1
|
Thomas ZO, Holm J, McCarthy M, Nguyen W, Pang Y, Chrisman LP, Guitart J, Burns MB, Zhou XA. Decreased gut short-chain fatty acids in cutaneous T-cell lymphoma: a novel insight. Arch Dermatol Res 2025; 317:781. [PMID: 40411534 DOI: 10.1007/s00403-025-04285-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/04/2025] [Accepted: 05/13/2025] [Indexed: 05/26/2025]
Abstract
Short-chain fatty acids (SCFAs) are critical metabolites produced by gut microbiota that play a key role in modulating inflammation and regulating systemic immunity, including against cancer. Decreases in SCFAs can foster a permissive tumor immune environment. Recent studies have shown that cutaneous T-cell lymphoma (CTCL) patients exhibit increasing gut dysbiosis and loss of bacteria predicted to produce SCFAs with increasing disease severity. To investigate this functional connection, we collected stool swab samples from 15 individuals- 8 mycosis fungoides (MF) patients and 7 matched healthy controls (HC)- and quantified concentrations of four SCFAs (acetate, propionate, isovalerate, butyrate) via liquid chromatography-mass spectrometry. Our results demonstrated significantly reduced acetate and propionate concentrations in MF patients when compared to HC (both p = 0.027). Total measured SCFA concentrations were on average lower in MF versus HC, but did not achieve statistical significance (p = 0.063). Both propionate and acetate have been previously demonstrated to promote tumor apoptosis, inhibit tumor proliferation, and enhance antitumor immunity. Thus, dysbiosis-associated reductions in SCFAs may be another contributive factor in the immune dysfunction observed in CTCL. Our pilot findings add to the growing body of knowledge implicating the gut microbiota-SCFA axis in CTCL pathogenesis and offer potential new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Zachary O Thomas
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - JoJo Holm
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - Morgan McCarthy
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - William Nguyen
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - Yanzhen Pang
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - Lauren P Chrisman
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - Joan Guitart
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA
| | - Michael B Burns
- Department of Biology, Loyola University Chicago, Chicago, IL, USA
| | - Xiaolong Alan Zhou
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, 676 N St Clair St Suite 1600, Chicago, IL, 60611, USA.
| |
Collapse
|
2
|
Hunjan G, Shah SS, Kosey S, Aran KR. Gut microbiota and the tryptophan-kynurenine pathway in anxiety: new insights and treatment strategies. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02938-8. [PMID: 40369368 DOI: 10.1007/s00702-025-02938-8] [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: 11/29/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025]
Abstract
Anxiety disorders are mental health disorders characterized by long-lasting fear, worry, nervousness, and alterations in gut microbiota (GM). The GM is a vital modulator of brain function through the gut-brain axis, which acts as the neural pathway between the central and peripheral nervous systems. Dysbiosis of GM plays an essential role in anxiety development because of alterations in the vagus nerve, increased intestinal permeability, and altered breakdown of tryptophan (TRP). The Kynurenine (KYN) pathway plays a crucial role in the pathogenesis of anxiety disorders, primarily through its neuroprotective (KYNA) and neurotoxic (QUIN) metabolites. Higher ratios of KYNA/QUIN result in neuroprotection, whereas higher KYN/TRP ratios indicate increased QUIN production causing neuroinflammation. Studies on germ-free models exhibit higher plasma TRP levels, which interrupt the metabolic balance of TRP-derived compounds, thus causing brain impairment. A key issue in anxiety disorders is the dysregulation of GM, which disrupts TRP metabolism and neuroinflammatory pathways, however, remains poorly understood. Hence, the proper understanding of these mechanisms is crucial for future therapeutic advancements. Here, we highlight the significance of the TRP-KYN pathway and the potential of modulating KYN pathway enzymes, such as kynurenine aminotransferases (KATs), to adjust KYNA levels and restore neurotransmitter balance. It further discusses new therapeutic methods with a particular focus on probiotics that may restore GM and modulate TRP metabolism. Advancing our understanding of the intricate relationship between GM and anxiety disorders may facilitate novel, microbiota-targeted interventions. This ultimately contributes to precision medicine approaches in mental health care, thereby enhancing treatment efficacy and patient outcomes.
Collapse
Affiliation(s)
- Garry Hunjan
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Shiv Shankar Shah
- Krupanidhi College of Pharmacy, Carmelaram Gunjur Road, Hobli, off Sarjapur Road, Varthur, Bengaluru, 560035, Karnataka, India
| | - Sourabh Kosey
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, 142001, Punjab, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, 142001, Punjab, India.
| |
Collapse
|
3
|
Perez Umana ER, Mendes E, Casaro MC, Lazarini M, Oliveira FA, Sperling AI, Ferreira CM. Exogenous acetate mitigates later enhanced allergic airway inflammation in a menopausal mouse model. Front Cell Infect Microbiol 2025; 15:1543822. [PMID: 40292217 PMCID: PMC12023485 DOI: 10.3389/fcimb.2025.1543822] [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: 12/11/2024] [Accepted: 03/06/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Asthma, an inflammatory lung disease, disproportionately affects women in adulthood and is associated with a decline in estrogen levels during the menstrual cycle and menopause. To study asthma symptoms during menopause, we used a mouse model of postmenopausal asthma via ovariectomy (OVx). Similar to human menopause, we previously discovered that re-exposure of allergic OVx mice to allergen exacerbates lung inflammation. Surprisingly, we found that probiotic treatment alleviates this inflammatory exacerbation and produces acetate as one of its metabolites. Here, we investigate whether exogenous acetate alone can inhibit the exacerbation of experimental asthma in menopause. Methods Mice received acetate administration before and during sensitization. After challenge and OVx the mice were subjected to a second challenge to test whether acetate protected against airway inflammation after menopause induction. Results Acetate administration reduced all lung T2 inflammatory responses, as well as the serum immunoglobulin (IgE) level. Early acetate treatment led to an increase in regulatory T cells, even 3 weeks after cessation of the treatment, suggesting that the increase in Treg percentage is associated with the reduction of type 2 inflammation in the airways after menopause induction, indicating its potential role in this process. Given the significant role of the lung-gut axis in asthma and the association of asthma and menopause with intestinal dysfunctions, this finding is particularly relevant; we also analyzed several markers of intestinal integrity. Compared with sham-operated mice, rechallenged allergic menopausal mice had a reduction in the intestinal epithelial genes, MUC2 and OCLN, and preventive supplementation with acetate returned their expression to normal. No change was found in menopausal mice without allergic inflammation. Conclusion In conclusion, treatment with acetate prior to estrogen level decline protects sensitized and challenged mice against later airway T2 inflammation and may restore gut homeostasis.
Collapse
Affiliation(s)
- Evelyn Roxana Perez Umana
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Eduardo Mendes
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Mateus Campos Casaro
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Mariana Lazarini
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| | - Fernando A. Oliveira
- Cellular and Molecular Neurobiology Laboratory (LaNeC), Center of Mathematics, Computing and Cognition (CMCC), Federal University of ABC, São Bernando do Campo, Brazil
| | - Anne I. Sperling
- Pulmonary and Critical Care Laboratory, Department of Medicine, University of Virginia, Charlottesville, VA, United States
| | - Caroline Marcantonio Ferreira
- Institute of Environmental, Chemistry and Pharmaceutical Sciences, Department of Pharmaceutics Sciences, University Federal de São Paulo, Diadema, Brazil
| |
Collapse
|
4
|
Wijaya AJ, Dolan SK, Kohlstedt M, Gläser L, Brear P, Geddis S, Wittmann C, Spring DR, Welch M. The 2-methylcitrate cycle and the glyoxylate shunt in Pseudomonas aeruginosa are linked through enzymatic redundancy. J Biol Chem 2025; 301:108355. [PMID: 40015638 PMCID: PMC11982470 DOI: 10.1016/j.jbc.2025.108355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/03/2025] [Accepted: 02/13/2025] [Indexed: 03/01/2025] Open
Abstract
The 2-methylcitrate cycle and the glyoxylate cycle are central metabolic pathways in Pseudomonas aeruginosa, enabling the organism to utilize organic acids such as propionate and acetate during infection. Here, we show that these cycles are linked through enzymatic redundancy, with isocitrate lyase (AceA) exhibiting secondary 2-methylisocitrate lyase activity. Furthermore, we use a combination of structural analyses, enzyme kinetics, metabolomics, and targeted mutation of PrpBPa to demonstrate that whereas loss of PrpB function impairs growth on propionate, the promiscuous 2-methylisocitrate lyase activity of AceA compensates for this by mitigating the accumulation of toxic 2-methylcitrate cycle intermediates. Our findings suggest that simultaneous inhibition of PrpB and AceA could present a robust antimicrobial strategy to target P. aeruginosa in propionate-rich environments, such as the cystic fibrosis airways. Our results emphasize the importance of understanding pathway interconnections in the development of novel antimicrobial agents.
Collapse
Affiliation(s)
- Andre J Wijaya
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Stephen K Dolan
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom; Department of Genetics and Biochemistry, Eukaryotic Pathogens Innovation Center, Clemson University, Clemson, USA.
| | - Michael Kohlstedt
- Institute of Systems Biotechnology, Saarland University, Saarbrücken, Germany
| | - Lars Gläser
- Institute of Systems Biotechnology, Saarland University, Saarbrücken, Germany
| | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Stephen Geddis
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Wittmann
- Institute of Systems Biotechnology, Saarland University, Saarbrücken, Germany
| | - David R Spring
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| | - Martin Welch
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
5
|
Yang J, Chen Y, Tian X, Yu H, Shi Y, Zhao D, Long X, Tan Q, Li H. Effect of dietary Dendrobium nobile Lindl. On production performance, immune function and caecal microbiota of Chishui black-bone hens. Sci Rep 2025; 15:9745. [PMID: 40118906 PMCID: PMC11928661 DOI: 10.1038/s41598-025-91901-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
The objective of this study was to investigate the effects of dietary supplementation of Dendrobium nobile Lindl. (DNL) on production performance, immune function, and caecal microbiota of Chishui black-bone hens. A total of one hundred and eighty 600-day-old Chishui black-bone hens were randomly allocated to two experimental groups, with six replicates of 15 chickens per group. The control group was fed a basal diet, and the experimental group was supplemented with 2100 mg/kg DNL in the basal diet. The results demonstrated that compared with the control group, the addition of 2100 mg/kg DNL to the diet significantly (P < 0.05) increased the average egg production rate of Chishui black-bone hens from 38.97 to 44.95%. Interferon-gamma (IFN-γ) levels were significantly lower in the experimental group than in the control group, whereas serum immunoglobulin A (IgA) levels were significantly higher (P < 0.05). However, adding DNL exerted no significant effect on egg quality. Asp, Ser, Gly, Leu, Phe, His, Pro, TAA, EAA, and NEAA levels in the eggs of the experimental group were significantly higher (P < 0.05) than in those in the control group. Additionally, the EAA/TAA in the control and experimental groups were 40.38% and 40.76%, respectively, and the EAA/NEAA was 76.62% and 74.72%, both of which were higher than the 40% and 60% thresholds specified in the WHO/HAO ideal protein guidelines. The results also indicated that eggs in the experimental group had significantly higher contents of palmitic acid (C16:0), stearic acid (C18:0), palmitic acid (C16:1), oleic acid (C18:ln9c), behenolic acid (C22:0), palmitic acid (C16:), oleic acid (C18:ln9c), saturated fatty acids (SFA), and monounsaturated fatty acids (MUFA) (P < 0.05). In contrast, the contents of hexadecenoic acid (C15:0), heptadecanoic acid (C17:0), α-linolenic acid (C18:3n3), arachidonic acid (C20:0), C10 heptadecanoic acid (C17:1), linoleic acid (C18:2n6c), α-linolenic acid (C18:3n3), eicosapentaenoic acid (C20:2), Docosapentaenoic Acid (DPA), polyunsaturated fatty acid (PUFA), and n-6PUFA were significantly reduced (P < 0.05). Moreover, when 2100 mg/kg DNL was added to the diet of Chishui black-bone hens, the composition of the intestinal flora was altered. Specifically, the relative abundances of Bacteroides, Lactobacillus, Subdoligranulum, and Parabacteroides increased, whereas those of Desulfovibrio, Lachnoclostridium, Ruminococcaceae, Fournierella, Faecalibacterium, and Oribacterium decreased. In conclusion, adding 2100 mg/kg DNL to the feed can increase serum IgA and reduce IFN-γ content in Chishui black-bone hens during the late laying period. Furthermore, it can enhance the laying rate and types of amino acids in eggs, as well as the ratio of ideal proteins. Simultaneously, it improved the caecal microbial community of Chishui black-bone hens and increased the relative abundance of beneficial bacteria.
Collapse
Affiliation(s)
- Jinlin Yang
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Youbo Chen
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Xingzhou Tian
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Huan Yu
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Yushi Shi
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Depeng Zhao
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Xia Long
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Qisong Tan
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China
| | - Hui Li
- College of Animal Science, Guizhou University, Guiyang, 550025, Guizhou, China.
- Key Laboratory of Plateau Mountain Animal Genetics Breeding and Reproduction of Ministry of Education, Guizhou Provincial Key Laboratory of Animal Genetics Breeding and Reproduction, Guizhou University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
6
|
LeGrand EK. Beyond nutritional immunity: immune-stressing challenges basic paradigms of immunometabolism and immunology. Front Nutr 2025; 12:1508767. [PMID: 40013164 PMCID: PMC11860096 DOI: 10.3389/fnut.2025.1508767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Pathogens have the well-known advantage of rapid evolution due to short generation times and large populations. However, pathogens have the rarely noted disadvantage of the vulnerability to stress involved in proliferation as well as being localized. Presented here are numerous new paradigms in immunology, and especially immunometabolism, which are derived from examining how hosts capitalize on pathogen vulnerabilities to stress. Universally, proliferation requires both resources and synthesis, which are vulnerable to resource-limiting stress and damaging/noxious stress, respectively. Pathogens are particularly vulnerable to stress at the time when they are most threatening-when they are proliferating. Since immune cells actively controlling pathogens (effector cells) typically do not proliferate at infected sites, there is a "stress vulnerability gap" wherein proliferating pathogens are more vulnerable to any type of stress than are the attacking effector cells. Hosts actively stress vulnerable proliferating pathogens by restricting resources (resource-limiting stress) and generating noxious waste products (damaging/disruptive stress) in a fundamental defense here-in termed "immune-stressing." While nutritional immunity emphasizes denying pathogens micronutrients, immune-stressing extends the concept to restricting all resources, especially glucose and oxygen, coupled with the generation of noxious metabolic products such as lactic acid, reactive oxygen species (ROS), and heat to further harm or stress the pathogens. At present much of the field of immunometabolism centers on how nutrition and metabolism regulate immune function, a central feature being the inefficient use of glucose via aerobic glycolysis (with much lactate/lactic acid production) by effector immune cells. In contrast, immune-stressing emphasizes how the immune system uses nutrition and metabolism to control infections. Immune-stressing addresses effector cell glycolysis at the infected site by noting that the high uptake of glucose linked with high output of lactic acid is an ideal double-pronged stressor targeting proliferating pathogens. Once the basic vulnerability of pathogen proliferation is recognized, numerous other paradigms of immunometabolism, and immunology as a whole, are challenged.
Collapse
Affiliation(s)
- Edmund K. LeGrand
- Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN, United States
| |
Collapse
|
7
|
Liu HY, Li S, Ogamune KJ, Ahmed AA, Kim IH, Zhang Y, Cai D. Fungi in the Gut Microbiota: Interactions, Homeostasis, and Host Physiology. Microorganisms 2025; 13:70. [PMID: 39858841 PMCID: PMC11767893 DOI: 10.3390/microorganisms13010070] [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: 12/10/2024] [Revised: 12/25/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
The mammalian gastrointestinal tract is a stage for dynamic inter-kingdom interactions among bacteria, fungi, viruses, and protozoa, which collectively shape the gut micro-ecology and influence host physiology. Despite being a modest fraction, the fungal community, also referred to as mycobiota, represents a critical component of the gut microbiota. Emerging evidence suggests that fungi act as early colonizers of the intestine, exerting a lasting influence on gut development. Meanwhile, the composition of the mycobiota is influenced by multiple factors, with diet, nutrition, drug use (e.g., antimicrobials), and physical condition standing as primary drivers. During its establishment, the mycobiota forms both antagonistic and synergistic relationships with bacterial communities within the host. For instance, intestinal fungi can inhibit bacterial colonization by producing alcohol, while certain bacterial pathogens exploit fungal iron carriers to enhance their growth. However, the regulatory mechanisms governing these complex interactions remain poorly understood. In this review, we first introduce the methodologies for studying the microbiota, then address the significance of the mycobiota in the mammalian intestine, especially during weaning when all 'primary drivers' change, and, finally, discuss interactions between fungi and bacteria under various influencing factors. Our review aims to shed light on the complex inter-kingdom dynamics between fungi and bacteria in gut homeostasis and provide insights into how they can be better understood and managed to improve host health and disease outcomes.
Collapse
Affiliation(s)
- Hao-Yu Liu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Shicheng Li
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Kennedy Jerry Ogamune
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Abdelkareem A. Ahmed
- Department of Veterinary Science, Botswana University of Agriculture and Natural Resources, Private Bag 0027, Gaborone P.O. Box 100, Botswana;
| | - In Ho Kim
- Department of Animal Resource & Science, Dankook University, 119 Dandero, Donnamgu Cheonan, Cheonan-si 31116, Republic of Korea;
| | - Yunzeng Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China;
| | - Demin Cai
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (H.-Y.L.); (S.L.); (K.J.O.)
- Jiangsu Key Laboratory of Animal Genetic Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
8
|
Green CG, Ong MLY, Rowland SN, Bongiovanni T, James LJ, Clifford T, Bailey SJ, Heaney LM. Investigating serum concentration profiles of orally ingested short-chain fatty acid supplements. Food Funct 2024; 15:11525-11536. [PMID: 39498577 DOI: 10.1039/d4fo04028g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Acetate, propionate, and butyrate are naturally-occurring short-chain fatty acids (SCFAs) derived from bacterial metabolism of dietary fibre and have been associated with numerous positive health outcomes. All three acids have been shown to offer unique physiological and metabolic effects and, therefore, could be targeted for co-ingestion as part of a nutritional/medicinal plan. However, a better understanding of the outcomes of supplementing in combination on circulating concentration profiles is necessary to confirm uptake efficacy. This study sought to investigate the acute circulating concentration profiles of acetate, propionate, and butyrate following oral supplementation. Three experimental trials were conducted including investigations to understand the impact of capsule coating on circulating concentration profiles, the effect of supplementation dose on uptake kinetics, and the outcome of a short, repeated, supplementation routine on circulating levels. Serum samples were analysed for SCFA content using a quantitative GC-MS assay. It was observed that an acid-resistant coated capsule caused a delayed and blunted blood concentration response, with the non-acid resistant trial displaying earlier and more intense peak serum concentrations. For dose comparison investigations, all SCFAs peaked within 60 min and returned to baseline concentrations by 120 min post-supplementation. A graded dose relationship was present for propionate and butyrate when considering the total circulating exposure across a 240 min monitoring period. In addition, a one-week, twice-daily, repeated supplementation protocol resulted in no changes in basal serum SCFA concentrations. Overall, these data indicate that acetate, propionate, and butyrate display relatively similar circulating concentration profiles following oral co-ingestion, adding knowledge to help inform supplementation strategies for future outcomes where acute elevation of circulating SCFAs is desired.
Collapse
Affiliation(s)
- Christopher G Green
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Marilyn L Y Ong
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
- Exercise and Sports Science Programme, School of Health Sciences, Health Campus, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Samantha N Rowland
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Tindaro Bongiovanni
- Player Health & Performance Department, Palermo Football Club, Palermo, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Lewis J James
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Tom Clifford
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| | - Liam M Heaney
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK.
| |
Collapse
|
9
|
Brochut M, Heinonen T, Snäkä T, Gilbert C, Le Roy D, Roger T. Using weight loss to predict outcome and define a humane endpoint in preclinical sepsis studies. Sci Rep 2024; 14:21150. [PMID: 39256525 PMCID: PMC11387420 DOI: 10.1038/s41598-024-72039-1] [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: 06/07/2024] [Accepted: 09/03/2024] [Indexed: 09/12/2024] Open
Abstract
Preclinical mouse models are critical for understanding the pathophysiological response to infections and developing treatment strategies for sepsis. In keeping with ethical values, researchers follow guidelines to minimize the suffering of the mice. Weight loss is a criteria used as a humane end point, but there is no official recommendation for a maximum weight loss leading to euthanasia. To evaluate whether the thresholds used in daily practice are optimal, we performed a comprehensive retrospective analysis of data generated over 10 years with > 2300 mice used in models of infection with Listeria monocytogenes, Streptococcus pneumoniae, Candida albicans and H1N1 influenza virus. Weight loss segregated mice that survived from those that did not. Statistical analyses revealed that lowering the weight loss thresholds used (none, 30% or 20%) would have increased mortality rates due to the sacrifice of mice that survived infections (p < 0.01-0.001). Power calculations showed high variability and reduction of power as weight loss thresholds approached 20% for S. pneumoniae and L. monocytogenes models. Hence, weight loss thresholds need to be adapted to each model of infection used in a laboratory. Overall, weight loss is a valuable predictor of mortality that contributes to the robustness of composite scores. To our knowledge, this is the most extensive study exploring the relationship between weight loss threshold and sepsis outcome. It underscores the importance of the infection-model-specific evaluation of weight loss for use in clinical scores defining humane endpoints to minimize mouse suffering without compromising statistical power and scientific objectives.
Collapse
Affiliation(s)
- Maëlick Brochut
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Tytti Heinonen
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Tiia Snäkä
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Charly Gilbert
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, CLED.04.407, Chemin des Boveresses 155, 1066, Epalinges, Switzerland.
| |
Collapse
|
10
|
Williams LM, Cao S. Harnessing and delivering microbial metabolites as therapeutics via advanced pharmaceutical approaches. Pharmacol Ther 2024; 256:108605. [PMID: 38367866 PMCID: PMC10985132 DOI: 10.1016/j.pharmthera.2024.108605] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/05/2024] [Accepted: 02/08/2024] [Indexed: 02/19/2024]
Abstract
Microbial metabolites have emerged as key players in the interplay between diet, the gut microbiome, and host health. Two major classes, short-chain fatty acids (SCFAs) and tryptophan (Trp) metabolites, are recognized to regulate inflammatory, immune, and metabolic responses within the host. Given that many human diseases are associated with dysbiosis of the gut microbiome and consequent reductions in microbial metabolite production, the administration of these metabolites represents a direct, multi-targeted treatment. While a multitude of preclinical studies showcase the therapeutic potential of both SCFAs and Trp metabolites, they often rely on high doses and frequent dosing regimens to achieve systemic effects, thereby constraining their clinical applicability. To address these limitations, a variety of pharmaceutical formulations approaches that enable targeted, delayed, and/or sustained microbial metabolite delivery have been developed. These approaches, including enteric encapsulations, esterification to dietary fiber, prodrugs, and nanoformulations, pave the way for the next generation of microbial metabolite-based therapeutics. In this review, we first provide an overview of the roles of microbial metabolites in maintaining host homeostasis and outline how compromised metabolite production contributes to the pathogenesis of inflammatory, metabolic, autoimmune, allergic, infectious, and cancerous diseases. Additionally, we explore the therapeutic potential of metabolites in these disease contexts. Then, we provide a comprehensive and up-to-date review of the pharmaceutical strategies that have been employed to enhance the therapeutic efficacy of microbial metabolites, with a focus on SCFAs and Trp metabolites.
Collapse
Affiliation(s)
- Lindsey M Williams
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States
| | - Shijie Cao
- Department of Pharmaceutics, School of Pharmacy, University of Washington, Seattle, WA 98195, United States.
| |
Collapse
|
11
|
Salvadori M, Rosso G. Update on the gut microbiome in health and diseases. World J Methodol 2024; 14:89196. [PMID: 38577200 PMCID: PMC10989414 DOI: 10.5662/wjm.v14.i1.89196] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/18/2023] [Accepted: 01/27/2024] [Indexed: 03/07/2024] Open
Abstract
The Human Microbiome Project, Earth Microbiome Project, and next-generation sequencing have advanced novel genome association, host genetic linkages, and pathogen identification. The microbiome is the sum of the microbes, their genetic information, and their ecological niche. This study will describe how millions of bacteria in the gut affect the human body in health and disease. The gut microbiome changes in relation with age, with an increase in Bacteroidetes and Firmicutes. Host and environmental factors affecting the gut microbiome are diet, drugs, age, smoking, exercise, and host genetics. In addition, changes in the gut microbiome may affect the local gut immune system and systemic immune system. In this study, we discuss how the microbiome may affect the metabolism of healthy subjects or may affect the pathogenesis of metabolism-generating metabolic diseases. Due to the high number of publications on the argument, from a methodologically point of view, we decided to select the best papers published in referred journals in the last 3 years. Then we selected the previously published papers. The major goals of our study were to elucidate which microbiome and by which pathways are related to healthy and disease conditions.
Collapse
Affiliation(s)
- Maurizio Salvadori
- Department of Renal Transplantation, Careggi University Hospital, Florence 50139, Tuscany, Italy
| | - Giuseppina Rosso
- Division of Nephrology, San Giovanni di Dio Hospital, Florence 50143, Toscana, Italy
| |
Collapse
|
12
|
Shvets Y, Khranovska N, Senchylo N, Ostapchenko D, Tymoshenko I, Onysenko S, Kobyliak N, Falalyeyeva T. Microbiota substances modulate dendritic cells activity: A critical view. Heliyon 2024; 10:e27125. [PMID: 38444507 PMCID: PMC10912702 DOI: 10.1016/j.heliyon.2024.e27125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/07/2024] Open
Abstract
Contemporary research in the field of microbiota shows that commensal bacteria influence physiological activity of different organs and systems of a human organism, such as brain, lungs, immune and metabolic systems. This influence is realized by various processes. One of them is trough modulation of immune mechanisms. Interactions between microbiota and the human immune system are known to be complex and ambiguous. Dendritic cells (DCs) are unique cells, which initiate the development and polarization of adaptive immune response. These cells also interconnect native and specific immune reactivity. A large set of biochemical signals from microbiota in the form of different microbiota associated molecular patterns (MAMPs) and bacterial metabolites that act locally and distantly in the human organism. As a result, commensal bacteria influence the maturity and activity of dendritic cells and affect the overall immune reactivity of the human organism. It then determines the response to pathogenic microorganisms, inflammation, associated with different pathological conditions and even affects the effectiveness of vaccination.
Collapse
Affiliation(s)
- Yuliia Shvets
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Natalia Khranovska
- National Cancer Institute of Ukraine, 33/43 Yuliia Zdanovska Str., Kyiv, Ukraine
| | - Natalia Senchylo
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Danylo Ostapchenko
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Iryna Tymoshenko
- Bogomolets National Medical University, 13 Shevchenka Blvd., Kyiv, Ukraine
| | - Svitlana Onysenko
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
| | - Nazarii Kobyliak
- Bogomolets National Medical University, 13 Shevchenka Blvd., Kyiv, Ukraine
- Medical Laboratory CSD, 22b Zhmerynska Str., Kyiv, Ukraine
| | - Tetyana Falalyeyeva
- Taras Shevchenko National University of Kyiv, 64/13 Volodymyrska Str., Kyiv, Ukraine
- Medical Laboratory CSD, 22b Zhmerynska Str., Kyiv, Ukraine
| |
Collapse
|
13
|
Abstract
Short-chain fatty acids (SCFAs) are a subset of fatty acids that play crucial roles in maintaining normal physiology and developing metabolic diseases, such as obesity, diabetes, cardiovascular disease, and liver disease. Even though dairy products and vegetable oils are the direct dietary sources of SCFAs, their quantities are highly restricted. SCFAs are produced indirectly through microbial fermentation of fibers. The biological roles of SCFAs in human health and metabolic diseases are mainly due to their receptors, GPR41 and GPR43, FFAR2 and FFAR3. Additionally, it has been demonstrated that SCFAs modulate DNMTs and HDAC activities, inhibit NF-κB-STAT signaling, and regulate G(i/o)βγ-PLC-PKC-PTEN signaling and PPARγ-UCP2-AMPK autophagic signaling, thus mitigating metabolic diseases. Recent studies have uncovered that SCFAs play crucial roles in epigenetic modifications of DNAs, RNAs, and post-translational modifications of proteins, which are critical regulators of metabolic health and diseases. At the same time, dietary recommendations for the purpose of SCFAs have been proposed. The objective of the review is to summarize the most recent research on the role of dietary SCFAs in metabolic diseases, especially the signal transduction of SCFAs in metabolic diseases and their functional efficacy in different backgrounds and models of metabolic diseases, at the same time, to provide dietary and nutritional recommendations for using SCFAs as food ingredients to prevent metabolic diseases.
Collapse
Affiliation(s)
- Zhipeng Tao
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Nutrition Sciences, Texas Woman's University, Denton, Texas, USA
| | - Yao Wang
- Diabetes Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Nabizadeh E, Memar MY, Hamishehkar H, Ghanbari H, Kadkhoda H, Asnaashari S, Kafil HS, Varshochi M, Mostafazadeh M, Hosseinpour R, Ghotaslou R. Short-chain fatty acids profile in patients with SARS-CoV-2: A case-control study. Health Sci Rep 2023; 6:e1411. [PMID: 37425235 PMCID: PMC10323717 DOI: 10.1002/hsr2.1411] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/27/2023] [Accepted: 06/27/2023] [Indexed: 07/11/2023] Open
Abstract
BACKGROUND AND AIMS SARS-CoV-2, as a new pandemic disease, affected the world. Short-chain fatty acids (SCFAs) such as acetic, propionic, and butyric acids are the main metabolites of human gut microbiota. The positive effects of SCFAs have been shown in infections caused by respiratory syncytial virus, adenovirus, influenza, and rhinovirus. Therefore, this study aimed to evaluate the concentration of SCFAs in patients with SARS-CoV-2 compared with the healthy group. METHODS This research was designed based on a case and control study. Twenty healthy individuals as the control group and 20 persons admitted to the hospital with a positive test of coronavirus disease (COVID-19) real-time polymerase chain reaction were included in the study as the patient group from September 2021 to October 2021, in Tabriz, Iran. Stool specimens were collected from volunteers, and analysis of SCFAs was carried out by a high-performance liquid chromatography system. RESULTS The amount of acetic acid in the healthy group was 67.88 ± 23.09 μmol/g, while in the group of patients with COVID-19 was 37.04 ± 13.29 μmol/g. Therefore, the concentration of acetic acid in the patient group was significantly (p < 0.001) lower than in the healthy group. Propionic and butyric acid were present in a higher amount in the control group compared with the case group; however, this value was not statistically significant (p > 0.05). CONCLUSION This study showed that the concentration of acetic acid as the metabolite caused by gut microbiota is significantly disturbed in patients with COVID-19. Therefore, therapeutic interventions based on gut microbiota metabolites in future research may be effective against COVID-19.
Collapse
Affiliation(s)
- Edris Nabizadeh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | - Hamed Hamishehkar
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
| | - Hadi Ghanbari
- Department of Pharmacognosy, Faculty of PharmacyTabriz University of Medical SciencesTabrizIran
| | - Hiva Kadkhoda
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Drug Applied Research CenterTabriz University of Medical SciencesTabrizIran
| | - Solmaz Asnaashari
- Biotechnology Research CenterTabriz University of Medical SciencesTabrizIran
| | | | - Mojtaba Varshochi
- Infectious and Tropical Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | - Mostafa Mostafazadeh
- Department of Biochemistry and Clinical LaboratoriesTabriz University of Medical SciencesTabrizIran
| | - Rasoul Hosseinpour
- Infectious and Tropical Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| | - Reza Ghotaslou
- Infectious and Tropical Diseases Research CenterTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
15
|
Guiducci L, Nicolini G, Forini F. Dietary Patterns, Gut Microbiota Remodeling, and Cardiometabolic Disease. Metabolites 2023; 13:760. [PMID: 37367916 DOI: 10.3390/metabo13060760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023] Open
Abstract
The cardiovascular and metabolic disorders, collectively known as cardiometabolic disease (CMD), are high morbidity and mortality pathologies associated with lower quality of life and increasing health-care costs. The influence of the gut microbiota (GM) in dictating the interpersonal variability in CMD susceptibility, progression and treatment response is beginning to be deciphered, as is the mutualistic relation established between the GM and diet. In particular, dietary factors emerge as pivotal determinants shaping the architecture and function of resident microorganisms in the human gut. In turn, intestinal microbes influence the absorption, metabolism, and storage of ingested nutrients, with potentially profound effects on host physiology. Herein, we present an updated overview on major effects of dietary components on the GM, highlighting the beneficial and detrimental consequences of diet-microbiota crosstalk in the setting of CMD. We also discuss the promises and challenges of integrating microbiome data in dietary planning aimed at restraining CMD onset and progression with a more personalized nutritional approach.
Collapse
Affiliation(s)
- Letizia Guiducci
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| | | | - Francesca Forini
- CNR Institute of Clinical Physiology, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
16
|
Tang H, Zhan Z, Liu X, Zhang Y, Huang X, Xu M. Propionate reduces the viability of Salmonella enterica Serovar Typhi in macrophages by propionylation of PhoP K102. Microb Pathog 2023; 178:106078. [PMID: 36965832 DOI: 10.1016/j.micpath.2023.106078] [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: 02/06/2023] [Revised: 03/15/2023] [Accepted: 03/19/2023] [Indexed: 03/27/2023]
Abstract
Propionate, a major constituent of short chain fatty acids, has recently been reported to be involved in both prokaryotic and eukaryotic lysine propionylation (Kpr). However, the propionylation characteristics of the enteric pathogen Salmonella enterica serovar Typhi (S. Typhi) following invasion of the human gut under the influence of propionate, whether virulence is affected, and the underlying mechanisms are not yet known. In the present study, we report that propionate significantly reduces the viability of S. Typhi in macrophages through intra-macrophage survival assays. We also demonstrate that the concentration of propionate and the propionate metabolic intermediate propionyl coenzyme A can affect the level of modification of PhoP by propionylation, which is tightly linked to intracellular survival. By expressing and purifying PhoP protein in vitro and performing EMSA and protein phosphorylation analyses, We provide evidence that K102 of PhoP is modified by Kpr propionate, which regulates S. Typhi viability in macrophages by decreasing the phosphorylation and DNA-binding ability of PhoP. In conclusion, our study reveals a potential molecular mechanism by which propionate reduces the viability of S. Typhi in macrophages via Kpr.
Collapse
Affiliation(s)
- Hao Tang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China; Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ziyang Zhan
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiucheng Liu
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Zhang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xinxiang Huang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China; Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
17
|
Elsasser TH, Ma B, Ravel J, Kahl S, Gajer P, Cross A. Short-term feeding of defatted bovine colostrum mitigates inflammation in the gut via changes in metabolites and microbiota in a chicken animal model. Anim Microbiome 2023; 5:6. [PMID: 36703224 PMCID: PMC9878500 DOI: 10.1186/s42523-023-00225-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 01/10/2023] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Nondrug supplement strategies to improve gut health have largely focused on the effects of individual compounds to improve one aspect of gut homeostasis. However, there is no comprehensive assessment of the reproducible effects of oral, short-term, low-level colostrum supplementation on gut inflammation status that are specific to the ileum. Herein, a chicken animal model highly responsive to even mild gut inflammatory stimuli was employed to compare the outcomes of feeding a standard diet (CON) to those of CON supplemented with a centrifuge-defatted bovine colostrum (BC) or a nonfat dried milk (NFDM) control on the efficiency of nutrient use, ileal morphology, gut nitro-oxidative inflammation status, metabolites, and the composition of the microbiota. RESULTS A repeated design, iterative multiple regression model was developed to analyze how BC affected ileal digesta-associated anti-inflammatory metabolite abundance coincident with observed changes in the ileal microbiome, mitigation of epithelial inflammation, and ileal surface morphology. An improved whole body nutrient use efficiency in the BC group (v CON and NFDM) coincided with the observed increased ileum absorptive surface and reduced epithelial cell content of tyrosine-nitrated protein (NT, biomarker of nitro-oxidative inflammatory stress). Metabolome analysis revealed that anti-inflammatory metabolites were significantly greater in abundance in BC-fed animals. BC also had a beneficial BC impact on microbiota, particularly in promoting the presence of the bacterial types associated with eubiosis and the segmented filamentous bacteria, Candidatus Arthromitus. CONCLUSION The data suggest that an anti-inflammatory environment in the ileum was more evident in BC than in the other feeding groups and associated with an increased content of statistically definable groups of anti-inflammatory metabolites that appear to functionally link the observed interactions between the host's improved gut health with an observed increase in whole body nutrient use efficiency, beneficial changes in the microbiome and immunometabolism.
Collapse
Affiliation(s)
- Ted H. Elsasser
- grid.463419.d0000 0001 0946 3608Animal Biosciences and Biotechnology Laboratory, USA Department of Agriculture (USDA), Agricultural Research Service (ARS), Beltsville, MD 20705 USA
| | - Bing Ma
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Jacques Ravel
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Stanislaw Kahl
- grid.463419.d0000 0001 0946 3608Animal Biosciences and Biotechnology Laboratory, USA Department of Agriculture (USDA), Agricultural Research Service (ARS), Beltsville, MD 20705 USA
| | - Pawel Gajer
- grid.411024.20000 0001 2175 4264Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| | - Alan Cross
- grid.411024.20000 0001 2175 4264Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201 USA
| |
Collapse
|
18
|
Im J, Lee D, Park OJ, Natarajan S, Park J, Yun CH, Han SH. RNA-Seq-based transcriptome analysis of methicillin-resistant Staphylococcus aureus growth inhibition by propionate. Front Microbiol 2022; 13:1063650. [PMID: 36620009 PMCID: PMC9814166 DOI: 10.3389/fmicb.2022.1063650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus is a pathogen that causes a variety of infectious diseases such as pneumonia, endocarditis, and septic shock. Methicillin-resistant S. aureus (MRSA) evades virtually all available treatments, creating the need for an alternative control strategy. Although we previously demonstrated the inhibitory effect of sodium propionate (NaP) on MRSA, the regulatory mechanism of this effect remains unclear. In this study, we investigated the regulatory mechanism responsible for the inhibitory effect of NaP on MRSA using RNA-Seq analysis. Total RNAs were isolated from non-treated and 50 mM NaP-treated S. aureus USA300 for 3 h and transcriptional profiling was conducted by RNA-Seq analysis. A total of 171 differentially expressed genes (DEGs) with log2 fold change ≥2 and p < 0.05 was identified in the NaP treatment group compared with the control group. Among the 171 genes, 131 were up-regulated and 40 were down-regulated. Upon gene ontology (GO) annotation analysis, total 26 specific GO terms in "Biological process," "Molecular function," and "Cellular component" were identified in MRSA treated with NaP for 3 h. "Purine metabolism"; "riboflavin metabolism"; and "glycine, serine, and threonine metabolism" were identified as major altered metabolic pathways among the eight significantly enriched KEGG pathways in MRSA treated with NaP. Furthermore, the MRSA strains deficient in purF, ilvA, ribE, or ribA, which were the up-regulated DEGs in the metabolic pathways, were more susceptible to NaP than wild-type MRSA. Collectively, these results demonstrate that NaP attenuates MRSA growth by altering its metabolic pathways, suggesting that NaP can be used as a potential bacteriostatic agent for prevention of MRSA infection.
Collapse
Affiliation(s)
- Jintaek Im
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Dongwook Lee
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Ok-Jin Park
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, South Korea
| | | | | | - Cheol-Heui Yun
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, South Korea,Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang, South Korea
| | - Seung Hyun Han
- Department of Oral Microbiology and Immunology, and Dental Research Institute, School of Dentistry, Seoul National University, Seoul, South Korea,*Correspondence: Seung Hyun Han,
| |
Collapse
|
19
|
Butyrate Supplementation Exacerbates Myocardial and Immune Cell Mitochondrial Dysfunction in a Rat Model of Faecal Peritonitis. Life (Basel) 2022; 12:life12122034. [PMID: 36556399 PMCID: PMC9785094 DOI: 10.3390/life12122034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/27/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction and immune cell dysfunction are commonplace in sepsis and are associated with increased mortality risk. The short chain fatty acid, butyrate, is known to have anti-inflammatory effects and promote mitochondrial biogenesis. We therefore explored the immunometabolic effects of butyrate in an animal model of sepsis. Isolated healthy human volunteer peripheral mononuclear cells were stimulated with LPS in the presence of absence of butyrate, and released cytokines measured. Male Wistar rats housed in metabolic cages received either intravenous butyrate infusion or placebo commencing 6 h following faecal peritonitis induction. At 24 h, splenocytes were isolated for high-resolution respirometry, and measurement of mitochondrial membrane potential (MMP), reactive oxygen species (mtROS), and intracellular cytokines (TNF alpha, IL-10) using flow cytometry. Isolated splenocytes from septic and septic butyrate treated rats were stimulated with LPS for 18 h and the effects of butyrate on cytokine release assessed. Ex vivo, butyrate (1.8 mM) reduced LPS-induced TNF alpha (p = 0.019) and IL-10 (p = 0.001) release by human PBMCs. In septic animals butyrate infusion reduced the respiratory exchange ratio (p < 0.001), consistent with increased fat metabolism. This was associated with a reduction in cardiac output (p = 0.001), and increased lactate (p = 0.031) compared to placebo-treated septic animals (p < 0.05). Butyrate treatment was associated with a reduction in splenocyte basal respiration (p = 0.077), proton leak (p = 0.022), and non-mitochondrial respiration (p = 0.055), and an increase in MMP (p = 0.007) and mtROS (p = 0.027) compared to untreated septic animals. Splenocyte intracellular cytokines were unaffected by butyrate, although LPS-induced IL-10 release was impaired (p = 0.039). In summary, butyrate supplementation exacerbates myocardial and immune cell mitochondrial dysfunction in a rat model of faecal peritonitis.
Collapse
|
20
|
Hou J, Xiang J, Li D, Liu X, Pan W. Gut microbial response to host metabolic phenotypes. Front Nutr 2022; 9:1019430. [PMID: 36419554 PMCID: PMC9676441 DOI: 10.3389/fnut.2022.1019430] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/21/2022] [Indexed: 09/10/2023] Open
Abstract
A large number of studies have proved that biological metabolic phenotypes exist objectively and are gradually recognized by humans. Gut microbes affect the host's metabolic phenotype. They directly or indirectly participate in host metabolism, physiology and immunity through changes in population structure, metabolite differences, signal transduction and gene expression. Obtaining comprehensive information and specific identification factors associated with gut microbiota and host metabolic phenotypes has become the focus of research in the field of gut microbes, and it has become possible to find new and effective ways to prevent or treat host metabolic diseases. In the future, precise treatment of gut microbes will become one of the new therapeutic strategies. This article reviews the content of gut microbes and carbohydrate, amino acid, lipid and nucleic acid metabolic phenotypes, including metabolic intermediates, mechanisms of action, latest research findings and treatment strategies, which will help to understand the relationship between gut microbes and host metabolic phenotypes and the current research status.
Collapse
Affiliation(s)
- Jinliang Hou
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Jianguo Xiang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Deliang Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xinhua Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | | |
Collapse
|
21
|
Wang W, Jiang S, Xu C, Tang L, Liang Y, Zhao Y, Zhu G. Interactions between gut microbiota and Parkinson's disease: The role of microbiota-derived amino acid metabolism. Front Aging Neurosci 2022; 14:976316. [PMID: 36408101 PMCID: PMC9667037 DOI: 10.3389/fnagi.2022.976316] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/29/2022] [Indexed: 11/05/2022] Open
Abstract
Non-motor symptoms (NMS) of Parkinson's disease (PD), such as constipation, sleep disorders, and olfactory deficits, may emerge up to 20 years earlier than motor symptoms. A series of evidence indicates that the pathology of PD may occur from the gastrointestinal tract to the brain. Numerous studies support that the gut microbiota communicates with the brain through the immune system, special amino acid metabolism, and the nervous system in PD. Recently, there is growing recognition that the gut microbiota plays a vital role in the modulation of multiple neurochemical pathways via the “gut microbiota-brain axis” (GMBA). Many gut microbiota metabolites, such as fatty acids, amino acids, and bile acids, convey signaling functions as they mediate the crosstalk between gut microbiota and host physiology. Amino acids' abundance and species alteration, including glutamate and tryptophan, may disturb the signaling transmission between nerve cells and disrupt the normal basal ganglia function in PD. Specific amino acids and their receptors are considered new potential targets for ameliorating PD. The present study aimed to systematically summarize all available evidence on the gut microbiota-derived amino acid metabolism alterations associated with PD.
Collapse
Affiliation(s)
- Wang Wang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shujun Jiang
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengcheng Xu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lili Tang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Liang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Yang Zhao
| | - Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Guoxue Zhu
| |
Collapse
|
22
|
Tang H, Zhan Z, Zhang Y, Huang X. Propionylation of lysine, a new mechanism of short-chain fatty acids affecting bacterial virulence. Am J Transl Res 2022; 14:5773-5784. [PMID: 36105019 PMCID: PMC9452321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/19/2022] [Indexed: 06/15/2023]
Abstract
Propionic acid (PA) is a major component of short-chain fatty acids produced by Bacteroidetes spp. Lysine propionylation is a novel type of protein regulatory posttranslational modification that is widespread in prokaryotes and eukaryotes, as well as in cellular processes, it affects DNA binding affinity, protein stability, and enzyme activity. In this review of published literature, we provide evidence that the level of propionyl modification is influenced by the concentration of PA and the PA metabolic intermediate (propionyl-CoA) and discuss the possibility of PA affecting enteropathogenic bacterial virulence. The understanding of propionyl modification is helpful to better understand the mechanism of PA-producing Bacteroidetes affecting the virulence of pathogenic intestinal bacteria. It may provide novel choices for the prevention and treatment of pathogenic intestinal bacteria.
Collapse
Affiliation(s)
- Hao Tang
- Department of Biochemistry & Molecular Biology, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Ziyang Zhan
- Department of Biochemistry & Molecular Biology, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Ying Zhang
- Department of Biochemistry & Molecular Biology, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| | - Xinxiang Huang
- Department of Biochemistry & Molecular Biology, School of Medicine, Jiangsu University 301 Xuefu Road, Zhenjiang 212013, Jiangsu, China
| |
Collapse
|
23
|
Winiarska-Mieczan A, Tomaszewska E, Donaldson J, Jachimowicz K. The Role of Nutritional Factors in the Modulation of the Composition of the Gut Microbiota in People with Autoimmune Diabetes. Nutrients 2022; 14:2498. [PMID: 35745227 PMCID: PMC9227140 DOI: 10.3390/nu14122498] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a disease marked by oxidative stress, chronic inflammation, and the presence of autoantibodies. The gut microbiota has been shown to be involved in the alleviation of oxidative stress and inflammation as well as strengthening immunity, thus its' possible involvement in the pathogenesis of T1DM has been highlighted. The goal of the present study is to analyze information on the relationship between the structure of the intestinal microbiome and the occurrence of T1DM. The modification of the intestinal microbiota can increase the proportion of SCFA-producing bacteria, which could in turn be effective in the prevention and/or treatment of T1DM. The increased daily intake of soluble and non-soluble fibers, as well as the inclusion of pro-biotics, prebiotics, herbs, spices, and teas that are sources of phytobiotics, in the diet, could be important in improving the composition and activity of the microbiota and thus in the prevention of metabolic disorders. Understanding how the microbiota interacts with immune cells to create immune tolerance could enable the development of new therapeutic strategies for T1DM and improve the quality of life of people with T1DM.
Collapse
Affiliation(s)
- Anna Winiarska-Mieczan
- Department of Bromatology and Nutrition Physiology, Institute of Animal Nutrition and Bromatology, University of Life Sciences in Lublin, Akademicka St. 13, 20-950 Lublin, Poland;
| | - Ewa Tomaszewska
- Department of Animal Physiology, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Akademicka St. 12, 20-950 Lublin, Poland
| | - Janine Donaldson
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg 2193, South Africa;
| | - Karolina Jachimowicz
- Department of Bromatology and Nutrition Physiology, Institute of Animal Nutrition and Bromatology, University of Life Sciences in Lublin, Akademicka St. 13, 20-950 Lublin, Poland;
| |
Collapse
|
24
|
Wang Y, Nan X, Zhao Y, Jiang L, Wang H, Zhang F, Hua D, Liu J, Yang L, Yao J, Xiong B. Changes in the Profile of Fecal Microbiota and Metabolites as Well as Serum Metabolites and Proteome After Dietary Inulin Supplementation in Dairy Cows With Subclinical Mastitis. Front Microbiol 2022; 13:809139. [PMID: 35479637 PMCID: PMC9037088 DOI: 10.3389/fmicb.2022.809139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The occurrence and development of mastitis is linked to dysbiostic gastrointestinal microbiota. Inulin is a dietary prebiotic that improves the profile of intestinal flora. Our previous study showed that inulin supplementation could improve the ruminal microbes of subclinical mastitis (SCM) cows. The current study attempted to further investigate the response of hindgut (fecal) microbiome and metabolites, serum metabolism, and protein expression to inulin in the in SCM cows. Different levels of inulin (0, 100, 200, 300, and 400 g/day per cow) were supplemented in SCM cows. Compared with control group, Bacteroides and Bifidobacteria were increased, and Paeniclostridium, Ruminococcaceae, Coprococcus, and Clostridia were decreased in the feces of inulin groups, and accompanied with elevated propionate and butyrate concentrations, while secondary bile acid (SBA) metabolites were increased and proinflammatory lipid oxidation products were dropped in both feces and serum. In serum, inulin intake suppressed the levels of triglyceride (TG) and low-density lipoprotein (LDL). Serum proteome analysis found that CD44 antigen, phosphatidylinositol-glycan-specific phospholipase D, apolipoprotein A-II, and superoxide dismutase [Cu-Zn] were upregulated, while cathelicidin-1, haptoglobin, serpin A3, inter-alpha-trypsin inhibitor heavy chain H4 were downregulated in inulin groups. These findings suggested further evidence for inulin supplementation in amelioration of inflammatory symptoms in SCM cows, which might provide alternative treatment for mastitis.
Collapse
Affiliation(s)
- Yue Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yiguang Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Linshu Jiang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing, China
| | - Hui Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Fan Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Dengke Hua
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Jun Liu
- Langfang Academy of Agriculture and Forestry, Langfang, China
| | - Liang Yang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
25
|
Korsten SGPJ, Peracic L, van Groeningen LMB, Diks MAP, Vromans H, Garssen J, Willemsen LEM. Butyrate Prevents Induction of CXCL10 and Non-Canonical IRF9 Expression by Activated Human Intestinal Epithelial Cells via HDAC Inhibition. Int J Mol Sci 2022; 23:ijms23073980. [PMID: 35409339 PMCID: PMC8999521 DOI: 10.3390/ijms23073980] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/23/2022] [Accepted: 03/31/2022] [Indexed: 12/11/2022] Open
Abstract
Non-communicable diseases are increasing and have an underlying low-grade inflammation in common, which may affect gut health. To maintain intestinal homeostasis, unwanted epithelial activation needs to be avoided. This study compared the efficacy of butyrate, propionate and acetate to suppress IFN-γ+/−TNF-α induced intestinal epithelial activation in association with their HDAC inhibitory capacity, while studying the canonical and non-canonical STAT1 pathway. HT-29 were activated with IFN-γ+/−TNF-α and treated with short chain fatty acids (SCFAs) or histone deacetylase (HDAC) inhibitors. CXCL10 release and protein and mRNA expression of proteins involved in the STAT1 pathway were determined. All SCFAs dose-dependently inhibited CXCL10 release of the cells after activation with IFN-γ or IFN-γ+TNF-α. Butyrate was the most effective, completely preventing CXCL10 induction. Butyrate did not affect phosphorylated STAT1, nor phosphorylated NFκB p65, but inhibited IRF9 and phosphorylated JAK2 protein expression in activated cells. Additionally, butyrate inhibited CXCL10, SOCS1, JAK2 and IRF9 mRNA in activated cells. The effect of butyrate was mimicked by class I HDAC inhibitors and a general HDAC inhibitor Trichostatin A. Butyrate is the most potent inhibitor of CXCL10 release compared to other SCFAs and acts via HDAC inhibition. This causes downregulation of CXCL10, JAK2 and IRF9 genes, resulting in a decreased IRF9 protein expression which inhibits the non-canonical pathway and CXCL10 transcription.
Collapse
Affiliation(s)
- Sandra G. P. J. Korsten
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
- Tiofarma B.V., 3261 ME Oud-Beijerland, The Netherlands;
- Correspondence: (S.G.P.J.K.); (L.E.M.W.)
| | - Laura Peracic
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
| | - Luka M. B. van Groeningen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
| | - Mara A. P. Diks
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
| | - Herman Vromans
- Tiofarma B.V., 3261 ME Oud-Beijerland, The Netherlands;
- Division of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
- Nutricia Research B.V., 3584 CT Utrecht, The Netherlands
| | - Linette E. M. Willemsen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands; (L.P.); (L.M.B.v.G.); (M.A.P.D.); (J.G.)
- Correspondence: (S.G.P.J.K.); (L.E.M.W.)
| |
Collapse
|
26
|
Song Z, Luo W, Huang B, Cao Y, Jiang R. A new predictive model for the concurrent risk of diabetic retinopathy in type 2 diabetes patients and the effect of metformin on amino acids. Front Endocrinol (Lausanne) 2022; 13:985776. [PMID: 36060930 PMCID: PMC9434554 DOI: 10.3389/fendo.2022.985776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE This study established a model to predict the risk of diabetic retinopathy (DR) with amino acids selected by partial least squares (PLS) method, and evaluated the effect of metformin on the effect of amino acids on DR in the model. METHODS In Jinzhou, Liaoning Province, China, we retrieved 1031 patients with type 2 diabetes (T2D) from the First Affiliated Hospital of Liaoning Medical University. After sorting the amino acids using the PLS method, the top 10 amino acids were included in the model. Multivariate logistic regression was used to analyze the relationship between different amino acids and DR. And then the effects of metformin on amino acids were explored through interaction. Finally, Spearman's rank correlation analysis was used to analyze the correlation between different amino acids. RESULTS After sorting by PLS, Gly, Pro, Leu, Lyr, Glu, Phe, Tyr, His, Val and Ser were finally included in the DR risk prediction model. The predictive model after adding amino acids was statistically different from the model that only included traditional risk factors (p=0.001). Metformin had a significant effect on the relationship between DR and 7 amino acids (Gly, Glu, Phe, Tyr, His, Val, Ser, p<0.05), and the population who are not using metformin and have high levels of Glu (OR: 0.44, 95%CI: 0.27-0.71) had an additive protection effect for the occurrence of DR. And the similar results can be seen in high levels of Gly (OR: 0.46, 95%CI: 0.29-0.75), Leu (OR: 0.48, 95%CI: 0.29-0.8), His (OR: 0.46, 95%CI: 0.29-0.75), Phe (OR: 0.24, 95%CI: 0.14-0.42) and Tyr (OR: 0.41, 95%CI: 0.24 -0.68) in population who are not using metformin. CONCLUSIONS We established a prediction model of DR by amino acids and found that the use of metformin reduced the protective effect of amino acids on DR developing, suggesting that amino acids as biomarkers for predicting DR would be affected by metformin use.
Collapse
Affiliation(s)
- Zicheng Song
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Weiming Luo
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Tianjin Medical University, Tianjin, China
| | - Bing Huang
- Research Department, Dalian Innovation Center of Laboratory Medicine Mass Spectrometry Technology, Dalian, China
- Research Department, Clinical Mass Spectrometry Profession Technology Innovation Center of Liaoning Province, Jinzhou, China
- Research Department, Dalian Laboratory Medicine Mass Spectrometry Technology Development Innovation Team, Dalian, China
| | - Yunfeng Cao
- Department of Scientific Research, Shanghai Institute of Planned Parenthood Research, Shanghai, China
- Dalian Institute of Chemical Physics. Chinese Academy of Sciences, Dalian, China
- *Correspondence: Yunfeng Cao, ; Rongzhen Jiang,
| | - Rongzhen Jiang
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
- *Correspondence: Yunfeng Cao, ; Rongzhen Jiang,
| |
Collapse
|
27
|
Begum N, Harzandi A, Lee S, Uhlen M, Moyes DL, Shoaie S. Host-mycobiome metabolic interactions in health and disease. Gut Microbes 2022; 14:2121576. [PMID: 36151873 PMCID: PMC9519009 DOI: 10.1080/19490976.2022.2121576] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/31/2022] [Accepted: 08/31/2022] [Indexed: 02/04/2023] Open
Abstract
Fungal communities (mycobiome) have an important role in sustaining the resilience of complex microbial communities and maintenance of homeostasis. The mycobiome remains relatively unexplored compared to the bacteriome despite increasing evidence highlighting their contribution to host-microbiome interactions in health and disease. Despite being a small proportion of the total species, fungi constitute a large proportion of the biomass within the human microbiome and thus serve as a potential target for metabolic reprogramming in pathogenesis and disease mechanism. Metabolites produced by fungi shape host niches, induce immune tolerance and changes in their levels prelude changes associated with metabolic diseases and cancer. Given the complexity of microbial interactions, studying the metabolic interplay of the mycobiome with both host and microbiome is a demanding but crucial task. However, genome-scale modelling and synthetic biology can provide an integrative platform that allows elucidation of the multifaceted interactions between mycobiome, microbiome and host. The inferences gained from understanding mycobiome interplay with other organisms can delineate the key role of the mycobiome in pathophysiology and reveal its role in human disease.
Collapse
Affiliation(s)
- Neelu Begum
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Azadeh Harzandi
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Sunjae Lee
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Mathias Uhlen
- Science for Life Laboratory, KTH–Royal Institute of Technology, Stockholm, Sweden
| | - David L. Moyes
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
| | - Saeed Shoaie
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London, UK
- Science for Life Laboratory, KTH–Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
28
|
Gut microbiota and metabolic changes towards improved gut health with supplementation of Woodfordia fruticosa, a medicinal plant: An in vitro study. INNOV FOOD SCI EMERG 2022. [DOI: 10.1016/j.ifset.2021.102896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Dietary Supplementation of Inulin Ameliorates Subclinical Mastitis via Regulation of Rumen Microbial Community and Metabolites in Dairy Cows. Microbiol Spectr 2021; 9:e0010521. [PMID: 34494854 PMCID: PMC8557905 DOI: 10.1128/spectrum.00105-21] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Subclinical mastitis (SCM) is one of the highly infectious diseases in dairy cows with the characteristics of high incidence and nonvisible clinical symptoms. The gastrointestinal microbiota is closely related to mastitis. Inulin is a prebiotic fiber with functions in improving intestinal microbial communities and enhancing the host’s immunity. However, the impact of dietary inulin on the rumen inner environment remains unknown. The current study investigated whether inulin could relieve SCM by affecting the profiles of ruminal bacterial and metabolites in dairy cows. Inulin inclusion rates were 0, 100, 200, 300, and 400 g/day per cow, respectively. Inulin increased milk yield, milk protein, and lactose and reduced the somatic cell counts (SCC) in milk. In serum, the concentration of proinflammatory cytokines, such as interleukin-6 (IL-6), IL-8, tumor necrosis factor α (TNF-α), and malondialdehyde (MDA) were decreased, and IL-4 and superoxide dismutase (SOD) were increased. Meanwhile, inulin increased the concentration of propionate, butyrate, and lactic acid (LA), while it decreased NH3-N in rumen. The propionate- and butyrate-producing bacteria (e.g., Prevotella and Butyrivibrio) and several beneficial commensal bacteria (e.g., Muribaculaceae and Bifidobacterium) as well as metabolites related to energy and amino acid metabolism (e.g., melibiose and l-glutamate) were increased. However, several proinflammatory bacteria (e.g., Clostridia UCG-014, Streptococcus, and Escherichia-Shigella) were decreased, accompanied by the downregulation of lipid proinflammatory metabolites, for example, ceramide(d18:0/15:0) [Cer(d18:0/15:0)] and 17-phenyl-18,19,20-trinor-prostaglandin E2. In the current study, the above indicators showed the best response in the 300 g/day inulin group. Overall, dietary supplementation of inulin could alleviate inflammatory responses in cows with SCM through improving the rumen inner environment. IMPORTANCE The correlation between mastitis and the gastrointestinal microbiome in dairy cows has been demonstrated. Regulating the profile of rumen microorganisms may contribute to remission of subclinical mastitis (SCM). Supplementation of inulin in the diets of cows with SCM could increase the abundance of short-chain fatty acid (SCFA)-producing bacteria and beneficial commensal bacteria in rumen and meanwhile the levels of amino acids and energy metabolism. Conversely, the abundance of ruminal bacteria and metabolites with proinflammatory effects were decreased. Our study suggests that the improvement of the rumen internal environment by inulin supplementation could ameliorate inflammatory responses during SCM in dairy cows and thus improve lactation performance and milk quality. Our results provide a theoretical basis for regulation measures of SCM in dairy cows.
Collapse
|
30
|
Kumar NG, Contaifer D, Wijesinghe DS, Jefferson KK. Staphylococcus aureus Lipase 3 (SAL3) is a surface-associated lipase that hydrolyzes short chain fatty acids. PLoS One 2021; 16:e0258106. [PMID: 34618844 PMCID: PMC8496776 DOI: 10.1371/journal.pone.0258106] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/20/2021] [Indexed: 11/18/2022] Open
Abstract
Bacterial lipases play important roles during infection. The Staphylococcus aureus genome contains several genes that encode well-characterized lipases and several genes predicted to encode lipases or esterases for which the function has not yet been established. In this study, we sought to define the function of an uncharacterized S. aureus protein, and we propose the annotation S. aureus lipase 3 (SAL3) (SAUSA300_0641). We confirmed that SAL3 is a lipase and that it is surface associated and secreted through an unknown mechanism. We determined that SAL3 specifically hydrolyzes short chain (4-carbon and fewer) fatty acids and specifically binds negatively charged lipids including phosphatidic acid, phosphatidylinositol phosphate, and phosphatidylglycerol, which is the most abundant lipid in the staphylococcal cell membrane. Mutating the catalytic triad S66-A, D167-A, S168-A, and H301-A in the recombinant protein abolished lipase activity without altering binding to host lipid substrates. Taken together we report the discovery of a novel lipase from S. aureus specific to short chain fatty acids with yet to be determined roles in host pathogen interactions.
Collapse
Affiliation(s)
- Naren Gajenthra Kumar
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Daniel Contaifer
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Dayanjan S. Wijesinghe
- Department of Pharmacotherapy and Outcomes Sciences, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Kimberly K. Jefferson
- Department of Microbiology and Immunology, School of Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
- * E-mail:
| |
Collapse
|
31
|
Théroude C, Reverte M, Heinonen T, Ciarlo E, Schrijver IT, Antonakos N, Maillard N, Pralong F, Le Roy D, Roger T. Trained Immunity Confers Prolonged Protection From Listeriosis. Front Immunol 2021; 12:723393. [PMID: 34603295 PMCID: PMC8484647 DOI: 10.3389/fimmu.2021.723393] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/06/2021] [Indexed: 12/11/2022] Open
Abstract
Trained immunity refers to the ability of the innate immune system exposed to a first challenge to provide an enhanced response to a secondary homologous or heterologous challenge. We reported that training induced with β-glucan one week before infection confers protection against a broad-spectrum of lethal bacterial infections. Whether this protection persists over time is unknown. To tackle this question, we analyzed the immune status and the response to Listeria monocytogenes (L. monocytogenes) of mice trained 9 weeks before analysis. The induction of trained immunity increased bone marrow myelopoiesis and blood counts of Ly6Chigh inflammatory monocytes and polymorphonuclear neutrophils (PMNs). Ex vivo, whole blood, PMNs and monocytes from trained mice produced increased levels of cytokines in response to microbial products and limited the growth of L. monocytogenes. In vivo, following challenge with L. monocytogenes, peripheral blood leukocytes were massively depleted in control mice but largely preserved in trained mice. PMNs were reduced also in the spleen from control mice, and increased in the spleen of trained mice. In transwell experiments, PMNs from trained mice showed increased spontaneous migration and CXCL2/MIP2α-induced chemotaxis, suggesting that training promotes the migration of PMNs in peripheral organs targeted by L. monocytogenes. Trained PMNs and monocytes had higher glycolytic activity and mitochondrial respiration than control cells when exposed to L. monocytogenes. Bacterial burden and dissemination in blood, spleen and liver as well as systemic cytokines and inflammation (multiplex bead assay and bioluminescence imaging) were reduced in trained mice. In full agreement with these results, mice trained 9 weeks before infection were powerfully protected from lethal listeriosis. Altogether, these data suggest that training increases the generation and the antimicrobial activity of PMNs and monocytes, which may confer prolonged protection from lethal bacterial infection.
Collapse
Affiliation(s)
- Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Marta Reverte
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Tytti Heinonen
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Eleonora Ciarlo
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Irene T Schrijver
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Nikolaos Antonakos
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Nicolas Maillard
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Florian Pralong
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
32
|
Herderschee J, Heinonen T, Fenwick C, Schrijver IT, Ohmiti K, Moradpour D, Cavassini M, Pantaleo G, Roger T, Calandra T. High-dimensional immune phenotyping of blood cells by mass cytometry in patients infected with hepatitis C virus. Clin Microbiol Infect 2021; 28:611.e1-611.e7. [PMID: 34474121 DOI: 10.1016/j.cmi.2021.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Chronic hepatitis C virus (HCV) infection affects the immune system. Whether elimination of HCV with direct-acting antivirals (DAA) restores immunity is unclear. We used mass cytometry to get a broad and in-depth assessment of blood cell populations of patients with chronic HCV before and after DAA therapy. METHODS Before and 12 weeks after sustained virological response (SVR12) to DAA therapy, 22 cell populations were analysed by mass cytometry in blood collected from ten healthy control individuals and 20 HCV-infected patients with (ten patients) or without (ten patients) human immunodeficiency virus (HIV) infection. RESULTS HCV infection altered the frequency of 14/22 (64%) blood cell populations. At baseline, the frequencies (median, interquartile range (IQR); control, HCV, HCV/HIV) of intermediate monocytes (1.2, IQR 0.47-1.46; 1.76, IQR 0.83-2.66; 0.78, IQR 0.28-1.77), non-classical monocytes (1.11, IQR 0.49-1.26; 0.9, IQR 0.18-0.99; 0.54, IQR 0.28-1.77), conventional dendritic cells type 2 (0.55, IQR 0.35-0.59; 0.31, IQR 0.16-0.38; 0.19, IQR 0.11-0.36) and CD56dim natural killer cells (8.08, IQR 5.34-9.79; 4.72, IQR 2.59-6.05) 3.61, IQR 2.98-5.07) were reduced by 35% to 65%, particularly in HCV/HIV co-infected patients. In contrast, activated double-negative T cells (0.07, IQR 0.06-0.10; 0.10, IQR 0.09-0.19; 0.19, IQR 0.12-0.25), activated CD4 T cells (0.28, IQR 0.21-0.36; 0.56, IQR 0.33-0.77; 0.40, IQR 0.22-0.53) and activated CD8 T cells (0.23, IQR 0.14-0.42; 0.74, IQR 0.30-1.65; 0.80, IQR 0.58-1.16) were increased 1.4 to 3.5 times. Upon stimulation with Toll-like receptor ligands, the expression of cytokines was up-regulated in 7/9 (78%) and 17/19 (89%) of the conditions in HCV- and HCV/HIV-infected patients, respectively. Most alterations persisted at SVR12. CONCLUSIONS Chronic HCV and HCV/HIV infections induce profound and durable perturbations of innate and adaptive immune homeostasis.
Collapse
Affiliation(s)
- Jacobus Herderschee
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Tytti Heinonen
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Craig Fenwick
- Division of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Irene T Schrijver
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Khalid Ohmiti
- Division of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Darius Moradpour
- Division of Gastroenterology and Hepatology, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Matthias Cavassini
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland; Swiss Vaccine Research Institute, Lausanne, Switzerland
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Thierry Calandra
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | | |
Collapse
|
33
|
Machado MG, Sencio V, Trottein F. Short-Chain Fatty Acids as a Potential Treatment for Infections: a Closer Look at the Lungs. Infect Immun 2021; 89:e0018821. [PMID: 34097474 PMCID: PMC8370681 DOI: 10.1128/iai.00188-21] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by the gut microbiota via the fermentation of complex carbohydrates and fibers. Evidence suggests that SCFAs play a role in the control of infections through direct action both on microorganisms and on host signaling. This review summarizes the main microbicidal effects of SCFAs and discusses studies highlighting the effect of SCFAs in the virulence and viability of microorganisms. We also describe the diverse and complex modes of action of the SCFAs on the immune system in the face of infections with a specific focus on bacterial and viral respiratory infections. A growing body of evidence suggests that SCFAs protect against lung infections. Finally, we present potential strategies that may be leveraged to exploit the biological properties of SCFAs for increasing effectiveness and optimizing patient benefits.
Collapse
Affiliation(s)
- Marina Gomes Machado
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, ICB, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, INSERM U1019, CNRS UMR 9017, University of Lille, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
34
|
Tobin D, Vige R, Calder PC. Review: The Nutritional Management of Multiple Sclerosis With Propionate. Front Immunol 2021; 12:676016. [PMID: 34394076 PMCID: PMC8355737 DOI: 10.3389/fimmu.2021.676016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/14/2021] [Indexed: 12/13/2022] Open
Abstract
Over the last 15 years there has been an accumulation of data supporting the concept of a gut-brain axis whereby dysbiosis of the gut microbiota can impact neurological function. Such dysbiosis has been suggested as a possible environmental exposure triggering multiple sclerosis (MS). Dysbiosis has been consistently shown to result in a reduction in short-chain fatty acid (SCFA) producing bacteria and a reduction in stool and plasma levels of propionate has been shown for MS patients independent of disease stage and in different geographies. A wealth of evidence supports the action of propionate on T-cell activity, resulting in decreased T-helper cell 1 (Th1) and T-helper cell 17 (Th17) numbers/activity and increased regulatory T cell (Treg cell) numbers/activity and an overall anti-inflammatory profile. These different T-cell populations play various roles in the pathophysiology of MS. A recent clinical study in MS patients demonstrated that supplementation of propionate reduces the annual relapse rate and slows disease progression. This review discusses this data and the relevant mechanistic background and discusses whether taming of the overactive immune system in MS is likely to allow easier bacterial and viral infection.
Collapse
Affiliation(s)
| | | | - Philip C Calder
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, United Kingdom.,NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| |
Collapse
|
35
|
Ives A, Le Roy D, Théroude C, Bernhagen J, Roger T, Calandra T. Macrophage migration inhibitory factor promotes the migration of dendritic cells through CD74 and the activation of the Src/PI3K/myosin II pathway. FASEB J 2021; 35:e21418. [PMID: 33774873 DOI: 10.1096/fj.202001605r] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/18/2022]
Abstract
Constitutively expressed by innate immune cells, the cytokine macrophage migration inhibitory factor (MIF) initiates host immune responses and drives pathogenic responses in infectious, inflammatory, and autoimmune diseases. Dendritic cells (DCs) express high levels of MIF, but the role of MIF in DC function remains poorly characterized. As migration is critical for DC immune surveillance, we investigated whether MIF promoted the migration of DCs. In classical transwell experiments, MIF-/- bone marrow-derived DCs (BMDCs) or MIF+/+ BMDCs treated with ISO-1, an inhibitor of MIF, showed markedly reduced spontaneous migration and chemotaxis. CD74-/- BMDCs that are deficient in the ligand-binding component of the cognate MIF receptor exhibited a migration defect similar to that of MIF-/- BMDCs. Adoptive transfer experiments of LPS-matured MIF+/+ and MIF-/- and of CD74+/+ and CD74-/- BMDCs injected into the hind footpads of homologous or heterologous mice showed that the autocrine and paracrine MIF activity acting via CD74 contributed to the recruitment of DCs to the draining lymph nodes. Mechanistically, MIF activated the Src/PI3K signaling pathway and myosin II complexes, which were required for the migration of BMDCs. Altogether, these data show that the cytokine MIF exerts chemokine-like activity for DC motility and trafficking.
Collapse
Affiliation(s)
- Annette Ives
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Jürgen Bernhagen
- Chair of Vascular Biology, Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München (KUM), Ludwig-Maximilians-University (LMU), Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Thierry Calandra
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
36
|
Xia D, Yang L, Li Y, Chen J, Zhang X, Wang H, Zhai S, Jiang X, Meca G, Wang S, Huang L, Zhu S, Fu Y, Ma W, Zhu Y, Ye H, Wang W. Melatonin alleviates Ochratoxin A-induced liver inflammation involved intestinal microbiota homeostasis and microbiota-independent manner. JOURNAL OF HAZARDOUS MATERIALS 2021; 413:125239. [PMID: 33582472 DOI: 10.1016/j.jhazmat.2021.125239] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 06/12/2023]
Abstract
Melatonin (MEL) shows an anti-inflammatory effect and regulates intestinal microbiota communities in animals and humans; Ochratoxin A (OTA) induces liver inflammation through intestinal microbiota. However, it remains to know whether MEL alleviates the liver inflammation induced by OTA. In this study, MEL reversed various adverse effects induced by OTA. MEL recovered the swarming and motility of intestinal microbiota, decreased the accumulation of lipopolysaccharide (LPS), enhanced the tight junction proteins of jejunum and cecum segments; ultimately alleviated OTA-induced liver inflammation in ducks. However, it is worth noting that MEL still had positive effects on the OTA-exposed ducks after antibiotic treatment. These results suggest that both the maintenance of intestinal microbiota homeostasis and intestinal microbiota-independent manner involved the MEL anti-inflammatory function in OTA-induced liver inflammation. MEL represent a promising protective approach for OTA, even other mycotoxins.
Collapse
Affiliation(s)
- Daiyang Xia
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Lin Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yu Li
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jianying Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiufen Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Heng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shuangshuang Zhai
- College of Animal Science, Yangtze University, Jingzhou 434000, China
| | - Xianzhi Jiang
- Microbiome Research Center, Moon (Guangzhou) Biotech Co. Ltd. Guangzhou 510535, China
| | - Giuseppe Meca
- Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, Burjassot 46100, Spain
| | | | - Liang Huang
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shanshan Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yang Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Weiqing Ma
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Yongwen Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hui Ye
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Wence Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition and Regulation, College of Animal Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
37
|
Haque M, Koski KG, Scott ME. A gastrointestinal nematode in pregnant and lactating mice alters maternal and neonatal microbiomes. Int J Parasitol 2021; 51:945-957. [PMID: 34081970 DOI: 10.1016/j.ijpara.2021.03.008] [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: 08/19/2020] [Revised: 03/24/2021] [Accepted: 03/28/2021] [Indexed: 11/29/2022]
Abstract
The maternal microbiome is understood to be the principal source of the neonatal microbiome but the consequences of intestinal nematodes on pregnant and lactating mothers and implications for the neonatal microbiome are unknown. Using pregnant CD1 mice infected with Heligmosomoides bakeri, we investigated the microbiomes in maternal tissues (intestine, vagina, and milk) and in the neonatal stomach using MiSeq sequencing of bacterial 16S rRNA genes. Our first hypothesis was that maternal nematode infection altered the maternal intestinal, vaginal, and milk microbiomes and associated metabolic pathways. Maternal nematode infection was associated with increased beta-diversity and abundance of fermenting bacteria as well as Lactobacillus in the maternal caecum 2 days after parturition, together with down-regulated carbohydrate, amino acid and vitamin biosynthesis pathways. Maternal nematode infection did not alter the vaginal or milk microbiomes. Our second hypothesis was that maternal infection would shape colonization of the neonatal microbiome. Although the pup stomach microbiome was similar to that of the maternal vaginal microbiome, pups of infected dams had higher beta-diversity at day 2, and a dramatic expansion in the abundance of Lactobacillus between days 2 and 7 compared with pups nursing uninfected dams. Our third hypothesis that maternal nematode infection altered the composition of neonatal microbiomes was confirmed as we observed up-regulation of several putatively beneficial microbial pathways associated with synthesis of essential and branched-chain amino acids, vitamins, and short-chain fatty acids. We believe this is the first study to show that a nematode living in the maternal intestine is associated with altered composition and function of the neonatal microbiome.
Collapse
Affiliation(s)
- Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21 111 Lakeshore Road, Ste-Anne-de-Bellevue, Québec H9X 3V9, Canada.
| |
Collapse
|
38
|
Abdallah A, Elemba E, Zhong Q, Sun Z. Gastrointestinal Interaction between Dietary Amino Acids and Gut Microbiota: With Special Emphasis on Host Nutrition. Curr Protein Pept Sci 2021; 21:785-798. [PMID: 32048965 DOI: 10.2174/1389203721666200212095503] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/25/2019] [Accepted: 07/31/2019] [Indexed: 12/31/2022]
Abstract
The gastrointestinal tract (GIT) of humans and animals is host to a complex community of different microorganisms whose activities significantly influence host nutrition and health through enhanced metabolic capabilities, protection against pathogens, and regulation of the gastrointestinal development and immune system. New molecular technologies and concepts have revealed distinct interactions between the gut microbiota and dietary amino acids (AAs) especially in relation to AA metabolism and utilization in resident bacteria in the digestive tract, and these interactions may play significant roles in host nutrition and health as well as the efficiency of dietary AA supplementation. After the protein is digested and AAs and peptides are absorbed in the small intestine, significant levels of endogenous and exogenous nitrogenous compounds enter the large intestine through the ileocaecal junction. Once they move in the colonic lumen, these compounds are not markedly absorbed by the large intestinal mucosa, but undergo intense proteolysis by colonic microbiota leading to the release of peptides and AAs and result in the production of numerous bacterial metabolites such as ammonia, amines, short-chain fatty acids (SCFAs), branched-chain fatty acids (BCFAs), hydrogen sulfide, organic acids, and phenols. These metabolites influence various signaling pathways in epithelial cells, regulate the mucosal immune system in the host, and modulate gene expression of bacteria which results in the synthesis of enzymes associated with AA metabolism. This review aims to summarize the current literature relating to how the interactions between dietary amino acids and gut microbiota may promote host nutrition and health.
Collapse
Affiliation(s)
- Abedin Abdallah
- Key laboratory of Straw Biology and Utilization (The Ministry of Education), Key Lab of Animal Nutrition and Feed
Science, Key Lab of Animal Production, Product Quality and Security, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Evera Elemba
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Qingzhen Zhong
- Key laboratory of Straw Biology and Utilization (The Ministry of Education), Key Lab of Animal Nutrition and Feed
Science, Key Lab of Animal Production, Product Quality and Security, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zewei Sun
- Key laboratory of Straw Biology and Utilization (The Ministry of Education), Key Lab of Animal Nutrition and Feed
Science, Key Lab of Animal Production, Product Quality and Security, College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
39
|
Fowler EC, Poudel P, White B, St-Pierre B, Brown M. Effects of a Bioprocessed Soybean Meal Ingredient on the Intestinal Microbiota of Hybrid Striped Bass, Morone chrysops x M. saxatilis. Microorganisms 2021; 9:microorganisms9051032. [PMID: 34064862 PMCID: PMC8151853 DOI: 10.3390/microorganisms9051032] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 12/12/2022] Open
Abstract
The hybrid striped bass (Morone chrysops x M. saxatilis) is a carnivorous species and a major product of US aquaculture. To reduce costs and improve resource sustainability, traditional ingredients used in fish diets are becoming more broadly replaced by plant-based products; however, plant meals can be problematic for carnivorous fish. Bioprocessing has improved nutritional quality and allowed higher inclusions in fish diets, but these could potentially affect other systems such as the gut microbiome. In this context, the effects of bioprocessed soybean meal on the intestinal bacterial composition in hybrid striped bass were investigated. Using high-throughput sequencing of amplicons targeting the V1-V3 region of the 16S rRNA gene, no significant difference in bacterial composition was observed between fish fed a control diet, and fish fed a diet with the base bioprocessed soybean meal. The prominent Operational Taxonomic Unit (OTU) in these samples was predicted to be a novel species affiliated to Peptostreptococcaceae. In contrast, the intestinal bacterial communities of fish fed bioprocessed soybean meal that had been further modified after fermentation exhibited lower alpha diversity (p < 0.05), as well as distinct and more varied composition patterns, with OTUs predicted to be strains of Lactococcus lactis, Plesiomonas shigelloides, or Ralstonia pickettii being the most dominant. Together, these results suggest that compounds in bioprocessed soybean meal can affect intestinal bacterial communities in hybrid striped bass.
Collapse
Affiliation(s)
- Emily Celeste Fowler
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA; (E.C.F.); (P.P.)
| | - Prakash Poudel
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA; (E.C.F.); (P.P.)
| | - Brandon White
- Department of Natural Resource Management, South Dakota State University, Brookings, SD 57007, USA;
| | - Benoit St-Pierre
- Department of Animal Science, South Dakota State University, Brookings, SD 57007, USA; (E.C.F.); (P.P.)
- Correspondence: (B.S.-P.); (M.B.)
| | - Michael Brown
- Department of Natural Resource Management, South Dakota State University, Brookings, SD 57007, USA;
- Correspondence: (B.S.-P.); (M.B.)
| |
Collapse
|
40
|
Langfeld LQ, Du K, Bereswill S, Heimesaat MM. A review of the antimicrobial and immune-modulatory properties of the gut microbiota-derived short chain fatty acid propionate - What is new? Eur J Microbiol Immunol (Bp) 2021; 11:50-56. [PMID: 33950857 PMCID: PMC8287978 DOI: 10.1556/1886.2021.00005] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 04/21/2021] [Indexed: 12/26/2022] Open
Abstract
As antimicrobial resistance poses a globally rising health problem, the identification of alternative antimicrobial agents is urgently required. The short chain fatty acid propionate which is physiologically produced by the gut microbiota constitutes a promising molecule given that it has been widely used as a cosmetics and food preservative due to its antimicrobial effects. This literature survey aims to determine the most recent state of knowledge about the antimicrobial and immune-modulatory properties of propionate. Both in vitro and in vivo studies published between 2011 and 2020 confirmed the ability of propionate to inhibit the growth of several cellular pathogens, including Gram-positive and Gram-negative multi-drug resistant bacteria and fungi. In addition, heterogenous immune-modulatory and in particular, anti-inflammatory effects of propionate could be assessed involving a diverse signaling network that needs further comprehension. In conclusion, our literature survey provides evidence that propionate displays a plethora of health-beneficial including antimicrobial and immune-modulatory effects. Future research is required to further unravel the underlying molecular mechanisms and to set the basis for in vivo infection and clinical studies to broaden the path of propionate as a promising adjunct antibiotics-independent option in the combat of infections caused by multi-drug resistant bacteria.
Collapse
Affiliation(s)
- Luis Q Langfeld
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany and Berlin Institute of Health, Berlin, Germany
| | - Ke Du
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany and Berlin Institute of Health, Berlin, Germany
| | - Stefan Bereswill
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany and Berlin Institute of Health, Berlin, Germany
| | - Markus M Heimesaat
- Institute of Microbiology, Infectious Diseases and Immunology, Gastrointestinal Microbiology Research Group, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
41
|
Qi X, Zhang L, Xu J, Tao Z, Wang X, Qiu Y, Pan T, Liu Z, Qu H, Tan R, Liu J. Association of Increased Circulating Acetic Acid With Poor Survival in Pseudomonas aeruginosa Ventilator-Associated Pneumonia Patients. Front Cell Infect Microbiol 2021; 11:669409. [PMID: 33996639 PMCID: PMC8117141 DOI: 10.3389/fcimb.2021.669409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background We previously found that microbial disruption in Pseudomonas aeruginosa ventilator-associated pneumonia (PA-VAP) patients are long-lasting. Long-term microbial dysbiosis may lead to changes in metabolites. Short-chain fatty acids (SCFAs) are microbial fermentation products and show beneficial effects in patients with pneumonia. In this study, we aimed to explore the association between circulating SCFA levels and clinical outcomes in patients with PA-VAP. Methods In this study, we analyzed SCFAs in the serum of 49 patients with PA-VAP by gas chromatography-mass spectrometry analysis. Twenty of these patients died, and 29 survived. The correlation between serum SCFAs and patient survival and immune parameters was analyzed. Results We developed a partial least squares discriminant analysis (PLS-DA) model to examine differential SCFAs in 49 patients with PA-VAP. Among the seven SCFAs, only acetic acid was increased in non-survivors (P = 0.031, VIP > 1). Furthermore, high levels of acetic acid (>1.96ug/ml) showed increased 90-day mortality compared to low levels of acetic acid (<1.96ug/ml) in Kaplan-Meier survival analyses (P = 0.027). Increased acetic acid also correlated with reduced circulating lymphocyte and monocyte counts. Conclusion Our study showed that increased circulating acetic acid is associated with 90-day mortality in PA-VAP patients. The decrease in lymphocytes and monocytes might be affected by acetic acid and involved in the poor prognosis.
Collapse
Affiliation(s)
- Xiaoling Qi
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Zhang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Xu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheying Tao
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoli Wang
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuzhen Qiu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tingting Pan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaojun Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongping Qu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruoming Tan
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jialin Liu
- Department of Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Poll BG, Xu J, Jun S, Sanchez J, Zaidman NA, He X, Lester L, Berkowitz DE, Paolocci N, Gao WD, Pluznick JL. Acetate, a Short-Chain Fatty Acid, Acutely Lowers Heart Rate and Cardiac Contractility Along with Blood Pressure. J Pharmacol Exp Ther 2021; 377:39-50. [PMID: 33414131 PMCID: PMC7985618 DOI: 10.1124/jpet.120.000187] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 01/04/2021] [Indexed: 12/14/2022] Open
Abstract
Short-chain fatty acids (SCFAs) are metabolites produced almost exclusively by the gut microbiota and are an essential mechanism by which gut microbes influence host physiology. Given that SCFAs induce vasodilation, we hypothesized that they might have additional cardiovascular effects. In this study, novel mechanisms of SCFA action were uncovered by examining the acute effects of SCFAs on cardiovascular physiology in vivo and ex vivo. Acute delivery of SCFAs in conscious radiotelemetry-implanted mice results in a simultaneous decrease in both mean arterial pressure and heart rate (HR). Inhibition of sympathetic tone by the selective β-1 adrenergic receptor antagonist atenolol blocks the acute drop in HR seen with acetate administration, yet the decrease in mean arterial pressure persists. Treatment with tyramine, an indirect sympathomimetic, also blocks the acetate-induced acute drop in HR. Langendorff preparations show that acetate lowers HR only after long-term exposure and at a smaller magnitude than seen in vivo. Pressure-volume loops after acetate injection show a decrease in load-independent measures of cardiac contractility. Isolated trabecular muscle preparations also show a reduction in force generation upon SCFA treatment, though only at supraphysiological concentrations. These experiments demonstrate a direct cardiac component of the SCFA cardiovascular response. These data show that acetate affects blood pressure and cardiac function through parallel mechanisms and establish a role for SCFAs in modulating sympathetic tone and cardiac contractility, further advancing our understanding of the role of SCFAs in blood pressure regulation. SIGNIFICANCE STATEMENT: Acetate, a short-chain fatty acid, acutely lowers heart rate (HR) as well as mean arterial pressure in vivo in radiotelemetry-implanted mice. Acetate is acting in a sympatholytic manner on HR and exerts negative inotropic effects in vivo. This work has implications for potential short-chain fatty acid therapeutics as well as gut dysbiosis-related disease states.
Collapse
Affiliation(s)
- Brian G Poll
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jiaojiao Xu
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Seungho Jun
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jason Sanchez
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Nathan A Zaidman
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Xiaojun He
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Laeben Lester
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Dan E Berkowitz
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Nazareno Paolocci
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Wei Dong Gao
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| | - Jennifer L Pluznick
- Department of Physiology (B.G.P., J.X., J.S., N.Z., J.L.P.), Division of Cardiology (S.J., N.P.), Department of Anesthesiology and Critical Care Medicine (X.H., L.L., W.D.G.), Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Anesthesiology and Perioperative Medicine, The University of Alabama at Birmingham (D.B.); and Department of Biomedical Sciences, University of Padova, Padova, Italy (N.P.)
| |
Collapse
|
43
|
Ciarlo E, Heinonen T, Théroude C, Asgari F, Le Roy D, Netea MG, Roger T. Trained Immunity Confers Broad-Spectrum Protection Against Bacterial Infections. J Infect Dis 2021; 222:1869-1881. [PMID: 31889191 PMCID: PMC7653089 DOI: 10.1093/infdis/jiz692] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/30/2019] [Indexed: 12/29/2022] Open
Abstract
Background The innate immune system recalls a challenge to adapt to a secondary challenge, a phenomenon called trained immunity. Training involves cellular metabolic, epigenetic and functional reprogramming, but how broadly trained immunity protects from infections is unknown. For the first time, we addressed whether trained immunity provides protection in a large panel of preclinical models of infections. Methods Mice were trained and subjected to systemic infections, peritonitis, enteritis, and pneumonia induced by Staphylococcus aureus, Listeria monocytogenes, Escherichia coli, Citrobacter rodentium, and Pseudomonas aeruginosa. Bacteria, cytokines, leukocytes, and hematopoietic precursors were quantified in blood, bone marrow, and organs. The role of monocytes/macrophages, granulocytes, and interleukin 1 signaling was investigated using depletion or blocking approaches. Results Induction of trained immunity protected mice in all preclinical models, including when training and infection were initiated in distant organs. Trained immunity increased bone marrow hematopoietic progenitors, blood Ly6Chigh inflammatory monocytes and granulocytes, and sustained blood antimicrobial responses. Monocytes/macrophages and interleukin 1 signaling were required to protect trained mice from listeriosis. Trained mice were efficiently protected from peritonitis and listeriosis for up to 5 weeks. Conclusions Trained immunity confers broad-spectrum protection against lethal bacterial infections. These observations support the development of trained immunity-based strategies to improve host defenses.
Collapse
Affiliation(s)
- Eleonora Ciarlo
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Tytti Heinonen
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Charlotte Théroude
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Fatemeh Asgari
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Didier Le Roy
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| | - Mihai G Netea
- Radboud Center for Infectious Diseases, and Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.,Department for Genomics & Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Thierry Roger
- Infectious Diseases Service, Department of Medicine, Lausanne University Hospital and University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
44
|
Yang G, Ahmad F, Zhou Q, Guo M, Liang S, Gaal HA, Mo J. Investigation of Physicochemical Indices and Microbial Communities in Termite Fungus-Combs. Front Microbiol 2021; 11:581219. [PMID: 33519727 PMCID: PMC7843810 DOI: 10.3389/fmicb.2020.581219] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/14/2020] [Indexed: 01/15/2023] Open
Abstract
Termitomyces species are wild edible mushrooms that possess high nutritional value and a wide range of medicinal properties. However, the cultivation of these mushrooms is very difficult because of their symbiotic association with termites. In this study, we aimed to examine the differences in physicochemical indices and microbial communities between combs with Termitomyces basidiomes (CF) and combs without Termitomyces basidiomes (CNF). High-performance liquid chromatography (HPLC), inductively coupled plasma optical emission spectrometry (ICP-OES), gas chromatography equipped with a flame ionization detector (GC-FID), some commercial kits, high-throughput sequencing of the 16s RNA, and internal transcribed spacer (ITS) were used. Humidity, pH, and elements, i.e., Al, Ba, Fe, Mn, Ni, S, Ca, and Mg were higher while amino acids particularly alanine, tyrosine, and isoleucine were lower in CF as compared to CNF. The average contents of fatty acids were not significantly different between the two comb categories. The bacterial genera Alistipes, Burkholderia, Sediminibacterium, and Thermus were dominant in all combs. Brevibacterium, Brevundimonas, and Sediminibacterium were significantly more abundant in CF. Basidiomycota and Ascomycota were also identified in combs. Termitomyces clypeatus, Termitomyces sp. Group3, and Termitomyces sp. were the most dominant species in combs. However, any single Termitomyces species was abundantly present in an individual comb.
Collapse
Affiliation(s)
- Guiying Yang
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Farhan Ahmad
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.,Entomology Section, Central Cotton Research Institute, Sakrand, Pakistan
| | - Qihuan Zhou
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Meixia Guo
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Shiyou Liang
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| | - Hassan Ahmed Gaal
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.,Department of Entomology, Faculty of Veterinary and Animal Husbandry, Somali National University, Mogadishu, Somalia
| | - Jianchu Mo
- Ministry of Agriculture Key Laboratory of Molecular Biology of Crop Pathogens and Insect Pests, Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
| |
Collapse
|
45
|
Cholan PM, Han A, Woodie BR, Watchon M, Kurz AR, Laird AS, Britton WJ, Ye L, Holmes ZC, McCann JR, David LA, Rawls JF, Oehlers SH. Conserved anti-inflammatory effects and sensing of butyrate in zebrafish. Gut Microbes 2020; 12:1-11. [PMID: 33064972 PMCID: PMC7575005 DOI: 10.1080/19490976.2020.1824563] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Short-chain fatty acids (SCFAs) are produced by microbial fermentation of dietary fiber in the gut. Butyrate is a particularly important SCFA with anti-inflammatory properties and is generally present at lower levels in inflammatory diseases associated with gut microbiota dysbiosis in mammals. We aimed to determine if SCFAs are produced by the zebrafish microbiome and if SCFAs exert conserved effects on zebrafish immunity as an example of the non-mammalian vertebrate immune system. We demonstrate that bacterial communities from adult zebrafish intestines synthesize all three main SCFA in vitro, although SCFA were below our detectable limits in zebrafish intestines in vivo. Immersion in butyrate, but not acetate or propionate, reduced the recruitment of neutrophils and M1-type pro-inflammatory macrophages to wounds. We found conservation of butyrate sensing by neutrophils via orthologs of the hydroxycarboxylic acid receptor 1 (hcar1) gene. Neutrophils from Hcar1-depleted embryos were no longer responsive to the anti-inflammatory effects of butyrate, while macrophage sensitivity to butyrate was independent of Hcar1. Our data demonstrate conservation of anti-inflammatory butyrate effects and identify the presence of a conserved molecular receptor in fish.
Collapse
Affiliation(s)
- Pradeep Manuneedhi Cholan
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia
| | - Alvin Han
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Brad R Woodie
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia.,Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Maxinne Watchon
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University , Macquarie Park, Australia.,Sydney Medical School, The University of Sydney , Camperdown, Australia
| | - Angela Rm Kurz
- Centenary Imaging and Sydney Cytometry at the Centenary Institute, The University of Sydney , Camperdown, Australia
| | - Angela S Laird
- Centre for Motor Neuron Disease Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University , Macquarie Park, Australia
| | - Warwick J Britton
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia.,The University of Sydney, Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, And Marie Bashir Institute , Camperdown, Australia.,Department of Clinical Immunology, Royal Prince Alfred Hospital , Camperdown, Australia
| | - Lihua Ye
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Zachary C Holmes
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Jessica R McCann
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - John F Rawls
- Department of Molecular Genetics and Microbiology, Duke Microbiome Center, Duke University School of Medicine , Durham, NC, USA
| | - Stefan H Oehlers
- Tuberculosis Research Program at the Centenary Institute, The University of Sydney , Camperdown, Australia.,The University of Sydney, Discipline of Infectious Diseases and Immunology, Faculty of Medicine and Health, And Marie Bashir Institute , Camperdown, Australia
| |
Collapse
|
46
|
Gomaa EZ. Human gut microbiota/microbiome in health and diseases: a review. Antonie van Leeuwenhoek 2020; 113:2019-2040. [PMID: 33136284 DOI: 10.1007/s10482-020-01474-7] [Citation(s) in RCA: 667] [Impact Index Per Article: 133.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022]
Abstract
The human gut microbiota has received considerable interest in the recent years and our knowledge of the inhabitant species and their potential applications is increased particularly after the development of metagenomic studies. Gut microbiota is highly diverse and harboring trillions of microorganisms in human digestive system. The shaping and multiplication of gut microbiome starts at birth, while the modification of their composition depends mainly on various genetic, nutritional and environmental factors. The modification in the composition and function of the gut microbiota can change intestinal permeability, digestion and metabolism as well as immune responses. The pro inflammatory state caused by alternation of gut microbiota balance lead to the onset of many diseases ranging from gastrointestinal and metabolic conditions to immunological and neuropsychiatric diseases. In this context, the present review clarifies the role of gut microbiota in maintaining host health and investigates how nutritional and environmental factors affect the gut microbial structure and function. In addition, many therapeutic strategies of gut microbiota aimed at modulating and restoring of the intestinal ecosystem balance have been surveyed.
Collapse
Affiliation(s)
- Eman Zakaria Gomaa
- Department of Biological and Geological Sciences, Faculty of Education, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
47
|
Usta-Gorgun B, Yilmaz-Ersan L. Short-chain fatty acids production by Bifidobacterium species in the presence of salep. ELECTRON J BIOTECHN 2020. [DOI: 10.1016/j.ejbt.2020.06.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
48
|
Silva CADME, Rojony R, Bermudez LE, Danelishvili L. Short-Chain Fatty Acids Promote Mycobacterium avium subsp. hominissuis Growth in Nutrient-Limited Environments and Influence Susceptibility to Antibiotics. Pathogens 2020; 9:pathogens9090700. [PMID: 32859077 PMCID: PMC7559849 DOI: 10.3390/pathogens9090700] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 12/13/2022] Open
Abstract
Mycobacterium avium subsp. hominissuis (MAH) is a common intracellular pathogen that infects immunocompromised individuals and patients with pre-existing chronic lung diseases, such as cystic fibrosis, who develop chronic and persistent pulmonary infections. The metabolic remodeling of MAH in response to host environmental stresses or within biofilms formed in bronchial airways plays an important role in development of the persistence phenotype contributing to the pathogen’s tolerance to antibiotic treatment. Recent studies suggest a direct relationship between bacterial metabolic state and antimicrobial susceptibility, and improved antibiotic efficacy has been associated with the enhanced metabolism in bacteria. In the current study, we tested approximately 200 exogenous carbon source-dependent metabolites and identified short-chain fatty acid (SCFA) substrates (propionic, butyric and caproic acids) that MAH can utilize in different physiological states. Selected SCFA enhanced MAH metabolic activity in planktonic and sessile states as well as in the static and established biofilms during nutrient-limited condition. The increased bacterial growth was observed in all conditions except in established biofilms. We also evaluated the influence of SCFA on MAH susceptibility to clinically used antibiotics in established biofilms and during infection of macrophages and found significant reduction in viable bacterial counts in vitro and in cultured macrophages, suggesting improved antibiotic effectiveness against persistent forms of MAH.
Collapse
Affiliation(s)
- Carlos Adriano de Matos e Silva
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (C.A.d.M.e.S.); (R.R.); (L.E.B.)
| | - Rajoana Rojony
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (C.A.d.M.e.S.); (R.R.); (L.E.B.)
| | - Luiz E. Bermudez
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (C.A.d.M.e.S.); (R.R.); (L.E.B.)
- Department of Microbiology, College of Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Lia Danelishvili
- Department of Biomedical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR 97331, USA; (C.A.d.M.e.S.); (R.R.); (L.E.B.)
- Correspondence: ; Tel.: +1-(541)-737-6544; Fax: +1-(541)-737-2730
| |
Collapse
|
49
|
Wu T, Xu F, Su C, Li H, Lv N, Liu Y, Gao Y, Lan Y, Li J. Alterations in the Gut Microbiome and Cecal Metabolome During Klebsiella pneumoniae-Induced Pneumosepsis. Front Immunol 2020; 11:1331. [PMID: 32849494 PMCID: PMC7411141 DOI: 10.3389/fimmu.2020.01331] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/26/2020] [Indexed: 12/15/2022] Open
Abstract
Klebsiella (K.) pneumoniae is a common cause of pneumonia-derived sepsis in human and is associated with high morbidity and mortality. The microbiota promotes and maintains host immune homeostasis during bacterial infections. However, the mechanisms by which the gut microbiota affects immune responses in the lung still remain poorly understood. Here, we performed cecal metabolomics sequencing and fecal 16s rRNA sequencing in K. pneumoniae-infected mice and uninfected controls and showed that K. pneumoniae infection led to profound alterations in the gut microbiome and thus the cecal metabolome. We observed that the levels of Lactobacillus reuteri and Bifidobacterium pseudolongum were significantly decreased in K. pneumoniae-infected mice. Spearman correlation analysis showed that alterations in the richness and composition of the gut microbiota were associated with profound changes in host metabolite concentrations. Further, short-chain fatty acids (SCFAs), including acetate, propionate, and butyrate, were detected in cecal contents and serum by gas chromatography-mass spectrometry (GC-MS). We observed that the concentrations of these three SCFAs were all lower in the infected groups than in the untreated controls. Lastly, oral supplementation with these three SCFAs reduced susceptibility to K. pneumoniae infections, as indicated by lower bacterial burdens in the lung and higher survival rates. Our data highlight the protective roles of gut microbiota and certain metabolites in K. pneumoniae-pneumonia and suggests that it is possible to intervene in this bacterial pneumonia by targeting the gut microbiota.
Collapse
Affiliation(s)
- Ting Wu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fangming Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cong Su
- Department of Infectious Diseases, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hongru Li
- Department of Neurology, Xiangya Hospital Central South University, Changsha, China
| | - Na Lv
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanyan Liu
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China.,Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Yufeng Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yanhu Lan
- Anhui Center for Surveillance of Bacterial Resistance, Hefei, China.,Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| | - Jiabin Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Infectious Diseases, The Chaohu Affiliated Hospital of Anhui Medical University, Hefei, China.,Anhui Center for Surveillance of Bacterial Resistance, Hefei, China.,Institute of Bacterial Resistance, Anhui Medical University, Hefei, China
| |
Collapse
|
50
|
Hu X, Guo J, Zhao C, Jiang P, Maimai T, Yanyi L, Cao Y, Fu Y, Zhang N. The gut microbiota contributes to the development of Staphylococcus aureus-induced mastitis in mice. THE ISME JOURNAL 2020; 14:1897-1910. [PMID: 32341472 PMCID: PMC7305118 DOI: 10.1038/s41396-020-0651-1] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/19/2022]
Abstract
Mastitis is one of the most prevalent diseases in dairy farming worldwide. The gut microbiota plays an important role in the regulation of systemic and local inflammatory diseases, such as mastitis. However, the regulatory mechanism of the gut microbiota on mastitis is still unclear. Thus, the aim of this study was to investigate the function and regulatory mechanisms of the gut microbiota in host defense against mastitis caused by Staphylococcus aureus (S. aureus) infection. Increased blood-milk barrier permeability, and S. aureus-induced mastitis severity were observed gut microbiota-dysbiosis mice compared with those in control mice. Moreover, feces microbiota transplantation (FMT) to microbbiota-dysbiosis mice reversed these effects. Furthermore, established disruption of commensal homeostasis results in significantly increased abundance of pathogenic Enterobacter bacteria, while the relative abundance of short-chain fatty acid (SCFAs)-producing bacterial phyla (Firmicutes and Bacteroidetes) was significantly reduced. However, FMT to gut microbiota-dysbiosis mice reversed these changes. In addition, dysbiosis reduced the levels of SCFAs, and administration of sodium propionate, sodium butyrate, and probiotics (butyrate-producing bacteria) reversed the changes in the blood-milk barrier and reduced the severity of mastitis induced by S. aureus. In conclusion, this new finding demonstrated that the gut microbiota acts as a protective factor in host defense against mastitis and that targeting the gut-mammary gland axis represents a promising therapeutic approach for mastitis treatment.
Collapse
Affiliation(s)
- Xiaoyu Hu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - Jian Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - Caijun Zhao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - Peng Jiang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - T Maimai
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - Li Yanyi
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China
| | - Yunhe Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China.
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, Changchun, 130062, Jilin, People's Republic of China.
| |
Collapse
|