - Chunyu Tan
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Yubin Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
- Rare Diseases Center, West China Hospital, Sichuan University, Chengdu, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Chengdu, China
| |
Collapse
|
13
|
Mendoza-León MJ, Mangalam AK, Regaldiz A, González-Madrid E, Rangel-Ramírez MA, Álvarez-Mardonez O, Vallejos OP, Méndez C, Bueno SM, Melo-González F, Duarte Y, Opazo MC, Kalergis AM, Riedel CA. Gut microbiota short-chain fatty acids and their impact on the host thyroid function and diseases. Front Endocrinol (Lausanne) 2023; 14:1192216. [PMID: 37455925 PMCID: PMC10349397 DOI: 10.3389/fendo.2023.1192216] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/23/2023] [Indexed: 07/18/2023] Open
Abstract
Thyroid disorders are clinically characterized by alterations of L-3,5,3',5'-tetraiodothyronine (T4), L-3,5,3'-triiodothyronine (T3), and/or thyroid-stimulating hormone (TSH) levels in the blood. The most frequent thyroid disorders are hypothyroidism, hyperthyroidism, and hypothyroxinemia. These conditions affect cell differentiation, function, and metabolism. It has been reported that 40% of the world's population suffers from some type of thyroid disorder and that several factors increase susceptibility to these diseases. Among them are iodine intake, environmental contamination, smoking, certain drugs, and genetic factors. Recently, the intestinal microbiota, composed of more than trillions of microbes, has emerged as a critical player in human health, and dysbiosis has been linked to thyroid diseases. The intestinal microbiota can affect host physiology by producing metabolites derived from dietary fiber, such as short-chain fatty acids (SCFAs). SCFAs have local actions in the intestine and can affect the central nervous system and immune system. Modulation of SCFAs-producing bacteria has also been connected to metabolic diseases, such as obesity and diabetes. In this review, we discuss how alterations in the production of SCFAs due to dysbiosis in patients could be related to thyroid disorders. The studies reviewed here may be of significant interest to endocrinology researchers and medical practitioners.
Collapse
Affiliation(s)
- María José Mendoza-León
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | | | - Alejandro Regaldiz
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Enrique González-Madrid
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Ma. Andreina Rangel-Ramírez
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Oscar Álvarez-Mardonez
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Omar P. Vallejos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Constanza Méndez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-González
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Yorley Duarte
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Ma. Cecilia Opazo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| |
Collapse
|
14
|
Shimizu J, Murayama MA, Mizukami Y, Arimitsu N, Takai K, Miyabe Y. Innate immune responses in Behçet disease and relapsing polychondritis. Front Med (Lausanne) 2023; 10:1055753. [PMID: 37435539 PMCID: PMC10331610 DOI: 10.3389/fmed.2023.1055753] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Behçet disease (BD) and relapsing polychondritis (RP) are chronic multisystem disorders characterized by recurrent flare-ups of tissue inflammation. Major clinical manifestations of BD are oral aphthae, genital aphthous ulcers, skin lesions, arthritis, and uveitis. Patients with BD may develop rare but serious neural, intestinal, and vascular complications, with high relapse rates. Meanwhile, RP is characterized by the inflammation of the cartilaginous tissues of the ears, nose, peripheral joints, and tracheobronchial tree. Additionally, it affects the proteoglycan-rich structures in the eyes, inner ear, heart, blood vessels, and kidneys. The mouth and genital ulcers with inflamed cartilage (MAGIC) syndrome is a common characteristic of BD and RP. The immunopathology of these two diseases may be closely related. It is established that the genetic predisposition to BD is related to the human leukocyte antigen (HLA)-B51 gene. Skin histopathology demonstrates the overactivation of innate immunity, such as neutrophilic dermatitis/panniculitis, in patients with BD. Monocytes and neutrophils frequently infiltrate cartilaginous tissues of patients with RP. Somatic mutations in UBA1, which encodes a ubiquitylation-related enzyme, cause vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic syndrome (VEXAS) with severe systemic inflammation and activation of myeloid cells. VEXAS prompts auricular and/or nasal chondritis, with neutrophilic infiltration around the cartilage in 52-60% of patients. Thus, innate immune cells may play an important role in the initiation of inflammatory processes underlying both diseases. This review summarizes the recent advances in our understanding of the innate cell-mediated immunopathology of BD and RP, with a focus on the common and distinct features of these mechanisms.
Collapse
Affiliation(s)
- Jun Shimizu
- Department of Immunology and Parasitology, St. Marianna University of School of Medicine, Kawasaki, Kanagawa, Japan
| | - Masanori A. Murayama
- Department of Animal Models for Human Diseases, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| | - Yoshihisa Mizukami
- Department of Immunology and Parasitology, St. Marianna University of School of Medicine, Kawasaki, Kanagawa, Japan
| | - Nagisa Arimitsu
- Department of Immunology and Parasitology, St. Marianna University of School of Medicine, Kawasaki, Kanagawa, Japan
| | - Kenji Takai
- Department of Immunology and Parasitology, St. Marianna University of School of Medicine, Kawasaki, Kanagawa, Japan
| | - Yoshishige Miyabe
- Department of Immunology and Parasitology, St. Marianna University of School of Medicine, Kawasaki, Kanagawa, Japan
| |
Collapse
|
15
|
Woo JS, Hwang SH, Yang S, Lee KH, Lee YS, Choi JW, Park JS, Jhun J, Park SH, Cho ML. Lactobacillus acidophilus and propionate attenuate Sjögren's syndrome by modulating the STIM1-STING signaling pathway. Cell Commun Signal 2023; 21:135. [PMID: 37316856 DOI: 10.1186/s12964-023-01141-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/22/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Sjögren's syndrome (SS) is an autoimmune disease characterized by inflammation of the exocrine gland. An imbalance of gut microbiota has been linked to SS. However, the molecular mechanism is unclear. We investigated the effects of Lactobacillus acidophilus (L. acidophilus) and propionate on the development and progression of SS in mouse model. METHODS We compared the gut microbiomes of young and old mice. We administered L. acidophilus and propionate up to 24 weeks. The saliva flow rate and the histopathology of the salivary glands were investigated, and the effects of propionate on the STIM1-STING signaling pathway were evaluated in vitro. RESULTS Lactobacillaceae and Lactobacillus were decreased in aged mice. SS symptoms were ameliorated by L. acidophilus. The abundance of propionate-producing bacterial was increased by L. acidophilus. Propionate ameliorated the development and progression of SS by inhibiting the STIM1-STING signaling pathway. CONCLUSIONS The findings suggest that Lactobacillus acidophilus and propionate have therapeutic potential for SS. Video Abstract.
Collapse
Affiliation(s)
- Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sun-Hee Hwang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - SeungCheon Yang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Yeon Su Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jeong Won Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jin-Sil Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
16
|
Lyu Z, Hu Y, Guo Y, Liu D. Modulation of bone remodeling by the gut microbiota: a new therapy for osteoporosis. Bone Res 2023; 11:31. [PMID: 37296111 PMCID: PMC10256815 DOI: 10.1038/s41413-023-00264-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 04/01/2023] [Accepted: 04/18/2023] [Indexed: 06/12/2023] Open
Abstract
The gut microbiota (GM) plays a crucial role in maintaining the overall health and well-being of the host. Recent studies have demonstrated that the GM may significantly influence bone metabolism and degenerative skeletal diseases, such as osteoporosis (OP). Interventions targeting GM modification, including probiotics or antibiotics, have been found to affect bone remodeling. This review provides a comprehensive summary of recent research on the role of GM in regulating bone remodeling and seeks to elucidate the regulatory mechanism from various perspectives, such as the interaction with the immune system, interplay with estrogen or parathyroid hormone (PTH), the impact of GM metabolites, and the effect of extracellular vesicles (EVs). Moreover, this review explores the potential of probiotics as a therapeutic approach for OP. The insights presented may contribute to the development of innovative GM-targeted therapies for OP.
Collapse
Affiliation(s)
- Zhengtian Lyu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yongfei Hu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dan Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
17
|
Liu Y, Li X, Cheng L, Zhan H, Huang Y, Li H, Li Y. Progress and challenges in the use of blood biomarkers in relapsing polychondritis. Clin Exp Immunol 2023; 212:199-211. [PMID: 36751132 PMCID: PMC10243844 DOI: 10.1093/cei/uxad014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/15/2022] [Accepted: 02/07/2023] [Indexed: 02/09/2023] Open
Abstract
Relapsing polychondritis (RP) is a rare inflammatory disease with significant individual heterogeneity that involves systemic organs. The diagnosis of RP mainly depends on the clinical manifestations; currently, there are no molecular biomarkers routinely evaluated in clinical practice. Biomarkers have diagnostic or monitoring values and can predict response to treatment or the disease course. Over the years, many biomarkers have been proposed to facilitate diagnosis and prognosis. Unfortunately, ideal biomarkers to diagnose RP have not yet been discovered. Most of the molecular biomarkers in RP are immunological biomarkers, with autoantibodies and proteins related to cartilage damage in the blood being the most common. Alterations in some genes (HLA typing and UBA1 somatic mutation) were detected in patients with RP, which could serve as a potential biomarker for the diagnosis of RP. Moreover, proinflammatory cytokines and lymphocyte levels, and certain laboratory tests, have certain values of RP diagnosis and disease activity assessment but lack specificity and sensitivity. This review describes the different types of biomarkers and their clinical correlation with respect to the diagnosis of RP and disease activity. Research on biomarkers and disease pathology is ongoing to identify the ideal biomarkers that are sensitive and specific for RP.
Collapse
Affiliation(s)
- Yongmei Liu
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiaomeng Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linlin Cheng
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haoting Zhan
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yuan Huang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Haolong Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
18
|
Laragione T, Harris C, Azizgolshani N, Beeton C, Bongers G, Gulko PS. Magnesium increases numbers of Foxp3+ Treg cells and reduces arthritis severity and joint damage in an IL-10-dependent manner mediated by the intestinal microbiome. EBioMedicine 2023; 92:104603. [PMID: 37201335 PMCID: PMC10203746 DOI: 10.1016/j.ebiom.2023.104603] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/10/2023] [Accepted: 04/19/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common autoimmune disease with emerging environmental and microbiome risk factors. The western diet is typically deficient in magnesium (Mg), and there is some evidence suggesting that Mg may have anti-inflammatory properties. But the actual role of Mg supplementation in arthritis or in T cell subsets has not been explored. METHODS We investigated the role of a high Mg diet in two different mouse models of RA induced with the KRN serum, and collagen-induced arthritis. We also characterized the phenotypes of splenocytes, gene expression, and an extensive intestinal microbiome analyses including fecal material transplantation (FMT). FINDINGS The high Mg diet group was significantly protected with reduced arthritis severity and joint damage, and reduced expression of IL-1β, IL-6, and TNFα. The high Mg group also had increased numbers of Foxp3+ Treg cells and IL-10-producing T cells. The high Mg protective effect disappeared in IL-10 knockout mice. FMT from the high Mg diet mice recreated the phenotypes seen in the diet-treated mice, with reduced arthritis severity, increased Foxp3+ Treg, and increased IL-10-producing T cells. Intestinal microbiome analyses using 16S rDNA sequencing revealed diet-specific changes, including reduced levels of RA-associated Prevotella in the high Mg group, while increasing levels of Bacteroides and other bacteria associated with increased production of short-chain fatty acids. Metagenomic analyses implicated additional pathways including L-tryptophan biosynthesis and arginine deiminase. INTERPRETATION We describe a new role for Mg in suppressing arthritis, in expanding Foxp3+ T reg cells and in the production of IL-10, and show that these effects are mediated by the intestinal microbiome. Our discoveries suggest a novel strategy for modifying the intestinal microbiome to treat RA and other autoimmune and inflammatory diseases. FUNDING None.
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Carolyn Harris
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Nasim Azizgolshani
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, United States
| | - Gerold Bongers
- Microbiome Translational Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Percio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States.
| |
Collapse
|
19
|
Zhao X, Kong M, Wang Y, Mao Y, Xu H, He W, He Y, Gu J. Nicotinamide mononucleotide improves the Alzheimer's disease by regulating intestinal microbiota. Biochem Biophys Res Commun 2023; 670:27-35. [PMID: 37271037 DOI: 10.1016/j.bbrc.2023.05.075] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 04/29/2023] [Accepted: 05/20/2023] [Indexed: 06/06/2023]
Abstract
Alzheimer's disease (AD) is the most common progressive neurodegenerative disease, and the intestinal flora and its metabolites play an important role in the amelioration of central nervous system (CNS) disorders such as AD through a bidirectional interaction between the gut-brain axis (GBA). Nicotinamide mononucleotide (NMN), one of the precursors for nicotinamide adenine dinucleotide (NAD+) synthesis, reduces the brain features of AD, including neuroinflammation, mitochondrial abnormalities, synaptic dysfunction, and cognitive impairment. However, the impact of NMN on the gut flora of AD is still unknown. In the current study, we investigated the relationship between gut flora and NMN treatment in APP/PS1 transgenic (AD) mice through the 16S ribosomal RNA (rRNA) high-throughput sequencing analysis of mouse feces after being treated with NMN for 16 weeks. The results show that the NMN significantly changed the intestinal microbial community composition in AD mice. The NMN also increased the relative abundance of short-chain fatty acids (SCFAs)-producing bacteria such as Lactobacillus and Bacteroides at the genus level by protecting intestinal health and improving AD. The overall results suggest novel therapeutic strategies for treating AD and highlight the critical role of gut microbiota in AD pathology, and layout the further research.
Collapse
Affiliation(s)
- Xiaodong Zhao
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Mengmeng Kong
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Yilin Wang
- College of Life Science, University of Jilin, Changchun, 130015, China.
| | - Yingxin Mao
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Huilian Xu
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Wenxing He
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| | - Yan He
- Department of Neurology, Shandong Provincial Qianfoshan Hospital, The First Hospital Affiliated with Shandong First Medical University, 250014, China.
| | - Jinsong Gu
- College of Biological Science and Technology, University of Jinan, Jinan, 250022, China.
| |
Collapse
|
20
|
Cesar T, Salgaço MK, Mesa V, Sartoratto A, Sivieri K. Exploring the Association between Citrus Nutraceutical Eriocitrin and Metformin for Improving Pre-Diabetes in a Dynamic Microbiome Model. Pharmaceuticals (Basel) 2023; 16:650. [PMID: 37242433 PMCID: PMC10221435 DOI: 10.3390/ph16050650] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Pre-diabetes is recognized as an altered metabolic state, which precedes type 2 diabetes, and it is associated with great dysfunction of the intestinal microbiota, known as dysbiosis. Natural compounds, capable of reducing blood glucose without side effects and with a beneficial effect on the microbiota, have been studied as substitutes or adjuvants to conventional hypoglycemic agents, such as metformin. In this work, the effect of the nutraceutical Eriomin®, a mixture of citrus flavonoids (eriocitrin, hesperidin, naringin, and didymin), which reduces glycemia and increases glucagon-like peptide-1 (GLP-1) in pre-diabetic patients, was tested in the Simulator of Human Intestinal Microbial Ecosystem (SHIME®), inoculated with pre-diabetic microbiota. After treatment with Eriomin® plus metformin, a significant increase in acetate and butyrate production was observed. Furthermore, sequencing of the 16S rRNA gene of the microorganisms showed that Eriomin® plus metformin stimulated the growth of Bacteroides and Subdoligranulum genera. Bacteroides are the largest fraction of the intestinal microbiota and are potential colonizers of the colon, with some species producing acetic and propionic fatty acids. In addition, Subdoligranulum species are associated with better host glycemic metabolism. In conclusion, Eriomin® associated with metformin improved the composition and metabolism of the intestinal microbiota, suggesting a potential use in pre-diabetes therapy.
Collapse
Affiliation(s)
- Thais Cesar
- Graduate Program in Food, Nutrition and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (T.C.)
| | - Mateus Kawata Salgaço
- Graduate Program in Food, Nutrition and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (T.C.)
| | - Victoria Mesa
- INSERM, UMR-S 1139 (3PHM), Faculty of Pharmacy, Université Paris Cité, F-75006 Paris, France
- Food and Human Nutrition Research Group, School of Nutrition and Dietetics, Universidad de Antioquia (UdeA), Medellín 050010, Antioquia, Colombia
| | | | - Katia Sivieri
- Graduate Program in Food, Nutrition and Food Engineering, Campus Araraquara, São Paulo State University (UNESP), Araraquara 14800-060, SP, Brazil; (T.C.)
| |
Collapse
|
21
|
Zhai Y, Chen L, Zhao Q, Zheng ZH, Chen ZN, Bian H, Yang X, Lu HY, Lin P, Chen X, Chen R, Sun HY, Fan LN, Zhang K, Wang B, Sun XX, Feng Z, Zhu YM, Zhou JS, Chen SR, Zhang T, Chen SY, Chen JJ, Zhang K, Wang Y, Chang Y, Zhang R, Zhang B, Wang LJ, Li XM, He Q, Yang XM, Nan G, Xie RH, Yang L, Yang JH, Zhu P. Cysteine carboxyethylation generates neoantigens to induce HLA-restricted autoimmunity. Science 2023; 379:eabg2482. [PMID: 36927018 DOI: 10.1126/science.abg2482] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Autoimmune diseases such as ankylosing spondylitis (AS) can be driven by emerging neoantigens that disrupt immune tolerance. Here, we developed a workflow to profile posttranslational modifications involved in neoantigen formation. Using mass spectrometry, we identified a panel of cysteine residues differentially modified by carboxyethylation that required 3-hydroxypropionic acid to generate neoantigens in patients with AS. The lysosomal degradation of integrin αIIb [ITGA2B (CD41)] carboxyethylated at Cys96 (ITGA2B-ceC96) generated carboxyethylated peptides that were presented by HLA-DRB1*04 to stimulate CD4+ T cell responses and induce autoantibody production. Immunization of HLA-DR4 transgenic mice with the ITGA2B-ceC96 peptide promoted colitis and vertebral bone erosion. Thus, metabolite-induced cysteine carboxyethylation can give rise to pathogenic neoantigens that lead to autoreactive CD4+ T cell responses and autoantibody production in autoimmune diseases.
Collapse
Affiliation(s)
- Yue Zhai
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Liang Chen
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Qian Zhao
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Zhao-Hui Zheng
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zhi-Nan Chen
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Huijie Bian
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xu Yang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Huan-Yu Lu
- Department of Occupational and Environmental Health and the Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Peng Lin
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xi Chen
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ruo Chen
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Hao-Yang Sun
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Lin-Ni Fan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Kun Zhang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Bin Wang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiu-Xuan Sun
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Zhuan Feng
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Meng Zhu
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jian-Sheng Zhou
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Shi-Rui Chen
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Tao Zhang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Si-Yu Chen
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jun-Jie Chen
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Kui Zhang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yan Wang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yang Chang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Rui Zhang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Bei Zhang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Li-Juan Wang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiao-Min Li
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Qian He
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Xiang-Min Yang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Gang Nan
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Rong-Hua Xie
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Liu Yang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jing-Hua Yang
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450001, China
| | - Ping Zhu
- Department of Clinical Immunology, Xijing Hospital, and Department of Cell Biology of National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
22
|
Liu Y, Zhu J, Wang H, Lu W, LEE YK, Zhao J, Zhang H. Machine learning framework for gut microbiome biomarkers discovery and modulation analysis in large-scale obese population. BMC Genomics 2022; 23:850. [PMID: 36564713 PMCID: PMC9789565 DOI: 10.1186/s12864-022-09087-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The gut microbiome has proven to be an important factor affecting obesity; however, it remains a challenge to identify consistent biomarkers across geographic locations and perform precisely targeted modulation for obese individuals. RESULTS This study proposed a systematic machine learning framework and applied it to 870 human stool metagenomes across five countries to obtain comprehensive regional shared biomarkers and conduct a personalized modulation analysis. In our pipeline, a heterogeneous ensemble feature selection diagram is first developed to determine an optimal subset of biomarkers through the aggregation of multiple techniques. Subsequently, a deep reinforcement learning method was established to alter the targeted composition to the desired healthy target. In this manner, we can realize personalized modulation by counterfactual inference. Consequently, a total of 42 species were identified as regional shared biomarkers, and they showed good performance in distinguishing obese people from the healthy group (area under curve (AUC) =0.85) when demonstrated on validation datasets. In addition, by pooling all counterfactual explanations, we found that Akkermansia muciniphila, Faecalibacterium prausnitzii, Prevotella copri, Bacteroides dorei, Bacteroides eggerthii, Alistipes finegoldii, Alistipes shahii, Eubacterium sp. _CAG_180, and Roseburia hominis may be potential broad-spectrum targets with consistent modulation in the multi-regional obese population. CONCLUSIONS This article shows that based on our proposed machine-learning framework, we can obtain more comprehensive and accurate biomarkers and provide modulation analysis for the obese population. Moreover, our machine-learning framework will also be very useful for other researchers to further obtain biomarkers and perform counterfactual modulation analysis in different diseases.
Collapse
Affiliation(s)
- Yaoliang Liu
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, P. R, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu China
| | - Jinlin Zhu
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, P. R, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu China
| | - Hongchao Wang
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, P. R, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu China
| | - Wenwei Lu
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, P. R, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu China
| | - Yuan Kun LEE
- grid.4280.e0000 0001 2180 6431Department of Microbiology & Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 21 Lower Kent Ridge Rd, Singapore, Singapore ,grid.258151.a0000 0001 0708 1323International Joint Research Laboratory for Pharmabiotics & Antibiotic Resistance, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, P. R, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu China
| | - Hao Zhang
- grid.258151.a0000 0001 0708 1323State Key Laboratory of Food Science and Technology, Jiangnan University, P. R, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323School of Food Science and Technology, Jiangnan University, Wuxi, 214122 Jiangsu China ,grid.258151.a0000 0001 0708 1323National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, 214122 Jiangsu China ,grid.89957.3a0000 0000 9255 8984Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, 214122 Jiangsu China
| |
Collapse
|
23
|
Liu W, Yang G, Liu P, Jiang X, Xin Y. Modulation of adipose tissue metabolism by microbial-derived metabolites. Front Microbiol 2022; 13:1031498. [PMID: 36569060 PMCID: PMC9783635 DOI: 10.3389/fmicb.2022.1031498] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022] Open
Abstract
Obesity and its complications, including type 2 diabetes, cardiovascular disease, and certain cancers, have posed a significant burden on health and healthcare systems over the years due to their high prevalence and incidence. Gut microbial derivatives are necessary for the regulation of energy metabolism and host immunity, as well as for maintaining homeostasis of the intestinal environment. Gut flora metabolites may be a link between gut microbes and diseases, such as obesity, and help understand why alterations in the microbiota can influence the pathophysiology of human disease. This is supported by emerging evidence that microbial-derived metabolites, such as short-chain fatty acids, bile acids, tryptophan, trimethylamine-N-oxide, and lipopolysaccharides, can be beneficial or detrimental to the host by affecting organs outside the gut, including adipose tissue. Adipose tissue is the largest lipid storage organ in the body and an essential endocrine organ that plays an indispensable role in the regulation of lipid storage, metabolism, and energy balance. Adipose tissue metabolism includes adipocyte metabolism (lipogenesis and lipolysis), thermogenesis, and adipose tissue metabolic maladaptation. Adipose tissue dysfunction causes the development of metabolic diseases, such as obesity. Here, we review the current understanding of how these microbial metabolites are produced and discuss both established mechanisms and the most recent effects of microbial products on host adipose tissue metabolism. We aimed to identify novel therapeutic targets or strategies for the prevention and treatment of obesity and its complications.
Collapse
Affiliation(s)
- Wenyun Liu
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Ge Yang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Pinyi Liu
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology & Therapy, The First Hospital of Jilin University, and Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China,NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China,*Correspondence: Xin Jiang,
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China,Ying Xin,
| |
Collapse
|
24
|
Dietary autolysed yeast modulates blood profiles, small intestinal morphology and caecal microbiota of weaning pigs. Animal 2022; 16:100660. [DOI: 10.1016/j.animal.2022.100660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/19/2022] Open
|
25
|
Deng L, Lee JWJ, Tan KSW. Infection with pathogenic Blastocystis ST7 is associated with decreased bacterial diversity and altered gut microbiome profiles in diarrheal patients. Parasit Vectors 2022; 15:312. [PMID: 36064620 PMCID: PMC9446694 DOI: 10.1186/s13071-022-05435-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/15/2022] [Indexed: 11/23/2022] Open
Abstract
Background Blastocystis is a common protistan parasite inhabiting the gastrointestinal tract of humans and animals. While there are increasing reports characterizing the associations between Blastocystis and the gut microbiome in healthy individuals, only a few studies have investigated the relationships between Blastocystis and the gut microbiota in diarrheal patients. Methods The effects of a specific subtype (ST7) of Blastocystis on the composition of gut microbiota in diarrheal patients were investigated using 16S ribosomal RNA (rRNA) gene sequencing and bioinformatic analyses. Results Compared with diarrheal patients without Blastocystis, diarrheal patients infected with Blastocystis ST7 exhibited lower bacterial diversity. Beta diversity analysis revealed significant differences in bacterial community structure between ST7-infected and Blastocystis-free patients. The proportion of Enterobacteriaceae and Escherichia-Shigella were significantly enriched in ST7-infected patients. In contrast, the abundance of Bacteroides and Parabacteroides were more prevalent in Blastocystis-free patients. Conclusions The results of this study revealed, for the first time, that infection with Blastocystis ST7 is associated with lower bacterial diversity and altered microbial structure in diarrheal patients. Our study on clinical diarrheal patients is also the first to reinforce the notion that ST7 is a pathogenic subtype of Blastocystis. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05435-z.
Collapse
Affiliation(s)
- Lei Deng
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore. .,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jonathan W J Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore.,Division of Gastroenterology & Hepatology, National University Hospital, Singapore, 119074, Singapore
| | - Kevin S W Tan
- Laboratory of Molecular and Cellular Parasitology, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Drive 2, Singapore, 117545, Singapore. .,Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
26
|
Kim H, Jeon S, Kim J, Seol D, Jo J, Cho S, Kim H. Investigation of memory-enhancing effects of Streptococcus thermophilus EG007 in mice and elucidating molecular and metagenomic characteristics using nanopore sequencing. Sci Rep 2022; 12:13274. [PMID: 35918353 PMCID: PMC9346115 DOI: 10.1038/s41598-022-14837-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 06/13/2022] [Indexed: 11/15/2022] Open
Abstract
Over the past decades, accumulating evidences have highlighted the gut microbiota as a key player in the brain functioning via microbiota–gut–brain axis, and accordingly, the beneficial role of several probiotic strains in cognitive ability also have been actively investigated. However, the majority of the research have demonstrated the effects against age-related cognitive decline or neurological disease. To this end, we aimed to investigate lactic acid bacteria strains having beneficial effects on the cognitive function of healthy young mice and elucidate underlying characteristics by carrying out nanopore sequencing-based genomics and metagenomics analysis. 8-week consumption of Streptococcus thermophilus EG007 demonstrated marked enhancements in behavior tests assessing short-term spatial and non-spatial learning and memory. It was revealed that EG007 possessed genes encoding various metabolites beneficial for a health condition in many aspects, including gamma-aminobutyric acid producing system, a neurotransmitter associated with mood and stress response. Also, by utilizing 16S–23S rRNA operon as a taxonomic marker, we identified more accurate species-level compositional changes in gut microbiota, which was increase of certain species, previously reported to have associations with mental health or down-regulation of inflammation or infection-related species. Moreover, correlation analysis revealed that the EG007-mediated altered microbiota had a significant correlation with the memory traits.
Collapse
Affiliation(s)
- Hyaekang Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Soomin Jeon
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Jina Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Donghyeok Seol
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.,eGnome, Inc, Seoul, Republic of Korea
| | - JinChul Jo
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Seoae Cho
- eGnome, Inc, Seoul, Republic of Korea
| | - Heebal Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea. .,eGnome, Inc, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Therapeutic potential of Short Chain Fatty acid production by gut microbiota in Neurodegenerative disorders. Nutr Res 2022; 106:72-84. [DOI: 10.1016/j.nutres.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 07/30/2022] [Indexed: 11/20/2022]
|
28
|
Moroishi Y, Gui J, Hoen AG, Morrison HG, Baker ER, Nadeau KC, Li H, Li Z, Madan JC, Karagas MR. The relationship between the gut microbiome and the risk of respiratory infections among newborns. COMMUNICATIONS MEDICINE 2022; 2:87. [PMID: 35847562 PMCID: PMC9283516 DOI: 10.1038/s43856-022-00152-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 06/27/2022] [Indexed: 12/15/2022] Open
Abstract
Background Emerging evidence points to a critical role of the developing gut microbiome in immune maturation and infant health; however, prospective studies are lacking. Methods We examined the occurrence of infections and associated symptoms during the first year of life in relation to the infant gut microbiome at six weeks of age using bacterial 16S rRNA V4-V5 gene sequencing (N = 465) and shotgun metagenomics (N = 185). We used generalized estimating equations to assess the associations between longitudinal outcomes and 16S alpha diversity and metagenomics species. Results Here we show higher infant gut microbiota alpha diversity was associated with an increased risk of infections or respiratory symptoms treated with a prescription medicine, and specifically upper respiratory tract infections. Among vaginally delivered infants, a higher alpha diversity was associated with an increased risk of all-cause wheezing treated with a prescription medicine and diarrhea involving a visit to a health care provider. Positive associations were specifically observed with Veillonella species among all deliveries and Haemophilus influenzae among cesarean-delivered infants. Conclusion Our findings suggest that intestinal microbial diversity and the relative abundance of key taxa in early infancy may influence susceptibility to respiratory infection, wheezing, and diarrhea.
Collapse
Affiliation(s)
- Yuka Moroishi
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| | - Jiang Gui
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| | - Anne G. Hoen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| | - Hilary G. Morrison
- Josephine Bay Paul Center for Comparative Molecular Biology and Evolution, Marine Biological Laboratory, Woods Hole, MA USA
| | - Emily R. Baker
- Department of Obstetrics and Gynecology, Dartmouth-Hitchcock Medical Center, Lebanon, NH USA
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, CA USA
| | - Hongzhe Li
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA USA
| | - Zhigang Li
- Department of Biostatistics, University of Florida, Gainesville, FL USA
| | - Juliette C. Madan
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
- Department of Pediatrics, Children’s Hospital at Dartmouth, Lebanon, NH USA
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH USA
| |
Collapse
|
29
|
O'Riordan KJ, Collins MK, Moloney GM, Knox EG, Aburto MR, Fülling C, Morley SJ, Clarke G, Schellekens H, Cryan JF. Short chain fatty acids: Microbial metabolites for gut-brain axis signalling. Mol Cell Endocrinol 2022; 546:111572. [PMID: 35066114 DOI: 10.1016/j.mce.2022.111572] [Citation(s) in RCA: 220] [Impact Index Per Article: 73.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 02/08/2023]
Abstract
The role of the intestinal microbiota as a regulator of gut-brain axis signalling has risen to prominence in recent years. Understanding the relationship between the gut microbiota, the metabolites it produces, and the brain will be critical for the subsequent development of new therapeutic approaches, including the identification of novel psychobiotics. A key focus in this regard have been the short-chain fatty acids (SCFAs) produced by bacterial fermentation of dietary fibre, which include butyrate, acetate, and propionate. Ongoing research is focused on the entry of SCFAs into systemic circulation from the gut lumen, their migration to cerebral circulation and across the blood brain barrier, and their potential to exert acute and chronic effects on brain structure and function. This review aims to discuss our current mechanistic understanding of the direct and indirect influence that SCFAs have on brain function, behaviour and physiology, which will inform future microbiota-targeted interventions for brain disorders.
Collapse
Affiliation(s)
| | - Michael K Collins
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Gerard M Moloney
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Emily G Knox
- APC Microbiome Ireland, University College Cork, Ireland; School of Pharmacy, University College Cork, Ireland
| | - María R Aburto
- APC Microbiome Ireland, University College Cork, Ireland
| | | | - Shane J Morley
- APC Microbiome Ireland, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland.
| |
Collapse
|
30
|
Shimizu J, Suzuki N. Mechanical model of steady-state and inflammatory conditions in patients with relapsing polychondritis: A review. Medicine (Baltimore) 2022; 101:e28852. [PMID: 35212285 PMCID: PMC8878696 DOI: 10.1097/md.0000000000028852] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/28/2022] [Indexed: 11/26/2022] Open
Abstract
Relapsing polychondritis (RP) is a multisystem inflammatory disorder, considered to associate with immune aberration.Increased T helper type-1 cell-related cytokines were reported in RP patients. mRNA expressions of a regulatory T cell cytokine interleukin (IL)-10 increased, whereas pro-inflammatory cytokines IL1β and IL6 mRNA expressions decreased in freshly isolated peripheral blood mononuclear cells of RP patients compared with those in healthy individuals. Upon in vitro stimulation with mitogen, IL10 mRNA expressions decreased, and IL1β and IL6 mRNA expressions increased in RP patients.This short-time dynamic change of gene expressions from anti-inflammatory to pro-inflammatory features of immune cells may be associated with the "relapsing" disease course of patients with RP. IL1β mRNA expressions of peripheral blood mononuclear cells exhibited positive correlations with serum matrix metalloproteinase (MMP)-3 concentrations in patients with respiratory involvement. Such positive correlation was not found in those without respiratory involvement.In a metagenomic analysis, an altered composition of gut microbes was found, suggesting that microbe metabolites such as short-chain fatty acids may affect T cell responses of the patients.In this review, the relationships among RP-related inflammatory molecules were summarized. The data support a hypothesis that the immune conditions are different between steady-state and inflammation in RP patients.
Collapse
|
31
|
Little M, Dutta M, Li H, Matson A, Shi X, Mascarinas G, Molla B, Weigel K, Gu H, Mani S, Cui JY. Understanding the physiological functions of the host xenobiotic-sensing nuclear receptors PXR and CAR on the gut microbiome using genetically modified mice. Acta Pharm Sin B 2022; 12:801-820. [PMID: 35256948 PMCID: PMC8897037 DOI: 10.1016/j.apsb.2021.07.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/29/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological activation of the xenobiotic-sensing nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) is well-known to increase drug metabolism and reduce inflammation. Little is known regarding their physiological functions on the gut microbiome. In this study, we discovered bivalent hormetic functions of PXR/CAR modulating the richness of the gut microbiome using genetically engineered mice. The absence of PXR or CAR increased microbial richness, and absence of both receptors synergistically increased microbial richness. PXR and CAR deficiency increased the pro-inflammatory bacteria Helicobacteraceae and Helicobacter. Deficiency in both PXR and CAR increased the relative abundance of Lactobacillus, which has bile salt hydrolase activity, corresponding to decreased primary taurine-conjugated bile acids (BAs) in feces, which may lead to higher internal burden of taurine and unconjugated BAs, both of which are linked to inflammation, oxidative stress, and cytotoxicity. The basal effect of PXR/CAR on the gut microbiome was distinct from pharmacological and toxicological activation of these receptors. Common PXR/CAR-targeted bacteria were identified, the majority of which were suppressed by these receptors. hPXR-TG mice had a distinct microbial profile as compared to wild-type mice. This study is the first to unveil the basal functions of PXR and CAR on the gut microbiome.
Collapse
Key Words
- BA, bile acid
- BSH, bile salt hydrolase
- Bile acids
- CA, cholic acid
- CAR
- CAR, constitutive androstane receptor
- CDCA, chenodeoxycholic acid
- CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
- CV, conventional
- CYP, cytochrome P450
- DCA, deoxycholic acid
- EGF, epidermal growth factor
- Feces
- GF, germ free
- GLP-1, glucagon-like peptide-1
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- Gut microbiome
- HDCA, hyodeoxycholic acid
- IBD, inflammatory bowel disease
- IFNγ, interferon-gamma
- IL, interleukin
- IS, internal standards
- Inflammation
- LCA, lithocholic acid
- LC–MS/MS, liquid chromatography–tandem mass spectrometry
- MCA, muricholic acid
- MCP-1, monocyte chemoattractant protein-1
- Mice
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NSAID, non-steroidal anti-inflammatory drug
- Nuclear receptor
- OH, hydroxylated
- OTUs, operational taxonomy units
- PA, indole-3 propionic acid
- PBDEs, polybrominated diphenyl ethers
- PCBs, polychlorinated biphenyls
- PCoA, Principle Coordinate Analysis
- PXR
- PXR, pregnane X receptor
- PiCRUSt, Phylogenetic Investigation of Communities by Reconstruction of Observed States
- QIIME, Quantitative Insights Into Microbial Ecology
- SCFAs, short-chain fatty acids
- SNP, single-nucleotide polymorphism
- SPF, specific-pathogen-free
- T, wild type
- T-, taurine conjugated
- TCPOBOP, 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene, 3,3′,5,5′-Tetrachloro-1,4-bis(pyridyloxy)benzene
- TGR-5, Takeda G-protein-coupled receptor 5
- TLR4, toll-like receptor 4
- TNF, tumor necrosis factor
- UDCA, ursodeoxycholic acid
- YAP, yes-associated protein
- hPXR-TG, humanized PXR transgenic
Collapse
Affiliation(s)
- Mallory Little
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Hao Li
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Adam Matson
- University of Connecticut, Hartford, CT 06106, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Gabby Mascarinas
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Bruk Molla
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Kris Weigel
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Sridhar Mani
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
32
|
Li M, Rajani C, Zheng X, Jia W. The microbial metabolome in metabolic-associated fatty liver disease. J Gastroenterol Hepatol 2022; 37:15-23. [PMID: 34850445 DOI: 10.1111/jgh.15746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/30/2022]
Abstract
Metabolism-associated fatty liver disease (MAFLD) is defined as the presence of excess fat in the liver in the absence of excess alcohol consumption and metabolic dysfunction. It has also been described as the hepatic manifestation of metabolic syndrome. The incidence of MAFLD has been reported to be 43-60% in diabetics, ~90% in patients with hyperlipidemia, and 91% in morbidly obese patients. Risk factors that have been associated with the development of MAFLD include male gender, increasing age, obesity, insulin resistance, diabetes, and hyperlipidemia. All of these risk factors have been linked to alterations of the gut microbiota, that is, gut dysbiosis. MAFLD can progress to non-alcoholic steatohepatitis with the presence of inflammation and ballooning, which can deteriorate into cirrhosis, MAFLD-related hepatocellular carcinoma, and liver failure. In this review, we will be focused on the role of the gut microbial metabolome in the development, progression, and potential treatment of MAFLD.
Collapse
Affiliation(s)
- Mengci Li
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Cynthia Rajani
- University of Hawaii Cancer Center, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Xiaojiao Zheng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| |
Collapse
|
33
|
Mirzaei R, Dehkhodaie E, Bouzari B, Rahimi M, Gholestani A, Hosseini-Fard SR, Keyvani H, Teimoori A, Karampoor S. Dual role of microbiota-derived short-chain fatty acids on host and pathogen. Biomed Pharmacother 2022; 145:112352. [PMID: 34840032 DOI: 10.1016/j.biopha.2021.112352] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
A growing body of documents shows microbiota produce metabolites such as short-chain fatty acids (SCFAs) as crucial executors of diet-based microbial influence the host and bacterial pathogens. The production of SCFAs depends on the metabolic activity of intestinal microflora and is also affected by dietary changes. SCFAs play important roles in maintaining colonic health as an energy source, as a regulator of gene expression and cell differentiation, and as an anti-inflammatory agent. Additionally, the regulated expression of virulence genes is critical for successful infection by an intestinal pathogen. Bacteria rely on sensing environmental signals to find preferable niches and reach the infectious state. This review will present data supporting the diverse functional roles of microbiota-derived butyrate, propionate, and acetate on host cellular activities such as immune modulation, energy metabolism, nervous system, inflammation, cellular differentiation, and anti-tumor effects, among others. On the other hand, we will discuss and summarize data about the role of these SCFAs on the virulence factor of bacterial pathogens. In this regard, receptors and signaling routes for SCFAs metabolites in host and pathogens will be introduced.
Collapse
Affiliation(s)
- Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| | - Elahe Dehkhodaie
- Department of Biology, Science and Research Branch, Islamic Azad University Tehran, Iran
| | - Behnaz Bouzari
- Department of Pathology, Firouzgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mandana Rahimi
- Department of Pathology, School of Medicine, Hasheminejad Kidney Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Gholestani
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Hosseini-Fard
- Department of Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Teimoori
- Department of Virology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Berthelot JM, Lioté F, Sibilia J. Tissue microbiota: a 'secondary-self', first target of autoimmunity? Joint Bone Spine 2021; 89:105337. [PMID: 34968748 DOI: 10.1016/j.jbspin.2021.105337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/07/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Department, Nantes University Hospital, Hôtel-Dieu, Place Alexis-Ricordeau, 44093, Nantes Cedex 01, France.
| | - Frédéric Lioté
- Rheumatology Department & Inserm UMR 1132 (centre Viggo Petersen), Hôpital Lariboisière, 2 rue Ambroise Paré, F-75010 Paris, France; Université de Paris, UFR de Médecine, F-75010 Paris, France
| | - Jean Sibilia
- Service de rhumatologie, Hôpitaux Universitaires de Strasbourg, F-67000 Strasbourg, France; RESO: Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, France; INSERM UMR_S1109, Université de Strasbourg, F-67000 Strasbourg, France
| |
Collapse
|
35
|
Soares E, Soares AC, Trindade PL, Monteiro EB, Martins FF, Forgie AJ, Inada KOP, de Bem GF, Resende A, Perrone D, Souza-Mello V, Tomás-Barberán F, Willing BP, Monteiro M, Daleprane JB. Jaboticaba (Myrciaria jaboticaba) powder consumption improves the metabolic profile and regulates gut microbiome composition in high-fat diet-fed mice. Biomed Pharmacother 2021; 144:112314. [PMID: 34634561 DOI: 10.1016/j.biopha.2021.112314] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
The consumption of a high-fat diet can cause metabolic syndrome and induces host gut microbial dysbiosis and non-alcoholic fatty liver disease (NAFLD). We evaluated the effect of polyphenol-rich jaboticaba peel and seed powder (JPSP) on the gut microbial community composition and liver health in a mouse model of NAFLD. Three-month-old C57BL/6 J male mice, received either a control (C, 10% of lipids as energy, n = 16) or high-fat (HF, 50% of lipids as energy, n = 64) diet for nine weeks. The HF mice were randomly subdivided into four groups (n = 16 in each group), three of which (HF-J5, HF-J10, and HF-J15) were supplemented with dietary JPSP for four weeks (5%, 10%, and 15%, respectively). In addition to attenuating weight gain, JPSP consumption improved dyslipidemia and insulin resistance. In a dose-dependent manner, JPSP consumption ameliorated the expression of hepatic lipogenesis genes (AMPK, SREBP-1, HGMCoA, and ABCG8). The effects on the microbial community structure were determined in all JPSP-supplemented groups; however, the HF-J10 and HF-J15 diets led to a drastic depletion in the species of numerous bacterial families (Bifidobacteriaceae, Mogibacteriaceae, Christensenellaceae, Clostridiaceae, Dehalobacteriaceae, Peptococcaceae, Peptostreptococcaceae, and Ruminococcaceae) compared to the HF diet, some of which represented a reversal of increases associated with HF. The Lachnospiraceae and Enterobacteriaceae families and the Parabacteroides, Sutterella, Allobaculum, and Akkermansia genera were enriched more in the HF-J10 and HF-J15 groups than in the HF group. In conclusion, JPSP consumption improved obesity-related metabolic profiles and had a strong impact on the microbial community structure, thereby reversing NAFLD and decreasing its severity.
Collapse
Affiliation(s)
- Elaine Soares
- Laboratory for studies of Interactions between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Aruanna C Soares
- Laboratory for studies of Interactions between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Patricia Leticia Trindade
- Laboratory for studies of Interactions between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Elisa B Monteiro
- Laboratory for studies of Interactions between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Fabiane F Martins
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Andrew J Forgie
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Kim O P Inada
- Laboratory for studies of Interactions between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Graziele F de Bem
- Laboratory of Cardiovascular Pharmacology and Medicinal Plants, Department of Pharmacology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Angela Resende
- Laboratory of Cardiovascular Pharmacology and Medicinal Plants, Department of Pharmacology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Daniel Perrone
- Laboratório de Bioquímica Nutricional e de Alimentos, Chemistry Institute, Federal University of Rio de Janeiro, Av. Athos da Silveira Ramos 149, CT, Bloco A, sala 528 A, 21941-909 Rio de Janeiro, Brazil
| | - Vanessa Souza-Mello
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Biomedical Center, Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Francisco Tomás-Barberán
- Research Group on Quality, Safety and Bioactivity of Plant Foods, Department of Food Science and Technology, CEBAS-CSIC, P.O. Box 164, 30100 Campus de Espinardo, Murcia, Spain
| | - Benjamin P Willing
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Mariana Monteiro
- Laboratório de Alimentos Funcionais, Instituto de Nutrição Josué de Castro, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julio B Daleprane
- Laboratory for studies of Interactions between Nutrition and Genetics, LEING, Department of Basic and Experimental Nutrition, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
36
|
Modulation of Adipocyte Metabolism by Microbial Short-Chain Fatty Acids. Nutrients 2021; 13:nu13103666. [PMID: 34684670 PMCID: PMC8538331 DOI: 10.3390/nu13103666] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/17/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Obesity and its complications—including type 2 diabetes, cardiovascular disease, and certain cancers—constitute a rising global epidemic that has imposed a substantial burden on health and healthcare systems over the years. It is becoming increasingly clear that there is a link between obesity and the gut microbiota. Gut dysbiosis, characterized as microbial imbalance, has been consistently associated with obesity in both humans and animal models, and can be reversed with weight loss. Emerging evidence has shown that microbial-derived metabolites such as short-chain fatty acids (SCFAs)—including acetate, propionate, and butyrate—provide benefits to the host by impacting organs beyond the gut, including adipose tissue. In this review, we summarize what is currently known regarding the specific mechanisms that link gut-microbial-derived SCFAs with adipose tissue metabolism, such as adipogenesis, lipolysis, and inflammation. In addition, we explore indirect mechanisms by which SCFAs can modulate adipose tissue metabolism, such as via perturbation of gut hormones, as well as signaling to the brain and the liver. Understanding how the modulation of gut microbial metabolites such as SCFAs can impact adipose tissue function could lead to novel therapeutic strategies for the prevention and treatment of obesity.
Collapse
|
37
|
Chen J, Wang W, Guo Z, Huang S, Lei H, Zang P, Lu B, Shao J, Gu P. Associations between gut microbiota and thyroidal function status in Chinese patients with Graves' disease. J Endocrinol Invest 2021; 44:1913-1926. [PMID: 33481211 DOI: 10.1007/s40618-021-01507-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/09/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The imbalance of gut microbiota has been linked to manifold endocrine diseases, but the association with Graves' disease (GD) is still unclear. The purpose of this study was to investigate the correlation between human gut microbiota and clinical characteristics and thyroidal functional status of GD. METHODS 14 healthy volunteers (CG) and 15 patients with primary GD (HG) were recruited as subjects. 16SrDNA high-throughput sequencing was performed on IlluminaMiSeq platform to analyze the characteristics of gut microbiota in patients with GD. Among them, the thyroid function of 13 patients basically recovered after treatment with anti-thyroid drugs (oral administration of Methimazole for 3-5 months). The fecal samples of patients after treatment (TG) were sequenced again, to further explore and investigate the potential relationship between dysbacteriosis and GD. RESULTS In terms of alpha diversity index, the observed OTUs, Simpson and Shannon indices of gut microbiota in patients with GD were significantly lower than those in healthy volunteers (P < 0.05).The difference of bacteria species was mainly reflected in the genus level, in which the relative abundance of Lactobacillus, Veillonella and Streptococcus increased significantly in GD. After the improvement of thyroid function, a significant reduction at the genus level were Blautia, Corynebacter, Ruminococcus and Streptococcus, while Phascolarctobacterium increased significantly (P < 0.05). According to Spearman correlation analysis, the correlation between the level of thyrotropin receptor antibody (TRAb) and the relative abundance of Lactobacillus and Ruminococcus was positive, while Synergistetes and Phascolarctobacterium showed a negative correlation with TRAb. Besides, there were highly significant negative correlation between Synergistetes and clinical variables of TRAb, TPOAb and TGAb (P < 0.05, R < - 0.6). CONCLUSIONS This study revealed that functional status and TRAb level in GD were associated with composition and biological function in the gut microbiota, with Synergistetes and Phascolarctobacterium protecting the thyroid probably, while Ruminococcus and Lactobacillus may be novel biomarkers of GD.
Collapse
Affiliation(s)
- J Chen
- Department of Endocrinology, Jinling Hospital, Southeast Univ, Sch Med, Nanjing, China
| | - W Wang
- Department of Endocrinology, Jinling Hospital, Nanjing Univ, Sch Med, Nanjing, China
| | - Z Guo
- Department of Endocrinology, Jinling Hospital, Nanjing Med Univ, Nanjing, China
| | - S Huang
- Department of Endocrinology, Jinling Hospital, Nanjing Univ, Sch Med, Nanjing, China
| | - H Lei
- Department of Endocrinology, Jinling Hospital, Southern Medical University, Nanjing, China
| | - P Zang
- Department of Endocrinology, Jinling Hospital, Nanjing Univ, Sch Med, Nanjing, China
| | - B Lu
- Department of Endocrinology, Jinling Hospital, Nanjing Univ, Sch Med, Nanjing, China
| | - J Shao
- Department of Endocrinology, Jinling Hospital, Nanjing Univ, Sch Med, Nanjing, China.
| | - P Gu
- Department of Endocrinology, Jinling Hospital, Nanjing Univ, Sch Med, Nanjing, China.
| |
Collapse
|
38
|
Spyrou N, Vallianou N, Kadillari J, Dalamaga M. The interplay of obesity, gut microbiome and diet in the immune check point inhibitors therapy era. Semin Cancer Biol 2021; 73:356-376. [PMID: 33989733 DOI: 10.1016/j.semcancer.2021.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 03/22/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
|
39
|
Leonard MM, Valitutti F, Karathia H, Pujolassos M, Kenyon V, Fanelli B, Troisi J, Subramanian P, Camhi S, Colucci A, Serena G, Cucchiara S, Trovato CM, Malamisura B, Francavilla R, Elli L, Hasan NA, Zomorrodi AR, Colwell R, Fasano A. Microbiome signatures of progression toward celiac disease onset in at-risk children in a longitudinal prospective cohort study. Proc Natl Acad Sci U S A 2021; 118:e2020322118. [PMID: 34253606 PMCID: PMC8307711 DOI: 10.1073/pnas.2020322118] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Other than exposure to gluten and genetic compatibility, the gut microbiome has been suggested to be involved in celiac disease (CD) pathogenesis by mediating interactions between gluten/environmental factors and the host immune system. However, to establish disease progression markers, it is essential to assess alterations in the gut microbiota before disease onset. Here, a prospective metagenomic analysis of the gut microbiota of infants at risk of CD was done to track shifts in the microbiota before CD development. We performed cross-sectional and longitudinal analyses of gut microbiota, functional pathways, and metabolites, starting from 18 mo before CD onset, in 10 infants who developed CD and 10 matched nonaffected infants. Cross-sectional analysis at CD onset identified altered abundance of six microbial strains and several metabolites between cases and controls but no change in microbial species or pathway abundance. Conversely, results of longitudinal analysis revealed several microbial species/strains/pathways/metabolites occurring in increased abundance and detected before CD onset. These had previously been linked to autoimmune and inflammatory conditions (e.g., Dialister invisus, Parabacteroides sp., Lachnospiraceae, tryptophan metabolism, and metabolites serine and threonine). Others occurred in decreased abundance before CD onset and are known to have anti-inflammatory effects (e.g., Streptococcus thermophilus, Faecalibacterium prausnitzii, and Clostridium clostridioforme). Additionally, we uncovered previously unreported microbes/pathways/metabolites (e.g., Porphyromonas sp., high mannose-type N-glycan biosynthesis, and serine) that point to CD-specific biomarkers. Our study establishes a road map for prospective longitudinal study designs to better understand the role of gut microbiota in disease pathogenesis and therapeutic targets to reestablish tolerance and/or prevent autoimmunity.
Collapse
Affiliation(s)
- Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02129
- Celiac Research Program, Harvard Medical School, Boston, MA 02114
| | - Francesco Valitutti
- European Biomedical Research Institute of Salerno, 84125 Salerno, Italy
- Pediatric Unit, Maternal and Child Health Department, Azienda Ospedaliera Universitaria San Giovanni di Dio e Ruggi d'Aragona, 84125 Salerno, Italy
| | | | | | - Victoria Kenyon
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02129
- Celiac Research Program, Harvard Medical School, Boston, MA 02114
| | | | - Jacopo Troisi
- European Biomedical Research Institute of Salerno, 84125 Salerno, Italy
- Theoreo srl, University of Salerno, 20851 Salerno, Italy
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84090 Salerno, Italy
| | | | - Stephanie Camhi
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02129
- Celiac Research Program, Harvard Medical School, Boston, MA 02114
| | - Angelo Colucci
- Theoreo srl, University of Salerno, 20851 Salerno, Italy
- Department of Medicine, Surgery and Dentistry, Scuola Medica Salernitana, University of Salerno, 84090 Salerno, Italy
| | - Gloria Serena
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02129
- Celiac Research Program, Harvard Medical School, Boston, MA 02114
| | | | | | - Basilio Malamisura
- Pediatric Unit, Maternal and Child Health Department, Azienda Ospedaliera Universitaria San Giovanni di Dio e Ruggi d'Aragona, 00185 Salerno, Italy
| | | | - Luca Elli
- Center for Prevention and Diagnosis of Celiac Disease, Fondazione Department and University Hospital (IRCCS) Ca' Granda Ospedale Maggiore Policlinico, 70126 Milan, Italy
| | | | - Ali R Zomorrodi
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02129
- Celiac Research Program, Harvard Medical School, Boston, MA 02114
| | - Rita Colwell
- CosmosID Inc., Rockville, MD 84100;
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD 20122
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02114;
- Mucosal Immunology and Biology Research Center, MassGeneral Hospital for Children, Harvard Medical School, Boston, MA 02129
- Celiac Research Program, Harvard Medical School, Boston, MA 02114
- European Biomedical Research Institute of Salerno, 84125 Salerno, Italy
| |
Collapse
|
40
|
Jang HR, Lee HY. Mechanisms linking gut microbial metabolites to insulin resistance. World J Diabetes 2021; 12:730-744. [PMID: 34168724 PMCID: PMC8192250 DOI: 10.4239/wjd.v12.i6.730] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/23/2021] [Accepted: 05/20/2021] [Indexed: 02/06/2023] Open
Abstract
Insulin resistance is the rate-limiting step in the development of metabolic diseases, including type 2 diabetes. The gut microbiota has been implicated in host energy metabolism and metabolic diseases and is recognized as a quantitatively important organelle in host metabolism, as the human gut harbors 10 trillion bacterial cells. Gut microbiota break down various nutrients and produce metabolites that play fundamental roles in host metabolism and aid in the identification of possible therapeutic targets for metabolic diseases. Therefore, understanding the various effects of bacterial metabolites in the development of insulin resistance is critical. Here, we review the mechanisms linking gut microbial metabolites to insulin resistance in various insulin-responsive tissues.
Collapse
Affiliation(s)
- Hye Rim Jang
- Laboratory of Mitochondrial and Metabolic Diseases, Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, South Korea
| | - Hui-Young Lee
- Laboratory of Mitochondrial and Metabolic Diseases, Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon 21999, South Korea
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, South Korea
- Division of Molecular Medicine, Department of Medicine, Gachon University College of Medicine, Incheon 21936, South Korea
| |
Collapse
|
41
|
Jaago M, Timmusk US, Timmusk T, Palm K. Drastic Effects on the Microbiome of a Young Rower Engaged in High-Endurance Exercise After a Month Usage of a Dietary Fiber Supplement. Front Nutr 2021; 8:654008. [PMID: 33996876 PMCID: PMC8119783 DOI: 10.3389/fnut.2021.654008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/22/2021] [Indexed: 01/12/2023] Open
Abstract
Food supplements are increasingly used worldwide. However, research on the efficacy of such supplements on athlete's well-being and optimal sports performance is very limited. This study performed in junior academic rowing explores the effects of nutritional supplements to aid to the high energy requirements at periods of intense exercise. Herein, the effects of prebiotic fibers on the intestinal microbiome composition of an 18-year-old athlete exercising at high loads during an 8-month period in a “real-life” setting were examined using next-generation sequencing analysis. Results demonstrated that although the alpha diversity of the subject's microbiome drastically decreased [from 2.11 precompetition to 1.67 (p < 0.05)] upon fiber consumption, the Firmicutes/Bacteroidetes ratio increased significantly [from 3.11 to 4.55, as compared with population average (p < 0.05)]. Underlying these macrolevel microbial alterations were demonstrable shifts from acetate- to butyrate-producing bacteria, although with stable effects on the Veillonella species. To our knowledge, this a unique study that shows pronounced changes in the gut microbiome of the young athlete at the competition season and their favorable compensation by the dietary fiber intake. The data here expand the overall understanding of how the high energy needs in high-intensity sports like academic rowing could be supported by dietary fiber supplement consumption.
Collapse
Affiliation(s)
- Mariliis Jaago
- Protobios Llc, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Tõnis Timmusk
- Protobios Llc, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Kaia Palm
- Protobios Llc, Tallinn, Estonia.,Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| |
Collapse
|
42
|
Abstract
The functional diversity of the mammalian intestinal microbiome far exceeds that of the host organism, and microbial genes contribute substantially to the well-being of the host. However, beneficial gut organisms can also be pathogenic when present in the gut or other locations in the body. Among dominant beneficial bacteria are several species of Bacteroides, which metabolize polysaccharides and oligosaccharides, providing nutrition and vitamins to the host and other intestinal microbial residents. These topics and the specific organismal and molecular interactions that are known to be responsible for the beneficial and detrimental effects of Bacteroides species in humans comprise the focus of this review. The complexity of these interactions will be revealed.
Collapse
Affiliation(s)
- Hassan Zafar
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, USA
- Department of Microbiology and Molecular Genetics, Faculty of Life Sciences, University of Okara,Okara, PunjabPakistan
| | - Milton H. Saier
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, USA
| |
Collapse
|
43
|
Linz B, Saljic A, Hohl M, Gawałko M, Jespersen T, Sanders P, Böhm M, Linz D. Inhibition of sodium-proton-exchanger subtype 3-mediated sodium absorption in the gut: A new antihypertensive concept. IJC HEART & VASCULATURE 2020; 29:100591. [PMID: 32760780 PMCID: PMC7390783 DOI: 10.1016/j.ijcha.2020.100591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/24/2020] [Accepted: 07/07/2020] [Indexed: 12/22/2022]
Abstract
Arterial hypertension is one of the main contributors to cardiovascular diseases, including stroke, heart failure, and coronary heart disease. Salt plays a major role in the regulation of blood pressure and is one of the most critical factors for hypertension and stroke. At the individual level, effective salt reduction is difficult to achieve and available methods for managing sodium balance are lacking for many patients. As part of the ingested food, salt is absorbed in the gastrointestinal tract by the sodium proton exchanger subtype 3 (NHE3 also known as Slc9a3), influencing extracellular fluid volume and blood pressure. In this review, we discuss the beneficial effects of pharmacological inhibition of NHE3-mediated sodium absorption in the gut and focus on the effect on blood pressure and end-organ damage.
Collapse
Affiliation(s)
- Benedikt Linz
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnela Saljic
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mathias Hohl
- Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes
| | - Monika Gawałko
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Department of Cardiology, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Thomas Jespersen
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Michael Böhm
- Kardiologie, Angiologie und Internistische Intensivmedizin, Universitätsklinikum des Saarlandes
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Centre, Maastricht, the Netherlands
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
- University Maastricht, Cardiovascular Research Institute Maastricht (CARIM), the Netherlands
- Corresponding author at: Maastricht UMC+, Maastricht Heart+Vascular Center, 6202 AZ Maastricht, the Netherlands.
| |
Collapse
|
44
|
Bibbò S, Abbondio M, Sau R, Tanca A, Pira G, Errigo A, Manetti R, Pes GM, Dore MP, Uzzau S. Fecal Microbiota Signatures in Celiac Disease Patients With Poly-Autoimmunity. Front Cell Infect Microbiol 2020; 10:349. [PMID: 32793511 PMCID: PMC7390951 DOI: 10.3389/fcimb.2020.00349] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022] Open
Abstract
To date, reliable tests enabling the identification of celiac disease (CD) patients at a greater risk of developing poly-autoimmune diseases are not yet available. We therefore aimed to identify non-invasive microbial biomarkers, useful to implement diagnosis of poly-autoimmunity. Twenty CD patients with poly-autoimmunity (cases) and 30 matched subjects affected exclusively by CD (controls) were selected. All patients followed a varied gluten-free diet for at least 1 year. Fecal microbiota composition was characterized using bacterial 16S ribosomal RNA gene sequencing. Significant differences in gut microbiota composition between CD patients with and without poly-autoimmune disease were found using the edgeR algorithm. Spearman correlations between gut microbiota and clinical, demographic, and anthropometric data were also examined. A significant reduction of Bacteroides, Ruminococcus, and Veillonella abundances was found in CD patients with poly-autoimmunity compared to the controls. Bifidobacterium was specifically reduced in CD patients with Hashimoto's thyroiditis and its abundance correlated negatively with abdominal circumference values in patients affected exclusively by CD. In addition, the duration of CD correlated with the abundance of Firmicutes (negatively) and Odoribacter (positively), whereas the abundance of Desulfovibrionaceae correlated positively with the duration of poly-autoimmunity. This study provides supportive evidence that specific variations of gut microbial taxa occur in CD patients with poly-autoimmune diseases. These findings open the way to future validation studies on larger cohorts, which might in turn lead to promising diagnostic applications.
Collapse
Affiliation(s)
- Stefano Bibbò
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Marcello Abbondio
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Rosangela Sau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandro Tanca
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Giovanna Pira
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Alessandra Errigo
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Roberto Manetti
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Giovanni Mario Pes
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy
| | - Maria Pina Dore
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Sassari, Italy.,Baylor College of Medicine, Houston, TX, United States
| | - Sergio Uzzau
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
45
|
Markey KA, Schluter J, Gomes ALC, Littmann ER, Pickard AJ, Taylor BP, Giardina PA, Weber D, Dai A, Docampo MD, Armijo GK, Slingerland AE, Slingerland JB, Nichols KB, Brereton DG, Clurman AG, Ramos RJ, Rao A, Bush A, Bohannon L, Covington M, Lew MV, Rizzieri DA, Chao N, Maloy M, Cho C, Politikos I, Giralt S, Taur Y, Pamer EG, Holler E, Perales MA, Ponce DM, Devlin SM, Xavier J, Sung AD, Peled JU, Cross JR, van den Brink MRM. The microbe-derived short-chain fatty acids butyrate and propionate are associated with protection from chronic GVHD. Blood 2020; 136:130-136. [PMID: 32430495 PMCID: PMC7332893 DOI: 10.1182/blood.2019003369] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 04/19/2020] [Indexed: 01/10/2023] Open
Abstract
Studies of the relationship between the gastrointestinal microbiota and outcomes in allogeneic hematopoietic stem cell transplantation (allo-HCT) have thus far largely focused on early complications, predominantly infection and acute graft-versus-host disease (GVHD). We examined the potential relationship of the microbiome with chronic GVHD (cGVHD) by analyzing stool and plasma samples collected late after allo-HCT using a case-control study design. We found lower circulating concentrations of the microbe-derived short-chain fatty acids (SCFAs) propionate and butyrate in day 100 plasma samples from patients who developed cGVHD, compared with those who remained free of this complication, in the initial case-control cohort of transplant patients and in a further cross-sectional cohort from an independent transplant center. An additional cross-sectional patient cohort from a third transplant center was analyzed; however, serum (rather than plasma) was available, and the differences in SCFAs observed in the plasma samples were not recapitulated. In sum, our findings from the primary case-control cohort and 1 of 2 cross-sectional cohorts explored suggest that the gastrointestinal microbiome may exert immunomodulatory effects in allo-HCT patients at least in part due to control of systemic concentrations of microbe-derived SCFAs.
Collapse
Affiliation(s)
- Kate A Markey
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | | | - Antonio L C Gomes
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eric R Littmann
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amanda J Pickard
- Department of Hematology and Oncology, Internal Medicine III, University Medical Center, Regensburg, Germany
| | | | - Paul A Giardina
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniela Weber
- Department of Hematology and Oncology, Internal Medicine III, University Medical Center, Regensburg, Germany
| | - Anqi Dai
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Melissa D Docampo
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gabriel K Armijo
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ann E Slingerland
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - John B Slingerland
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katherine B Nichols
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel G Brereton
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Annelie G Clurman
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ruben J Ramos
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Arka Rao
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Amy Bush
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - Lauren Bohannon
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - Megan Covington
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - Meagan V Lew
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - David A Rizzieri
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - Nelson Chao
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - Molly Maloy
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Christina Cho
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ioannis Politikos
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Sergio Giralt
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Ying Taur
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Infectious Disease Service, Department of Medicine, and
| | - Eric G Pamer
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Infectious Disease Service, Department of Medicine, and
| | - Ernst Holler
- Department of Hematology and Oncology, Internal Medicine III, University Medical Center, Regensburg, Germany
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Doris M Ponce
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Joao Xavier
- Program for Computational and Systems Biology, and
| | - Anthony D Sung
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University Medical Center, Durham, NC; and
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
- Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
46
|
Jin JB, Cha JW, Shin IS, Jeon JY, Cha KH, Pan CH. Supplementation with Chlorella vulgaris, Chlorella protothecoides, and Schizochytrium sp. increases propionate-producing bacteria in in vitro human gut fermentation. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2020; 100:2938-2945. [PMID: 32031246 DOI: 10.1002/jsfa.10321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Gut microbiota are major contributors to host metabolism and are considered as potential targets of novel therapeutics. Microalgae have a strong potential for use as prebiotics because they are a rich source of proteins, fatty acids, fiber, and minerals for nutritional supplementation in humans. Nevertheless, there has been insufficient research into the effect of microalgae on gut microbiota. To investigate the effects of three edible microalgae (Chlorella vulgaris, Chlorella protothecoides, and Schizochytrium sp.) on gut microbiota, simulated digestion and colonic fermentation were examined. RESULTS Following in vitro digestion, the microalgae displayed different levels of bioaccessibility and the nutrient analysis revealed that unabsorbed nutrients during the digestion process could be used for colonic fermentation. Following colonic fermentation, the control, inulin, and microalgae groups displayed different metabolite tendencies when investigated with nuclear magnetic resonance (NMR) spectroscopic analysis. In particular, microalgae supplementation increased the proportion of propionate in the colonic culture (control: 19.14%, Inulin: 18.38%, C. vulgaris: 25.80%, C. protothecoides: 25.46%, and Schizochytrium sp.: 25.56%). Microbial profiling analysis using 16S rRNA gene sequencing also disclosed that the relative abundance of Bacteroides (control: 1.91%, inulin: 2.61%, C. vulgaris: 14.77%, C. protothecoides: 11.17%, and Schizochytrium sp.: 5.51%) and Dialister (control: 0.08%, inulin: 2.06%, C. vulgaris: 6.79%, C. protothecoides: 4.45%, and Schizochytrium sp.: 4.48%), involved in propionate metabolism increased more than in the inulin group. CONCLUSION Our findings suggest the potential use of microalgae as a functional food to increase propionate generation because propionate has been reported to be effective in weight loss and the inhibition of pathogen infection. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jong Beom Jin
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, Republic of Korea
| | - Jin Wook Cha
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, Republic of Korea
| | - Il-Shik Shin
- Department of Marine Food Science and Technology, Gangneung-Wonju National University, Gangneung, Republic of Korea
| | - Jin Young Jeon
- Research & Technology Division Ingredient Research Department 2, Ingredient Business Unit, Daesang Corporation, Seoul, Republic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, Republic of Korea
| | - Cheol-Ho Pan
- Natural Product Informatics Research Center, KIST Gangneung Institute of Natural Products, Gangneung, Republic of Korea
| |
Collapse
|
47
|
Lee TH, Park D, Kim YJ, Lee I, Kim S, Oh CT, Kim JY, Yang J, Jo SK. Lactobacillus salivarius BP121 prevents cisplatin‑induced acute kidney injury by inhibition of uremic toxins such as indoxyl sulfate and p‑cresol sulfate via alleviating dysbiosis. Int J Mol Med 2020; 45:1130-1140. [PMID: 32124946 PMCID: PMC7053870 DOI: 10.3892/ijmm.2020.4495] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 12/24/2019] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota is important for maintaining the integrity of the intestinal barrier, promoting immunological tolerance and carrying out metabolic activities that have not evolved in hosts. Intestinal dysbiosis is associated with chronic kidney disease and probiotic supplementation has been shown to be beneficial. However, it is not known whether gut microorganisms‑specifically, lactic acid bacteria (LAB) can protect against acute kidney injury (AKI). To address this issue, the present study investigated the effects of Lactobacillus salivarius BP121, an intestinal LAB isolated from the feces of newborns, in a rat model of cisplatin‑induced AKI and also in Caco‑2 human intestinal epithelial cells. BP121 prevented cisplatin‑induced AKI in rats, as demonstrated by decreases in inflammation and oxidative stress in kidney tissue and in serum levels of uremic toxins such as indoxyl sulfate (IS) and p‑cresol sulfate (PCS). BP121 also reduced intestinal permeability, as determined using fluorescein isothiocyanate‑dextran by immunohistochemical detection of tight junction (TJ) proteins such as zona occludens‑1 and occludin. The abundance of Lactobacillus spp., which are beneficial intestinal flora, was increased by BP121; this was accompanied by an increase in the concentrations of short‑chain fatty acids in feces. Additionally, H2O2‑induced TJ protein damage was reduced in Caco‑2 cells treated with BP121 culture supernatant, an effect that was reversed by the 5' AMP‑activated protein kinase (AMPK) inhibitor Compound C and Toll‑like receptor (TLR)4 inhibitor TLR4‑IN‑C34. In conclusion, this study demonstrated that L. salivarius BP121 protects against cisplatin‑induced AKI by decreasing inflammation and oxidative stress and this renoprotective effect is partially mediated by modulating the gut environment and thereby suppressing IS and PCS production as well as by regulating AMPK and TLR4 dependent TJ assembly.
Collapse
Affiliation(s)
- Tae-Hee Lee
- Department of Toxicity Evaluation, Centralbio Co., Ltd., Gimpo, Gyeonggi 31499
| | - Dongsun Park
- Department of Biology Education, Korea National University of Education, Cheongju, North Chungcheong 28173
| | - Yang Ji Kim
- Department of Toxicity Evaluation, Centralbio Co., Ltd., Gimpo, Gyeonggi 31499
- Department of Toxicity Evaluation, Konyang University, Daejeon 35365
| | - Isaac Lee
- Hoseo Toxicological Research Center, Hoseo University, Asan, Chungnam 31499
| | - Suae Kim
- Research Institute, Research and Development Center, Green Cross WellBeing Corporation, Seongnam, Gyeonggi 13595
| | - Chang-Taek Oh
- Research Institute, Research and Development Center, Green Cross WellBeing Corporation, Seongnam, Gyeonggi 13595
| | - Joem-Yong Kim
- Research Institute, Research and Development Center, Green Cross WellBeing Corporation, Seongnam, Gyeonggi 13595
| | - Jihyun Yang
- Department of Internal Medicine, Division of Nephrology, Korea University Medical College, Seoul 02841, Republic of Korea
| | - Sang-Kyung Jo
- Department of Internal Medicine, Division of Nephrology, Korea University Medical College, Seoul 02841, Republic of Korea
| |
Collapse
|
48
|
Ma S, Yeom J, Lim YH. Dairy Propionibacterium freudenreichii ameliorates acute colitis by stimulating MUC2 expression in intestinal goblet cell in a DSS-induced colitis rat model. Sci Rep 2020; 10:5523. [PMID: 32218552 PMCID: PMC7099060 DOI: 10.1038/s41598-020-62497-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/09/2020] [Indexed: 12/21/2022] Open
Abstract
An intact mucus layer is important in managing inflammatory bowel disease (IBD). Dairy Propionibacterium freudenreichii has probiotic potential, produces propionic acid and is known to promote health. The aim of this study was to evaluate the effects of P. freudenreichii on the improvement of colitis. LS 174T goblet cells and a dextran sodium sulfate (DSS)-induced colitis rat model were used to investigate the P. freudenreichii-induced stimulation of mucin production in vitro and in vivo, respectively. The mRNA and protein expression levels of MUC2, a main component of intestinal mucus, increased in the supernatant of P. freudenreichii culture (SPFC)-treated LS 174 cells. The SPFC and live P. freudenreichii (LPF) reduced the disease activity index (DAI) in the rats with DSS-induced colitis. After treatment with SPFC or LPF, the mRNA levels of typical pro-inflammatory cytokines decreased and the inflammatory state was histologically improved in the rats with DSS-induced colitis. The SPFC and LPF treatments increased the gene and protein expression levels of MUC2 in the rats with DSS-induced colitis compared with the expression levels in the negative control rats, and immunohistochemistry (IHC) showed an increase of the intestinal MUC2 level. In addition, SPFC and LPF augmented the level of propionate in the faeces of the rats with DSS-induced colitis. In conclusion, P. freudenreichii might improve acute colitis by restoring goblet cell number and stimulating the expression of MUC2 in intestinal goblet cells.
Collapse
Affiliation(s)
- Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Public Health Science (Brain Korea 21 PLUS program), Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Laboratory Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
49
|
Kao HF, Wang YC, Tseng HY, Wu LSH, Tsai HJ, Hsieh MH, Chen PC, Kuo WS, Liu LF, Liu ZG, Wang JY. Goat Milk Consumption Enhances Innate and Adaptive Immunities and Alleviates Allergen-Induced Airway Inflammation in Offspring Mice. Front Immunol 2020; 11:184. [PMID: 32132998 PMCID: PMC7040033 DOI: 10.3389/fimmu.2020.00184] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/23/2020] [Indexed: 11/13/2022] Open
Abstract
Goat milk (GM), as compared to cow milk (CM), is easier for humans to digest. It also has antioxidant and anti-inflammatory effects and can improve minor digestive disorders and prevent allergic diseases in infants. It is unclear whether GM consumed in pregnant mothers has any protective effects on allergic diseases in infants. In this experimental study with mice, we found GM feeding enhanced immunoglobulin production, antigen-specific (ovalbumin, OVA) immune responses, and phagocytosis activity. The GM-fed mice had an increasing proportion of CD3+ T lymphocytes in the spleen. Splenocytes isolated from these animals also showed significantly increased production of cytokines IFN-γ and IL-10. More importantly, GM feeding during pregnancy and lactation periods can confer protective activity onto offspring by alleviating the airway inflammation of allergic asthma induced by mite allergens. There was a remarkably different composition of gut microbiota between offspring of pregnant mice fed with water or with milk (GM or CM). There was a greater proportion of beneficial bacterial species, such as Akkermansia muciniphila, Bacteroides eggerthii, and Parabacteroides goldsteinii in the gut microbiota of offspring from GM- or CM-fed pregnant mice compared to the offspring of water-fed pregnant mice. These results suggested that improving the nutrition of pregnant mice can promote immunological maturation and colonization of gut microbiota in offspring. This mother-to-child biological action may provide a protective effect on atopy development and alleviate allergen-induced airway inflammation in offspring.
Collapse
Affiliation(s)
- Hui-Fang Kao
- Department of Nursing, National Tainan Junior College of Nursing, Tainan, Taiwan
| | - Yu-Chin Wang
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Hsiu-Ying Tseng
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | | | - Hui-Ju Tsai
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Miao-Hsi Hsieh
- Graduate Institute of Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chi Chen
- Graduate Institute of Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wen-Shou Kuo
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,School of Chemistry and Materials Science, Nanjing University of Information Science and Technology, Nanjing, China
| | - Li-Fan Liu
- Institute of Gerontology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Zhi-Gang Liu
- Department of Respirology and Allergy, Third Affiliated Hospital of Shengzhen University, Shengzhen, China
| | - Jiu-Yao Wang
- Center for Allergy and Clinical Immunology Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
50
|
Engevik MA, Morra CN, Röth D, Engevik K, Spinler JK, Devaraj S, Crawford SE, Estes MK, Kalkum M, Versalovic J. Microbial Metabolic Capacity for Intestinal Folate Production and Modulation of Host Folate Receptors. Front Microbiol 2019; 10:2305. [PMID: 31649646 PMCID: PMC6795088 DOI: 10.3389/fmicb.2019.02305] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022] Open
Abstract
Microbial metabolites, including B complex vitamins contribute to diverse aspects of human health. Folate, or vitamin B9, refers to a broad category of biomolecules that include pterin, para-aminobenzoic acid (pABA), and glutamate subunits. Folates are required for DNA synthesis and epigenetic regulation. In addition to dietary nutrients, the gut microbiota has been recognized as a source of B complex vitamins, including folate. This study evaluated the predicted folate synthesis capabilities in the genomes of human commensal microbes identified in the Human Microbiome Project and folate production by representative strains of six human intestinal bacterial phyla. Bacterial folate synthesis genes were ubiquitous across 512 gastrointestinal reference genomes with 13% of the genomes containing all genes required for complete de novo folate synthesis. An additional 39% of the genomes had the genetic capacity to synthesize folates in the presence of pABA, an upstream intermediate that can be obtained through diet or from other intestinal microbes. Bacterial folate synthesis was assessed during exponential and stationary phase growth through the evaluation of expression of select folate synthesis genes, quantification of total folate production, and analysis of folate polyglutamylation. Increased expression of key folate synthesis genes was apparent in exponential phase, and increased folate polyglutamylation occurred during late stationary phase. Of the folate producers, we focused on the commensal Lactobacillus reuteri to examine host-microbe interactions in relation to folate and examined folate receptors in the physiologically relevant human enteroid model. RNAseq data revealed segment-specific folate receptor distribution. Treatment of human colonoid monolayers with conditioned media (CM) from wild-type L. reuteri did not influence the expression of key folate transporters proton-coupled folate transporter (PCFT) or reduced folate carrier (RFC). However, CM from L. reuteri containing a site-specific inactivation of the folC gene, which prevents the bacteria from synthesizing a polyglutamate tail on folate, significantly upregulated RFC expression. No effects were observed using L. reuteri with a site inactivation of folC2, which results in no folate production. This work sheds light on the contributions of microbial folate to overall folate status and mammalian host metabolism.
Collapse
Affiliation(s)
- Melinda A. Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Christina N. Morra
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, United States
| | - Daniel Röth
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - Kristen Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Jennifer K. Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Sridevi Devaraj
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| | - Sue E. Crawford
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Department of Medicine – Gastroenterology, Hepatology and Infectious Diseases, Baylor College of Medicine, Houston, TX, United States
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, CA, United States
- Mass Spectrometry and Proteomics Core, Beckman Research Institute of the City of Hope, Duarte, CA, United States
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States
- Department of Pathology, Texas Children’s Hospital, Houston, TX, United States
| |
Collapse
|