1
|
Zhang Y, Zhang X, Lv L, Gao S, Li X, Wang R, Wang P, Shi F, She J, Wang Y. Versatile inulin/trans-ferulic acid/silk sericin nanoparticles-nourished probiotic complex with prolonged intestinal retention for synergistic therapy of inflammatory bowel disease. Carbohydr Polym 2025; 350:123063. [PMID: 39647933 DOI: 10.1016/j.carbpol.2024.123063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/31/2024] [Accepted: 11/23/2024] [Indexed: 12/10/2024]
Abstract
To achieve effective long-term synergistic treatment of inflammatory bowel disease (IBD) with probiotics, we developed a versatile inulin/trans-ferulic acid/silk sericin nanoparticles-nourished probiotic complex. Inulin/TFA/SS nanoparticles were fabricated by inulin, trans-ferulic acid (TFA), and silk sericin (SS), and then loaded onto the surface of poly-l-lysine (PLL) and poly-glutamic acid (PGA)-coated Bifidobacterium longum (BL) to obtain BL@PLL-PGA-Inulin/TFA/SS NPs (BL@PP-NPs). This design simultaneously endowed the complex with excellent gastrointestinal resistance, antioxidant, and anti-inflammation abilities. Moreover, the inulin in the nanoparticles acts as a prebiotic, promoting the Bifidobacterium's rapid proliferation to exert effects within a short period. Compared with uncoated BL, BL@PP-NPs exhibited excellent gastric acid tolerance and up to 31.32-fold colonic colonization, and the ROS scavenging and proliferative capacity were increased by 5.61- and 1.39-fold, respectively. In a mouse model of dextran sulfate sodium (DSS)- and trinitrobenzene sulfonic acid (TNBS)-induced colitis, the components of Inulin/TFA/SS NPs synergized with probiotics to efficiently treat IBD by attenuating oxidative stress, decreasing inflammation, repairing intestinal barrier, and promoting the rapid proliferation of probiotics to reverse gut microbial disorders. Collectively, food-grade BL@PP-NPs represent a novel approach to probiotic modification that offers an effective, safe, and synergistic therapy for IBD.
Collapse
Affiliation(s)
- Yujie Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China
| | - Xiaojiang Zhang
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China
| | - Lianxi Lv
- Health Science Center, Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China
| | - Sheng Gao
- Health Science Center, Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China
| | - Xiang Li
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China; Department of Otorhinolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China
| | - Ruochen Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China
| | - Pengqian Wang
- Department of Chemical Engineering, School of Water and Environment, Chang'an University, 710064 Xi'an, Shaanxi, PR China.
| | - Feiyu Shi
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China.
| | - Junjun She
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China; Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China; Department of High Talent, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China.
| | - Ya Wang
- Center for Gut Microbiome Research, Med-X Institute, The First Affiliated Hospital of Xi'an Jiaotong University, 710061 Xi'an, Shaanxi, PR China.
| |
Collapse
|
2
|
Tchinda Defo SH, Moussa D, Bouvourné P, Guédang Nyayi SD, Woumitna GC, Kodji K, Wado EK, Ngatanko Abaissou HH, Foyet HS. Unpredictable chronic mild stress induced anxio-depressive disorders and enterobacteria dysbiosis: Potential protective effects of Detariummicrocarpum. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118940. [PMID: 39423942 DOI: 10.1016/j.jep.2024.118940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/11/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Detarium microcarpum Guill. & Perr. is used traditionally in Far North Cameroun to treat stomach aches, anxiety, epilepsy, and other mental disorders. AIM OF THE STUDY Evaluate the anxiolytic and antidepressant-like effects of D. microcarpum (DM) in unpredictable chronic mild stress (UCMS) model of depression in male rats and its impact on fecal enterobacteria of stressed rats. MATERIALS AND METHODS Rats were handled daily (control) or subjected to the UCMS procedure for 42 days. Anxiety-like behaviors were assessed using the light and dark box test (LBD) and the open field test (OFT). Depressive-like behaviors were assessed using the forced swimming test (FST), the sucrose preference test (SPT), and the novelty suppressed feeding test (NSFT). Feces were then collected, followed by blood, brain, and duodenum sections after sacrifice. Monoamine levels, pro-inflammatory cytokines, oxidative stress factors, and nitrosative stress were assessed. Feces were introduced into Hectoen enteric agar for the identification of enterobacteria. An in vitro growth test was performed. RESULTS The DM ethanolic extract has significantly increased the time spent in the light box, in the LBD, and in the center area of the OFT. Moreover, the extract has significantly reduced the preference for sucrose in the SPT, the time of immobility in the FST, and the latency period to consume the pet in the NSFT. DM extract has significantly reduced serum cortisol levels. It also significantly decreased the pro-inflammatory cytokines TNF-α and Il-1β in both brain and duodenum homogenate. DM has increased the brain's serotonin, GABA, and dopamine levels. The DM extract also decreased the MDA and nitrite levels. It also increased the SOD and CAT activities in both brain and duodenal homogenate. Histologically, the DM extract restored the cell's density in hippocampi sections and prevented gut inflammation and peroxidation characterizing leaky gut syndrome. DM extract has no effect on the growth of enterobacteria species isolated in vitro. CONCLUSION The ethanolic extract of DM would have anxiolytic and antidepressant effects via the modulation of the HPA axis, brain antioxidant enzyme activities, inflammation, and nitrosative stress. Moreover, it could act by preventing leaky gut syndrome.
Collapse
Affiliation(s)
- Serge Hermann Tchinda Defo
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Djaouda Moussa
- Department of Life and Earth Sciences, Higher Teachers' Training College, University of Maroua, P.O. Box: 55, Maroua, Cameroon.
| | - Parfait Bouvourné
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Simon Désiré Guédang Nyayi
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Guillaume Camdi Woumitna
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Kalib Kodji
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Eglantine Keugong Wado
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Hervé Hervé Ngatanko Abaissou
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| | - Harquin Simplice Foyet
- Laboratory of Cognitive and Behavioural Neuroscience, Faculty of Science, University of Maroua, Cameroon P.O. Box: 814, Maroua, Cameroon.
| |
Collapse
|
3
|
Correa Lopes B, Turck J, Tolbert MK, Giaretta PR, Suchodolski JS, Pilla R. Prolonged storage reduces viability of Peptacetobacter (Clostridium) hiranonis and core intestinal bacteria in fecal microbiota transplantation preparations for dogs. Front Microbiol 2025; 15:1502452. [PMID: 39839105 PMCID: PMC11747423 DOI: 10.3389/fmicb.2024.1502452] [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: 09/26/2024] [Accepted: 12/17/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Fecal microbiota transplantation (FMT) has been described useful as an adjunct treatment for chronic enteropathy in dogs. Different protocols can be used to prepare and store FMT preparations, however, the effect of these methods on microbial viability is unknown. We aimed (1) to assess the viability of several core intestinal bacterial species by qPCR and (2) to assess Peptacetobacter (Clostridium) hiranonis viability through culture to further characterize bacterial viability in different protocols for FMT preparations. Methods Bacterial abundances were assessed in feces from six healthy dogs by qPCR after propidium monoazide (PMA-qPCR) treatment for selective quantitation of viable bacteria. Conservation methods tested included lyophilization (stored at 4°C and at -20°C) and freezing with glycerol-saline solution (12.5%) and without any cryoprotectant (stored at -20°C). Additionally, the abundance of P. hiranonis was quantified using bacterial culture. Results Using PMA-qPCR, the viability of Faecalibacterium, Escherichia coli, Streptococcus, Blautia, Fusobacterium, and P. hiranonis was reduced in lyophilized fecal samples kept at 4°C and -20°C up to 6 months (p < 0.05). In frozen feces without cryoprotectant, only Streptococcus and E. coli were not significantly reduced for up to 3 months (p > 0.05). Lastly, no differences were observed in the viability of those species in glycerol-preserved samples up to 6 months (p > 0.05). When using culture to evaluate the viability of P. hiranonis, we observed that P. hiranonis abundance was lower in lyophilized samples kept at 4°C than -20°C; and P. hiranonis abundance was higher in glycerol-preserved samples for up to 6 months than in samples preserved without glycerol for up to 3 months. Moreover, the highest abundance of P. hiranonis was observed in glycerol-preserved feces. After 3 months, P. hiranonis was undetectable by culture in 83% (5/6) of the frozen samples without glycerol. Discussion While the lyophilization procedure initially reduced P. hiranonis abundance, P. hiranonis viability was stable thereafter for up to 6 months at -20°C. The higher bacterial viability detected in fecal samples preserved with glycerol confirms the use of this cryoprotectant as a reliable method to keep bacteria alive in the presence of fecal matrix for FMT purposes.
Collapse
Affiliation(s)
- Bruna Correa Lopes
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jonathan Turck
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - M. Katherine Tolbert
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Paula R. Giaretta
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Jan S. Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX, United States
- Department of Veterinary Pathology, Hygiene and Public Health, University of Milan, Milan, Italy
| |
Collapse
|
4
|
Ghare S, Warner D, Warner J, Chilton PM, Lee J, Zhang J, Wang M, Hardesty J, Treves R, Gabbard J, Anderson C, Batra L, Sreenivasan C, Kraenzle J, McCulley M, McCoy S, Zhang L, Feng W, Gondim DD, Barve S, Zheng J, Palmer K, McClain C, Kirpich I. Impact of chronic ethanol consumption and SARS-COV-2 on the liver and intestine: A pilot dose-response study in mice. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2025. [PMID: 39757351 DOI: 10.1111/acer.15528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND During the coronavirus disease 2019 (COVID-19) pandemic, there was a marked increase in alcohol consumption. COVID-19 superimposed on underlying liver disease notably worsens the outcome of many forms of liver injury. The goal of a current pilot study was to test the dual exposure of alcohol and COVID-19 infection in an experimental animal model of alcohol-associated liver disease (ALD). METHODS After 4 weeks of ethanol (EtOH) feeding, C57BL/6 male mice received SARS-CoV-2 (SARS2-N501YMA30) intranasally at 3 × 102, 1 × 103, 3 × 103, and 1 × 104 plaque-forming units (PFU). Mice were then weighed/monitored daily for morbidity/mortality for 10 days while continuing EtOH consumption. Markers of liver inflammation, injury, and intestinal barrier integrity were evaluated. RESULTS A similar gradual weight loss was observed in all inoculated mice (slightly less in the 3 × 102 group) up to post-infection day 4. Greater mortality was observed in mice receiving the highest viral dose at days 3 and 4 post-infection. The majority of the surviving mice subjected to EtOH and inoculated with 3 × 103 or 1 × 104 PFU rapidly lost 25% of their body weight and were euthanized on post-infection day 4. Analysis of liver health in animals that survived to the end of the experiment exhibited no significant changes in hepatic steatosis but had a limited increase in plasma alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels at all viral doses versus EtOH alone. However, the 1 × 104 PFU viral dose exacerbated EtOH-induced hepatic inflammation characterized by elevated levels of several pro-inflammatory cytokines, including Il-6 and Tnf-α. There was limited effect of viral infection on the intestine. CONCLUSIONS SARS-CoV-2 infection caused a dose-dependent negative impact on body weight and survival in mice fed EtOH. This pilot study suggests that early mortality observed after high-dose SARS-CoV-2 challenge could be due, in part, to hepatic dysfunction following chronic EtOH feeding.
Collapse
Affiliation(s)
- Smita Ghare
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Dennis Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jeffrey Warner
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Paula M Chilton
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jiyeon Lee
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - JingWen Zhang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Min Wang
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Josiah Hardesty
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Rui Treves
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jon Gabbard
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Charles Anderson
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Lalit Batra
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Chithra Sreenivasan
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Jennifer Kraenzle
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Matthew McCulley
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Stephanie McCoy
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Lihua Zhang
- Department of Structural & Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Wenke Feng
- Department of Structural & Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Dibson Dibe Gondim
- Department of Pathology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Shirish Barve
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Jian Zheng
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Kenneth Palmer
- Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Craig McClain
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Robley Rex Veterans Affairs Medical Center, Louisville, Kentucky, USA
| | - Irina Kirpich
- Alcohol Research Center, University of Louisville, Louisville, Kentucky, USA
- Hepatobiology and Toxicology Center, School of Medicine, University of Louisville, Louisville, Kentucky, USA
- Department of Microbiology and Immunology, School of Medicine, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
5
|
Balmori V, Marnpae M, Kamonsuwan K, Chusak C, Nungarlee U, Sivapornnukul P, Chanchaem P, Payungporn S, Charoensiddhi S, Suantawee T, Thilavech T, Adisakwattana S. Comparative effects of non-fermented and Lacticaseibacillus paracasei-fermented pomelo juice on gut microbiota composition and short-chain fatty acid production: An in vitro colonic model. Food Chem X 2024; 24:102041. [PMID: 39697599 PMCID: PMC11652756 DOI: 10.1016/j.fochx.2024.102041] [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: 08/11/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
Pomelo juice, especially from the Tubtim Siam cultivar, may offer prebiotic benefits by promoting beneficial gut bacteria. This study evaluated the impact of non-fermented and Lacticaseibacillus paracasei (L. paracasei)-fermented pomelo juice on gut microbiota using an in vitro colonic fermentation model. The L. paracasei-fermented juice significantly increased lactobacilli levels compared to the non-fermented juice, while both treatments similarly suppressed coliforms within 24 h. Microbiota analysis revealed increased richness and significant community shifts in both treatments. Moreover, the fermented juice demonstrated a greater decrease in the Firmicutes/Bacteroidetes ratio, indicating a greater impact on gut metabolism. Fermented juice promoted beneficial bacteria like L. paracasei, Bifidobacterium longum, and Faecalibacterium prauznitzii while inhibiting pathogens. These changes coincided with higher production of short-chain fatty acids (SCFAs), including acetic, propionic, and n-butyric acids. Therefore, fermenting pomelo juice with L. paracasei improves its ability to beneficially influence the gut microbiota, suggesting its potential for gut health enhancement.
Collapse
Affiliation(s)
- Vernabelle Balmori
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Food Science and Technology, Southern Leyte State University, Sogod 6606, Southern Leyte, Philippines
| | - Marisa Marnpae
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- The Halal Science Center, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kritmongkhon Kamonsuwan
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Charoonsri Chusak
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Uarna Nungarlee
- The Halal Science Center, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pavaret Sivapornnukul
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Prangwalai Chanchaem
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sunchai Payungporn
- Center of Excellence in Systems Microbiology, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Suvimol Charoensiddhi
- Department of Food Science and Technology, Faculty of Agro-Industry, Kasetsart University, Bangkok 10900, Thailand
| | - Tanyawan Suantawee
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Thavaree Thilavech
- Department of Food Chemistry, Faculty of Pharmacy, Mahidol University, Bangkok 10400, Thailand
| | - Sirichai Adisakwattana
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
6
|
Reges BM, da Silva Oliveira FA, Fonteles TV, Rodrigues S. Changes in Human Colonic Microbiota Promoted by Synbiotic Açai Juice Composed of Gluco-Oligosaccharides, Dextran, and Bifidobacterium breve NRRL B-41408. Foods 2024; 13:4121. [PMID: 39767062 PMCID: PMC11675832 DOI: 10.3390/foods13244121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/10/2024] [Accepted: 12/14/2024] [Indexed: 01/11/2025] Open
Abstract
The present study evaluates the effects of açai juice containing gluco-oligosaccharides and dextran, fermented by Bifidobacterium breve NRRL B-41408 (synbiotic juice), on the human fecal microbiota. The juice is subjected to simulated digestion and fecal fermentation after production and 42 days of refrigerated storage. High throughput 16S rRNA sequencing and HPLC are used to identify the bacterial cells and metabolites. The results show that the viability of B. breve is stable during the refrigerated storage, indicating that the metabolism is maintained even under low temperatures and pH. Furthermore, gluco-oligosaccharides and dextran prove to be resistant to gastrointestinal conditions and are quickly consumed during fecal fermentation. The synbiotic açai juice enhances the microbial diversity and stimulates the production of short-chain fatty acids (SCFA), including acetate, propionate, and isobutyrate. Elevated propionate levels are directly associated with an increased abundance of Bacteroides thetaiotaomicron, Bacteroides uniformis, Bacteroides xylanisolvens, Bacteroides dorei, Bacteroides stercoris, and Bacteroides massiliensis after 48 h of fermentation. This highlights the potential of synbiotic açai juice as a functional beverage, supported by the significant increase in microbial diversity reflected in the Shannon and Simpson's diversity indexes (Shannon = 116.6%, 117.2%, 125.15%, and 116.02%; Simpson's = 151.86%, 177.22%, 152.5%, and 163.73%).
Collapse
Affiliation(s)
- Bianca Mara Reges
- Food Engineering Department, Federal University of Ceara, Fortaleza 60440-900, CE, Brazil; (B.M.R.); (S.R.)
| | | | - Thatyane Vidal Fonteles
- Food Engineering Department, Federal University of Ceara, Fortaleza 60440-900, CE, Brazil; (B.M.R.); (S.R.)
| | - Sueli Rodrigues
- Food Engineering Department, Federal University of Ceara, Fortaleza 60440-900, CE, Brazil; (B.M.R.); (S.R.)
| |
Collapse
|
7
|
Holani R, Bar-Yoseph H, Krekhno Z, Serapio-Palacios A, Moon KM, Stacey RG, Donald KA, Deng W, Bressler B, Magaña AA, Foster LJ, Atser MG, Johnson JD, Finlay B. Bile acid-induced metabolic changes in the colon promote Enterobacteriaceae expansion and associate with dysbiosis in Crohn's disease. Sci Signal 2024; 17:eadl1786. [PMID: 39689182 DOI: 10.1126/scisignal.adl1786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 06/10/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024]
Abstract
Bile acids (BAs) affect the growth of potentially pathogenic commensals, including those from the Enterobacteriaceae family, which are frequently overrepresented in inflammatory bowel disease (IBD). BAs are normally reabsorbed in the ileum for recycling and are often increased in the colonic lumina of patients with IBD, including those with Crohn's disease (CD). Here, we investigated the influence of BAs on gut colonization by Enterobacteriaceae. We found increased abundance of Enterobacteriaceae in the colonic mucosae of patients with CD with a concomitant decrease in the transporters that resorb BAs in the ileum. The increase in Enterobacteriaceae colonization was greater in the colons of patients who had undergone terminal ileum resection compared with those with intact ileum, leading us to hypothesize that BAs promote intestinal colonization by Enterobacteriaceae. Exposure of human colonic epithelial cell lines to BAs reduced mitochondrial respiration, increased oxygen availability, and enhanced the epithelial adherence of several Enterobacteriaceae members. In a publicly available human dataset, mucosal Enterobacteriaceae was negatively associated with the expression of genes related to mitochondrial function. In a murine model, increased intestinal BA availability enhanced colonization by Escherichia coli in a manner that depended on bacterial respiration. Together, our findings demonstrate that BAs reduce mitochondrial respiration in the colon, leading to an increase in oxygen availability that facilitates Enterobacteriaceae colonization. This identification of BAs as facilitators of host-commensal interactions may be relevant to multiple intestinal diseases.
Collapse
Affiliation(s)
- Ravi Holani
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haggai Bar-Yoseph
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
- Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Zakhar Krekhno
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Antonio Serapio-Palacios
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyung-Mee Moon
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - Richard G Stacey
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
| | - Katherine A Donald
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Wanyin Deng
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Brian Bressler
- Division of Gastroenterology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Armando A Magaña
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael G Atser
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - James D Johnson
- Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Barton Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, British Columbia, Canada
- Biochemistry and Molecular Biology Department, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
8
|
Guo Y, Wu X, Wang Y, Zeqian Y, Cao L, Zhu F. Prediction of early remission after infliximab in Crohn's disease using baseline microbiome and metabolomics. J Pharm Biomed Anal 2024; 251:116424. [PMID: 39180897 DOI: 10.1016/j.jpba.2024.116424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
To characterize the microbiome and metabolic profile in Crohn's disease (CD) patients with different outcome after infliximab (IFX) treatment. The clinical data of a cohort of 35 patients with moderate-to-severe CD admitted at Jinling hospital between Oct 2022 and Dec 2023 were collected. Stool samples at baseline were collected to perform 16SrRNA and ITS2 sequencing and LC-MS untargeted metabolomics. Of these, seven discontinued IFX and underwent surgery during the induction period, and 28 received IFX at weeks 0, 2, and 6, each administered intravenously. Clinical remission was assessed based on the clinical symptoms and HBI at baseline and week 14. Baseline microbial richness and evenness was not significantly different between remission and non-remission group. The taxonomic community analysis identified decrease of Ruminococcus, Lachnoclostridium, Akkermansia in bacterial community and decrease of Asterotremella and Wallemia in fungal community in the non-remission group. LC-MS analysis showed that histamine, creatinine and L-proline significantly increased in remission group, while androsterone, berberine and episterol significantly decreased. The combined prediction model of histamine, androsterone, and episterol demonstrated a high predictive value of remission in patients after IFX treatment (AUC=0.898, p<0.001). Together, these data might facilitate a priori determination of optimal therapeutics for CD patients.
Collapse
Affiliation(s)
- Yanzhe Guo
- Department of general surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China
| | - Xianhai Wu
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, PR China; Department of Anorectum, Shandong Second Provincial General Hospital, PR China
| | - Yixiao Wang
- Jinling Clinical Medical College, Nanjing University of Chinese Medicine, PR China
| | - Yu Zeqian
- Department of general surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China
| | - Lei Cao
- Department of general surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China.
| | - Feng Zhu
- Department of general surgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, PR China.
| |
Collapse
|
9
|
Islam SMS, Singh S, Keshavarzian A, Abdel-Mohsen M. Intestinal Microbiota and Aging in People with HIV-What We Know and What We Don't. Curr HIV/AIDS Rep 2024; 22:9. [PMID: 39666149 DOI: 10.1007/s11904-024-00717-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] [Accepted: 11/29/2024] [Indexed: 12/13/2024]
Abstract
PURPOSE OF REVIEW People with HIV (PWH) experience premature aging and an elevated risk of age-related comorbidities, even with viral suppression through antiretroviral therapy (ART). We examine gastrointestinal disruptions, specifically impaired intestinal barrier integrity and microbial dysbiosis, as contributors to these comorbidities. RECENT FINDINGS HIV infection compromises the intestinal epithelial barrier, increasing permeability and microbial translocation, which trigger inflammation and cellular stress. ART does not fully restore gut barrier integrity, leading to persistent inflammation and cellular stress. Additionally, HIV-associated microbial dysbiosis favors pro-inflammatory bacteria, intensifying inflammation and tissue damage, which may contribute to premature aging in PWH. Understanding the interactions between intestinal microbiota, chronic inflammation, cellular stress, and aging is essential to developing therapies aimed at reducing inflammation and slowing age-related diseases in PWH. In this review, we discuss critical knowledge gaps and highlight the therapeutic potential of microbiota-targeted interventions to mitigate inflammation and delay age-associated pathologies in PWH.
Collapse
Affiliation(s)
| | - Shalini Singh
- Northwestern University, 300 E Superior St, Chicago, IL, 60611, USA
| | - Ali Keshavarzian
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, USA
- Departments of Internal Medicine, physiology Rush University Medical Center, Anatomy & Cell Biology, Chicago, IL, USA
| | | |
Collapse
|
10
|
García G, Soto J, Netherland M, Hasan NA, Buchaca E, Martínez D, Carlin M, de Jesus Cano R. Evaluating the Effects of Sugar Shift ® Symbiotic on Microbiome Composition and LPS Regulation: A Double-Blind, Placebo-Controlled Study. Microorganisms 2024; 12:2525. [PMID: 39770729 PMCID: PMC11678924 DOI: 10.3390/microorganisms12122525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
(1) Background: This study evaluated the effects of BiotiQuest® Sugar Shift®, a novel probiotic formulation, for its impact on gut microbiome composition and metabolic health in type 2 diabetes mellitus (T2D). T2D is characterized by chronic inflammation and gut microbiome imbalances, yet the therapeutic potential of targeted probiotics remains underexplored. (2) Methods: In a 12-week randomized, double-blind, placebo-controlled trial, 64 adults with T2D received either Sugar Shift or placebo capsules twice daily. Each dose provided 18 billion CFU of eight GRAS-certified bacterial strains and prebiotics. Clinical samples were analyzed for metabolic markers, and microbiome changes were assessed using 16S rRNA sequencing and metagenomics. (3) Results: Sugar Shift significantly reduced serum lipopolysaccharide (LPS) levels, improved insulin sensitivity (lower HOMA-IR scores), and increased short-chain fatty acid (SCFA)-producing genera, including Bifidobacterium, Faecalibacterium, Fusicatenibacter, and Roseburia. Pro-inflammatory taxa like Enterobacteriaceae decreased, with reduced LPS biosynthesis genes and increased SCFA production genes. The Lachnospiraceae:Enterobactericeae ratio emerged as a biomarker of reduced inflammation. (4) Conclusions: These findings demonstrate the potential of Sugar Shift to restore gut homeostasis, reduce inflammation, and improve metabolic health in T2D. Further studies are warranted to explore its long-term efficacy and broader application in metabolic disease management.
Collapse
Affiliation(s)
- Gissel García
- Pathology Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | - Josanne Soto
- Clinical Laboratory Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | | | - Nur A. Hasan
- EzBiome, 704 Quince Orchard Rd, Gaithersburg, MD 20878, USA (N.A.H.)
| | - Emilio Buchaca
- Internal Medicine Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | - Duniesky Martínez
- Research and Development Department, Center for Genetic Engineering and Biotechnology of Sancti Spíritus (CIGBSS), Circunvalante Norte S/N, Olivos 3, Apartado Postal 83, Sancti Spíritus 60200, Cuba;
| | - Martha Carlin
- The BioCollective, LLC, 4800 Dahlia Street, G8, Denver, CO 80216, USA;
| | - Raúl de Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| |
Collapse
|
11
|
Haro-Reyes J, Raghupathi JK, Reddivari L. Composition of Human-Associated Gut Microbiota Determines 3-DF and 3-HF Anti-Colitic Activity in IL-10 -/- Mice. Nutrients 2024; 16:4232. [PMID: 39683625 DOI: 10.3390/nu16234232] [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: 11/21/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Gut bacterial dysbiosis along with intestinal mucosal disruption plays a critical role in inflammatory disorders like ulcerative colitis. Flavonoids and other food bioactives have been studied in mice models as alternative treatments with minimal side effects. However, most of the research has been carried out with mice-native microbiota, which limits the comprehension of the interaction between flavonoids and human-associated bacteria. Hence, the objective of our study was to determine the effect of healthy human-associated microbiota on the anti-colitic activity of diets rich in anthocyanins (3-HF) and phlobaphenes (3-DF). METHODS In this regard, the interleukin (IL)-10 -/- mice model was utilized. Mice were divided into three groups for inoculation with human gut bacteria from three different healthy donors and assigned to four diets. A purified diet (Diet P) and three diets containing 25% near-isogenic lines (NILs) of corn were evaluated. Diets were substituted with NILs expressing only 3-DFs (diet B), only 3-HFs (diet C), and both 3-DF and 3-HF (diet D). RESULTS In an overall analysis, flavonoid-rich diets did not affect inflammatory markers, microbiota diversity, or gut metabolites, but diets containing anthocyanins improved barrier function parameters. However, when data was segmented by the recipient's microbiota from different human donors, the diet effects became significant. Furthermore, 3-HFs showed more beneficial effects than 3-DFs across the recipient's microbiota. CONCLUSIONS Our study suggests that the anti-colitic activity of 3-DF and 3-HF and their gut metabolites depends on the donor's microbial composition.
Collapse
Affiliation(s)
- Jose Haro-Reyes
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Jayaprakash Kanijam Raghupathi
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
- Department of Chemistry, Acharya Nagarjuna University, Guntur 522510, Andhra Pradesh, India
| | - Lavanya Reddivari
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
12
|
Costa CM, Pedrosa SS, Kirkland JL, Reis F, Madureira AR. The senotherapeutic potential of phytochemicals for age-related intestinal disease. Ageing Res Rev 2024; 104:102619. [PMID: 39638096 DOI: 10.1016/j.arr.2024.102619] [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: 09/17/2024] [Revised: 11/18/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
During the last few decades, life expectancy has increased worldwide along with the prevalence of several age-related diseases. Among aging pathways, cellular senescence and chronic inflammation (or "inflammaging") appear to be connected to gut homeostasis and dysbiosis of the microbiome. Cellular senescence is a state of essentially irreversible cell cycle arrest that occurs in response to stress. Although senescent cells (SC) remain metabolically active, they do not proliferate and can secrete inflammatory and other factors comprising the senescence-associated secretory phenotype (SASP). Accumulation of SCs has been linked to onset of several age-related diseases, in the brain, bones, the gastrointestinal tract, and other organs and tissues. The gut microbiome undergoes substantial changes with aging and is tightly interconnected with either successful (healthy) aging or disease. Senotherapeutic drugs are compounds that can clear senescent cells or modulate the release of SASP factors and hence attenuate the impact of the senescence-associated pro-inflammatory state. Phytochemicals, phenolic compounds and terpenes, which have antioxidant and anti-inflammatory activities, could also be senotherapeutic given their ability to act upon senescence-linked cellular pathways. The aim of this review is to dissect links among the gut microbiome, cellular senescence, inflammaging, and disease, as well as to explore phytochemicals as potential senotherapeutics, focusing on their interactions with gut microbiota. Coordinated targeting of these inter-related processes might unveil new strategies for promoting healthy aging.
Collapse
Affiliation(s)
- Célia Maria Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| | - Sílvia Santos Pedrosa
- Biorbis, Unipessoal LDA, Edifício de Biotecnologia da Universidade Católica Portuguesa, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| | - James L Kirkland
- Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048, USA.
| | - Flávio Reis
- Institute of Pharmacology and Experimental Therapeutics & Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra 3004-504, Portugal; Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra 3000-548, Portugal; Clinical Academic Center of Coimbra, Coimbra 3004-531, Portugal.
| | - Ana Raquel Madureira
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto 4169-005, Portugal.
| |
Collapse
|
13
|
Scanlan PD, Baquero F, Levin BR. Short-sighted evolution of virulence for invasive gut microbes: From hypothesis to tests. Proc Natl Acad Sci U S A 2024; 121:e2409905121. [PMID: 39570365 PMCID: PMC11626195 DOI: 10.1073/pnas.2409905121] [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] [Indexed: 11/22/2024] Open
Abstract
Why microbes harm their hosts is a fundamental question in evolutionary biology with broad relevance to our understanding of infectious diseases. Several hypotheses have been proposed to explain this "evolution of virulence." In this perspective, we reexamine one of these hypotheses in the specific context of the human gut microbiome, namely short-sighted evolution. According to the short-sighted evolution hypothesis, virulence is a product of niche expansion within a colonized host, whereby variants of commensal microbes establish populations in tissues and sites where the infection causes morbidity or mortality. This evolution is short-sighted in that the evolved variants that infect those tissues and sites are not transmitted to other hosts. The specific hypothesis that we propose is that some bacteria responsible for invasive infections and disease are the products of the short-sighted evolution of commensal bacteria residing in the gut microbiota. We present observations in support of this hypothesis and discuss the challenges inherent in assessing its general application to infections and diseases associated with specific members of the gut microbiota. We then describe how this hypothesis can be tested using genomic data and animal model experiments and outline how such studies will serve to provide fundamental information about both the evolution and genetic basis of virulence, and the bacteria of intensively studied yet poorly understood habitats including the gut microbiomes of humans and other mammals.
Collapse
Affiliation(s)
- Pauline D. Scanlan
- APC Microbiome Ireland, University College Cork, CorkT12 YT20, Ireland
- School of Microbiology, University College Cork, CorkT12 Y337, Ireland
| | - Fernando Baquero
- Servicio de Microbiología, Instituto Ramón y Cajal de Investigación Sanitaria, Madrid28034, Spain
- Centro de Investigación Médica en Red, Epidemiología y Salud Pública, Madrid28007, Spain
| | - Bruce R. Levin
- Department of Biology, Emory University, Atlanta, GA30322
| |
Collapse
|
14
|
Uemura T, Kawashima A, Jingushi K, Motooka D, Saito T, Sassi N, Horibe Y, Yamamoto A, Liu Y, Tani M, Yoshimura A, Oka T, Okuda Y, Yamamichi G, Ishizuya Y, Yamamoto Y, Kato T, Hatano K, Tsujikawa K, Wada H, Nonomura N. Bacterial information in serum extracellular vesicles reflects the inflammation of adherent perinephric fat. Cancer Sci 2024. [PMID: 39566543 DOI: 10.1111/cas.16410] [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: 08/02/2024] [Revised: 10/18/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024] Open
Abstract
Adipose tissue and bacterial flora are involved in metabolism in the human body. However, the relationship between the two remains unclear. Recently, the presence of circulating bacterial DNAs has been reported. We previously reported the utility of bacterial DNA in serum extracellular vesicles (EVs) for diagnosing patients with renal cell carcinoma (RCC). In this study, we aimed to assess whether there is a correlation between bacterial DNA in serum EVs and inflammation in adipose tissue. We undertook 16S rRNA metagenomic analysis of bacterial DNA in serum EVs from 77 patients with RCC (the derivation cohort). We discovered that DNAs from Enterobacteriaceae, Polaromonas, and Coxiellaceae were highly expressed in patients with low Mayo adhesive probability (MAP) scores. A lower MAP score reflects a reduced risk of dense adipose tissue and adhesions. Additionally, we combined these bacterial DNAs to create the EPC (Enterobacteriaceae, Polaromonas, Coxiellaceae) index that predicts a MAP score of 0. Subsequently, we undertook 16S rRNA metagenomic analysis of bacterial DNA in serum EVs from 32 patients with RCC (the validation cohort). The EPC index could distinguish patients with low MAP scores from those with high MAP scores in the derivation (area under the curve [AUC], 0.76; sensitivity, 56%; specificity, 85%) and validation (AUC, 0.81; sensitivity, 100%; specificity, 62%) cohorts. These results suggest that bacterial DNA in serum EVs could reflect the inflammation of adherent perinephric fat around the kidney.
Collapse
Affiliation(s)
- Toshihiro Uemura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Atsunari Kawashima
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kentaro Jingushi
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Daisuke Motooka
- Department of Infection Metagenomics, Genome Information Research Center, Osaka University Research Institute for Microbial Diseases, Suita, Japan
| | - Takuro Saito
- Department of Surgery, Graduate School of Medicine, Osaka University, Suita, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Nesrine Sassi
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yuki Horibe
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akinaru Yamamoto
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yutong Liu
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Masaru Tani
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Akihiro Yoshimura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Toshiki Oka
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yohei Okuda
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Gaku Yamamichi
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yu Ishizuya
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Yoshiyuki Yamamoto
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Taigo Kato
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Koji Hatano
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Suita, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Norio Nonomura
- Department of Urology, Graduate School of Medicine, Osaka University, Suita, Japan
| |
Collapse
|
15
|
Lee KR, Gulnaz A, Chae YJ. Drug Interaction-Informed Approaches to Inflammatory Bowel Disease Management. Pharmaceutics 2024; 16:1431. [PMID: 39598554 PMCID: PMC11597736 DOI: 10.3390/pharmaceutics16111431] [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: 09/09/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a complex and chronic condition that requires the use of various pharmacological agents for its management. Despite advancements in IBD research, the multifaceted mechanisms involved continue to pose significant challenges for strategic prevention. Therefore, it is crucial to prioritize safe and effective treatment strategies using the currently available pharmacological agents. Given that patients with IBD often require multiple medications due to combination therapy or other underlying conditions, a comprehensive understanding of drug interactions is essential for optimizing treatment regimens. In this review, we examined the pharmacological treatment options recommended in the current IBD management guidelines and provided a comprehensive analysis of the known pharmacokinetic interactions associated with these medications. In particular, this review includes recent research results for the impact of anti-drug antibodies (ADAs) on the concentrations of biological agents used in IBD treatment. By leveraging detailed interaction data and employing personalized dosing strategies, healthcare providers can improve therapeutic outcomes and minimize adverse effects, ultimately improving the quality of care for patients with IBD.
Collapse
Affiliation(s)
- Kyeong-Ryoon Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
- Department of Bioscience, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Aneela Gulnaz
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
| | - Yoon-Jee Chae
- College of Pharmacy, Woosuk University, Wanju 55338, Republic of Korea
- Research Institute of Pharmaceutical Sciences, Woosuk University, Wanju 55338, Republic of Korea
| |
Collapse
|
16
|
Wang ZQ, Zhang JY, Tang X, Zhou JB. Hypoglycemic drugs, circulating inflammatory proteins, and gallbladder diseases: A mediation mendelian randomization study. Diabetes Res Clin Pract 2024; 217:111882. [PMID: 39366640 DOI: 10.1016/j.diabres.2024.111882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/31/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND The relationship of hypoglycemic drugs, inflammatory proteins and gallbladder diseases remain unknown. METHODS Four hypoglycemic drugs were selected as exposure: glucagon-like peptide-1 receptor agonists (GLP-1RA), dipeptidyl peptidase-4 inhibitors (DPP-4i), sodium-glucose cotransporter 2 inhibitors (SGLT-2i), and metformin. The outcome were two gallbladder diseases: cholecystitis and cholelithiasis. Mendelian Randomization (MR) was employed to determine the association between hypoglycemic drugs and gallbladder diseases. RESULTS DPP-4i and SGLT-2i had no effect on cholecystitis and cholelithiasis. However, a causal relationship was found between inhibition of ETFDH gene, a target of metformin expressed in cultured fibroblasts, and cholelithiasis (OR: 0.84, 95 %CI: (0.72,0.97), p = 0.021), as well as between GLP1R expression in the brain caudate basal ganglia and cholecystitis (OR: 1.29, 95 %CI: (1.11,1.49), p = 0.001). The effect of ETFDH inhibition on cholelithiasis through Interleukin-10 receptor subunit beta (IL-10RB) levels and Neurotrophin-3 (NT-3) levels, with a mediated proportion of 8 % and 8 %, respectively. CONCLUSION Metformin plays a protective role in cholelithiasis, while GLP-1RA have a harmful effect on the risk of cholecystitis. Metformin may reduce the risk of cholelithiasis by modulating the levels of Neurotrophin-3 (NT-3) and Interleukin-10 receptor subunit beta (IL-10RB). Further clinical and mechanistic studies are required to confirm these findings.
Collapse
Affiliation(s)
- Zi-Qi Wang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jin-Yan Zhang
- Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | | | - Jian-Bo Zhou
- Department of Endocrinology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
17
|
Abuljadayel D, Alotibi A, Algothmi K, Basingab F, Alhazmi S, Almuhammadi A, Alharthi A, Alyoubi R, Bahieldin A. Gut microbiota of children with autism spectrum disorder and healthy siblings: A comparative study. Exp Ther Med 2024; 28:430. [PMID: 39328398 PMCID: PMC11425773 DOI: 10.3892/etm.2024.12719] [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: 10/31/2023] [Accepted: 05/01/2024] [Indexed: 09/28/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental abnormality that impairs social communication. The human gut microbiome (GM) influences a variety of local processes, including dysbiosis and the defense against pathogenic microorganisms. The aim of the present study was to categorize and identify molecular biomarkers for ASD. In the present study, metagenomics whole genome shotgun sequencing was used to identify the gut microbiota in autistic individuals. Fecal samples from four children with ASD and four healthy control siblings, aged 3-10 years old, were examined using bioinformatics analysis. A total of 673,091 genes were cataloged, encompassing 25 phyla and 2 kingdoms based on the taxonomy analysis. The results revealed 257 families, 34 classes, 84 orders, and 1,314 genera among 4,339 species. The top 10 most abundant genes and corresponding functional genes for each group were determined after the abundance profile was screened. The results showed that children with ASD had a higher abundance of certain gut microbiomes than their normal siblings and vice versa. The phyla Firmicutes and Proteobacteria were the most abundant in ASD. The Thermoanaerobacteria class was also restricted to younger healthy individuals. Moreover, the Lactobacillaceae family was more abundant in children with ASD. Additionally, it was discovered that children with ASD had a higher abundance of the Bacteroides genus and a lower abundance of the Bifidobacterium and Prevotella genera. In conclusion, there were more pathogenic genera and species and higher levels of biomass, diversity and richness in the GM of children with ASD.
Collapse
Affiliation(s)
- Dalia Abuljadayel
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asalah Alotibi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Khloud Algothmi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Fatemah Basingab
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Safiah Alhazmi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Immunology Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah 22252, Saudi Arabia
- Central Lab of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Asma Almuhammadi
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Amani Alharthi
- Department of Biology, College of Science in Zulfi, Majmaaha University, Zulfi 11932, Saudi Arabia
| | - Reem Alyoubi
- College of Medicine, King Abdulaziz University, Jeddah 22252, Saudi Arabia
| | - Ahmad Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
18
|
Moglad E, Elekhnawy E, Alanazi N, Al-Fakhrany OM. Repurposing simvastatin for treatment of Klebsiella pneumoniae infections: in vitro and in vivo study. BIOFOULING 2024; 40:801-815. [PMID: 39390775 DOI: 10.1080/08927014.2024.2413652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/20/2024] [Accepted: 09/27/2024] [Indexed: 10/12/2024]
Abstract
Simvastatin had minimum inhibitory concentrations of 32 to 128 µg/mL against Klebsiella pneumoniae isolates and hindered the biofilm-formation ability of 58.54% of the isolates. It considerably diminished the bacterial cell counts in the biofilms as revealed by scanning electron microscope. Also, qRT-PCR revealed a downregulation of the biofilm genes (bcsA, wza, and luxS) by simvastatin in 48.78% of the isolates. Moreover, simvastatin has significantly improved the survival of mice and decreased the burden of bacteria in the infected lungs. Also, the histological architecture was substantially improved in the simvastatin-treated group, as the alveolar sacs and bronchioles appeared normal with minimal collagen fiber deposition. The immunohistochemical studies exposed that the TNF-α, NF-kβ, and COX-2 immunostaining considerably declined in the simvastatin-treated group. Furthermore, ELISA exposed that both IL-1β and IL-6 were considerably diminished in the lungs of the simvastatin-treated group.
Collapse
Affiliation(s)
- Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Engy Elekhnawy
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Nuor Alanazi
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | | |
Collapse
|
19
|
Wang H, Liu Z, Zhan K, Ma Q, Xu L, Li Y, Liu Y. Vitamin K2 alleviates dextran sulfate sodium-induced colitis via inflammatory responses, gut barrier integrity, and the gut microbiota in mice. Int J Biol Macromol 2024; 280:136091. [PMID: 39353519 DOI: 10.1016/j.ijbiomac.2024.136091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/11/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Vitamin K2 (VK2) has been shown to have potential benefits in improving intestinal integrity, but its potential and mechanisms for alleviating intestinal inflammation are still unclear. The present results showed that VK2 supplementation significantly alleviated the symptoms of colitis and maintained the intestinal barrier integrity. In addition, VK2 significantly down-regulated the mRNA expression levels of pro-inflammatory cytokines including IL-1β, IL-6, and TNF-α, while up-regulated the mRNA expression level of anti-inflammatory cytokines such as IL-10. Moreover, VK2 significantly alleviated DSS-induced intestinal epithelial barrier dysfunction by maintaining the tight junction function. Furthermore, VK2 also regulated DSS-induced gut microbiota dysbiosis by reshaping the structure of gut microbiota, such as increasing the relative abundance of Firmicutes, Euryarchaeota, Prevotellaceae, and Prevotella and reducing the relative abundance of Proteobacteria, Rikenellaceae, Enterobacteriaceae, Acetatifactor, and Alistioes. In conclusion, these results indicated that VK2 effectively alleviates DSS-induced colitis in mice by modulating the gut microbiota.
Collapse
Affiliation(s)
- Huakai Wang
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Zhen Liu
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Kai Zhan
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China.
| | - Qiugang Ma
- Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Lei Xu
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei 230031, China
| | - Yinghao Li
- College of Animal Science, Anhui Science and Technology University, Chuzhou 233100, China
| | - Yun Liu
- Guangde City animal husbandry and veterinary aquatic services center, Xuancheng 242299, China
| |
Collapse
|
20
|
Shalmon G, Ibrahim R, Israel-Elgali I, Grad M, Shlayem R, Shapira G, Shomron N, Youngster I, Scheinowitz M. Gut Microbiota Composition Positively Correlates with Sports Performance in Competitive Non-Professional Female and Male Runners. Life (Basel) 2024; 14:1397. [PMID: 39598196 PMCID: PMC11595618 DOI: 10.3390/life14111397] [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: 09/28/2024] [Revised: 10/27/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
There is still a pressing need for further investigation to bridge the gap in understanding the differences in gut microbiota composition between female runners and their male counterparts. We aimed to determine the gut microbiota composition in competitive non-professional female and male runners and to correlate the gut bacteria to performance. Our study included 40 subjects, of which 22 were runners (13 males and 9 females) and 18 control subjects (9 males and 9 females, representing the general population who perform light physical activity with a weekly running volume of ≤5 km per week). Fecal specimens were collected and analyzed for taxonomic profiling to compare species' relative abundances between males and females based on the results of 16SrRNA analysis. Bacterial alpha and beta diversity were assessed to determine the differences in microbial composition between runners and controls, and between sexes. Each participant underwent a maximal oxygen consumption test and a time-to-exhaustion test at 85% of the measured VO2max. Blood lactate was collected every 5 min during the tests. Bacterial alpha diversity showed a significant difference (p = 0.04) between runners and controls. Taxonomic analysis of gut microbiota composition showed a lower Enterobacteriaceae abundance and a higher Methanosphaera abundance in runners compared with the control group. Ten different bacteria (Methanosphaera, Mitsuokella, Prevotellaceae, Megamonas, Rothia, Oscillospira, Bacteroides, Odoribacter, Blautia massiliensis, Butyricicoccus_pullicaecorum) were positively correlated with exercise (VO2max, lactate blood levels, time to exhaustion, and weekly training volume). We found no significant differences in the gut microbiota composition between male and female runners. Gut microbiota composition positively correlates with sports performance in competitive non-professional female and male runners, and female runners show similar gut microbiome diversity to male runners.
Collapse
Affiliation(s)
- Guy Shalmon
- Sylvan Adams Sports Institute, School of Public Health, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel;
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Rawan Ibrahim
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Ifat Israel-Elgali
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Meitar Grad
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Rani Shlayem
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Guy Shapira
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Noam Shomron
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
- Edmond J. Safra Center for Bioinformatics, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| | - Ilan Youngster
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
- Pediatric Infectious Diseases Unit, The Center for Microbiome Research, Shamir Medical Center, Tel Aviv 6997801, Israel
| | - Mickey Scheinowitz
- Sylvan Adams Sports Institute, School of Public Health, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel;
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv-Yafo 6997801, Israel
| |
Collapse
|
21
|
Pinto C, Carrasco-Loncharic T, González-Mienert E, de Solminihac J, Gálvez-Jirón F, Cifuentes F, Pino-Lagos K. IL-33 Induces a Switch in Intestinal Metabolites Revealing the Tryptophan Pathway as a Target for Inducing Allograft Survival. Nutrients 2024; 16:3655. [PMID: 39519488 PMCID: PMC11547499 DOI: 10.3390/nu16213655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 10/24/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND IL-33, a pleiotropic cytokine, has been associated with a plethora of immune-related processes, both inflammatory and anti-inflammatory. T regulatory (Treg) cells, the main leukocyte population involved in immune tolerance, can be induced by the administration of IL-33, the local microbiota, and its metabolites. Here, we demonstrate that IL-33 drastically induces the production of intestinal metabolites involved on tryptophan (Trp) metabolism. METHODS naïve mice were treated with IL-33 for 4 days and leukocyte populations were analyzed by flow cytometry, and feces were processed for microbiota and intestinal metabolites studies. Using a murine skin transplantation model, the effect of Kynurenic acid (KA) on allograft survival was tested. RESULTS Under homeostatic conditions, animals treated with IL-33 showed an increment in Treg cell frequencies. Intestinal bacterial abundance analysis indicates that IL-33 provokes dysbiosis, demonstrated by a reduction in Enterobacteria and an increment in Lactobacillus genera. Furthermore, metabolomics analysis showed a dramatic IL-33 effect on the abundance of intestinal metabolites related to amino acid synthesis pathways, highlighting molecules linked to Trp metabolism, such as kynurenic acid (KA), 5-Hydroxyindoleacetic acid (5-HIAA), and 6-Hydroxynicotinic acid (6-HNA), which was supported by an enhanced expression of Ido and Kat mRNA in MLN cells, which are two enzymes involved on KA synthesis. Interestingly, animals receiving KA in drinking water and subjected to skin transplantation showed allograft acceptance, which is associated with an increment in Treg cell frequencies. CONCLUSIONS Our study reveals a new property for IL-33 as a modulator of the intestinal microbiota and metabolites, especially those involved with Trp metabolism. In addition, we demonstrate that KA favors Tregs in vivo, positively affecting skin transplantation survival.
Collapse
Affiliation(s)
- Camila Pinto
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago 755000, Chile
| | - Tomás Carrasco-Loncharic
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago 755000, Chile
| | - Eduardo González-Mienert
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago 755000, Chile
| | - Javiera de Solminihac
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago 755000, Chile
| | - Felipe Gálvez-Jirón
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago 755000, Chile
| | - Federico Cifuentes
- Escuela de Medicina Veterinaria, Facultad de Agronomía e Ingeniería Forestal, Facultad de Ciencias Biológicas y Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Karina Pino-Lagos
- Facultad de Medicina, Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago 755000, Chile
| |
Collapse
|
22
|
Liu B, Schnider A, DeArmond M, Banach DB, Haubrich BA. Cryptosporidiosis in individuals with inflammatory bowel disease: a scoping review protocol. BMJ Open 2024; 14:e086529. [PMID: 39414295 PMCID: PMC11481120 DOI: 10.1136/bmjopen-2024-086529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/12/2024] [Indexed: 10/18/2024] Open
Abstract
INTRODUCTION Cryptosporidiosis is a leading cause of moderate-to-severe diarrhoea globally, and, while it is often self-limited, in immunocompromised individuals, the infection can be associated with significant morbidity and mortality. Diagnosis might be missed or delayed in patients with inflammatory bowel disease (IBD) due to similar presentation, and these patients may also be on immunosuppressive therapies, increasing their risk of infection. Additionally, gastrointestinal infection and dysbiosis may be a risk factor for IBD. Diagnosis, presentation and treatment of cryptosporidiosis in individuals with IBD, as well as any epidemiologic correlations between the two diseases, will be investigated. METHODS AND ANALYSIS MEDLINE, Embase, Cochrane Library, CINAHL, Dissertations and Theses Global and grey literature will be searched. Joanna Briggs Institute (JBI) methodology for scoping reviews was used for the protocol and will be for the review. Two reviewers will independently screen studies and extract data. The evidence and presentation of the results will be analysed with input from the review team. Studies of cryptosporidiosis in patients with IBD will be included. Paediatric, adolescent and adult studies in all patient environments will be included. Cases in which Crohn's disease does not affect the intestine and cases in which cryptosporidial infection is not in the intestine will be excluded. ETHICS AND DISSEMINATION Published clinical literature will be systematically reviewed, and this work does not directly involve patients. Consequently, ethical review by an institutional review board is not required. Data will be presented at academic conferences, and a culminating report will be published in a peer-reviewed journal. OPEN SCIENCE FRAMEWORK REGISTRATION: https://osf.io/j47mb.
Collapse
Affiliation(s)
- Belinda Liu
- College of Osteopathic Medicine, Touro University Nevada, Henderson, Nevada, USA
| | - Alexander Schnider
- College of Osteopathic Medicine, Touro University Nevada, Henderson, Nevada, USA
| | - Megan DeArmond
- Jay Sexter Library, Touro University Nevada, Henderson, Nevada, USA
- Touro University Nevada: A JBI Affiliated Group, Touro University Nevada, Henderson, Nevada, USA
| | - David B Banach
- School of Medicine, University of Connecticut, Farmington, Connecticut, USA
| | - Brad A Haubrich
- College of Osteopathic Medicine, Touro University Nevada, Henderson, Nevada, USA
| |
Collapse
|
23
|
Rahangdale S, Deshmukh P, Sammeta S, Aglawe M, Kale M, Umekar M, Kotagale N, Taksande B. Agmatine modulation of gut-brain axis alleviates dysbiosis-induced depression-like behavior in rats. Eur J Pharmacol 2024; 981:176884. [PMID: 39134294 DOI: 10.1016/j.ejphar.2024.176884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/20/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Depression is a global health concern affecting nearly 280 million individuals. It not only imposes a significant burden on economies and healthcare systems but also manifests complex physiological connections and consequences. Agmatine, a putative neuromodulator derived primarily from beneficial gut microbes specially Lactobacillus, has emerged as a potential therapeutic agent for mental health. The microbiota-gut-brain axis is involved in the development of depression through the peripheral nervous system, endocrine system, and immune system and may be a key factor in the effect of agmatine. Therefore, this study aimed to investigate the potential mechanism of agmatine in antibiotic-induced dysbiosis and depression-like behavior in rats, focusing on its modulation of the gut-brain axis. Depression-like behavior associated with dysbiosis was induced through a seven-day regimen of the broad-spectrum antibiotic, comprising ampicillin and metronidazole and validated through microbial, biochemical, and behavioral alterations. On day 8, antibiotic-treated rats exhibited loose fecal consistency, altered fecal microbiota, and depression-like behavior in forced swim test. Pro-inflammatory cytokines were elevated, while agmatine and monoamine levels decreased in the hippocampus and prefrontal cortex. Antibiotic administration disrupted tight junction proteins in the ileum, affecting gut architecture. Oral administration of agmatine alone or combined with probiotics significantly reversed antibiotic-induced dysbiosis, restoring gut microbiota and mitigating depression-like behaviors. This intervention also restored neuro-inflammatory markers, increased agmatine and monoamine levels, and preserved gut integrity. The study highlights the regulatory role of endogenous agmatine in the gut-brain axis in broad-spectrum antibiotic induced dysbiosis and associated depression-like behavior.
Collapse
Affiliation(s)
- Sandip Rahangdale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India
| | - Pankaj Deshmukh
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India
| | - Shivkumar Sammeta
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India
| | - Manish Aglawe
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India
| | - Mayur Kale
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India
| | - Milind Umekar
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India
| | - Nandkishor Kotagale
- Government College of Pharmacy, Kathora Naka, VMV Road, Amravati, M.S., 44604, India
| | - Brijesh Taksande
- Division of Neuroscience, Department of Pharmacology, Smt. Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur, M.S., 441 002, India.
| |
Collapse
|
24
|
Chu Z, Hu Z, Yang F, Zhou Y, Tang Y, Luo F. Didymin Ameliorates Dextran Sulfate Sodium (DSS)-Induced Ulcerative Colitis by Regulating Gut Microbiota and Amino Acid Metabolism in Mice. Metabolites 2024; 14:547. [PMID: 39452928 PMCID: PMC11509612 DOI: 10.3390/metabo14100547] [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: 08/30/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Didymin is a dietary flavonoid derived from citrus fruits and has been shown to have extensive biological functions, especially anti-inflammatory effects, but its mechanism is unclear. The purpose of this study was to investigate the potential mechanism of didymin that alleviates ulcerative colitis. Methods and Results: Our results indicated that didymin could alleviate the symptoms of ulcerative colitis, as it inhibited the expressions of interleukin-6 (IL-6), interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). Didymin also promoted the expressions of claudin-1 and zona occludens-1(ZO-1), which are closely related with restoring colon barrier function. Didymin also increased the abundance of Firmicutes and Verrucomicobiota, while decreasing the abundance of Bacteroidota and Proteobacteria. Meanwhile, didymin significantly altered the levels of metabolites related to arginine synthesis and metabolism, and lysine degradation in the colitis mice. Utilizing network pharmacology and molecular docking, our results showed that the metabolites L-ornithine and saccharin could interact with signal transducer and activator of transcription 3 (STAT3) and nuclear factor kappa-B (NF-κB). In this in vitro study, L-ornithine could reduce the expressions of transcription factors STAT3 and NF-κB, and it also inhibited the expressions of IL-6 and IL-1β in the lipopolysaccharides (LPS) induced in RAW264.7 cells, while saccharin had the opposite effect. Conclusions: Taken together, didymin can regulate gut microbiota and alter metabolite products, which can modulate STAT3 and NF-κB pathways and inhibit the expressions of inflammatory factors and inflammatory response in the DSS-induced colitis mice.
Collapse
Affiliation(s)
| | | | | | | | | | - Feijun Luo
- Hunan Provincial Key Laboratory of Deeply Processing and Quality Control of Cereals and Oils, Hunan Provincial Key Laboratory of Forestry Edible Resources Safety and Processing, National Research Center of Rice Deep Process and Byproducts, Central South University of Forestry and Technology, Changsha 410004, China; (Z.C.); (Z.H.); (F.Y.); (Y.Z.); (Y.T.)
| |
Collapse
|
25
|
Feng J, Peng J, Hsiao YC, Liu CW, Yang Y, Zhao H, Teitelbaum T, Wang X, Lu K. Non/Low-Caloric Artificial Sweeteners and Gut Microbiome: From Perturbed Species to Mechanisms. Metabolites 2024; 14:544. [PMID: 39452925 PMCID: PMC11509705 DOI: 10.3390/metabo14100544] [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: 09/15/2024] [Revised: 10/03/2024] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Non/low-caloric artificial sweeteners (NAS) are recognized as chemical additives substituting sugars to avoid caloric intake and subsequent sugar-derived diseases such as diabetes and hyperglycemia. Six NAS have been claimed safe and are authorized by the US Food and Drug Administration (FDA) for public use, with acceptable daily intake information available: aspartame, acesulfame-K, saccharin, sucralose, neotame, and advantame. However, the impacts of NAS on the gut microbiome have raised potential concerns, since sporadic research revealed NAS-induced microbial changes in the gastrointestinal tracts and alterations in the microbiome-host interactive metabolism. METHODS Given the fact that the gut microbiome influences kaleidoscopic physiological functions in host health, this review aimed to decipher the impacts of NAS on the gut microbiome by implementing a comprehensive two-stage literature analysis based on each NAS. RESULTS This review documented disturbed microbiomes due to NAS exposure to a maximal resolution of species level using taxonomic clustering analysis, and recorded metabolism alterations involved in gut microbiome-host interactions. CONCLUSIONS The results elucidated that specific NAS exhibited discrepant impacts on the gut microbiome, even though overlapping on the genera and species were identified. Some NAS caused glucose tolerance impairment in the host, but the key metabolites and their underlying mechanisms were different. Furthermore, this review embodied the challenges and future directions of current NAS-gut microbiome research to inspire advanced examination of the NAS exposure-gut microbiome-host metabolism axis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kun Lu
- Department of Environmental Sciences and Engineering, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
26
|
Kazemian N, Pakpour S. Understanding the impact of the gut microbiome on opioid use disorder: Pathways, mechanisms, and treatment insights. Microb Biotechnol 2024; 17:e70030. [PMID: 39388360 PMCID: PMC11466222 DOI: 10.1111/1751-7915.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
The widespread use of opioids for chronic pain management not only poses a significant public health issue but also contributes to the risk of tolerance, dependence, and addiction, leading to opioid use disorder (OUD), which affects millions globally each year. Recent research has highlighted a potential bidirectional relationship between the gut microbiome and OUD. This emerging perspective is critical, especially as the opioid epidemic intensifies, emphasizing the need to investigate how OUD may alter gut microbiome dynamics and vice versa. Understanding these interactions could reveal new insights into the mechanisms of addiction and tolerance, as well as provide novel approaches for managing and potentially mitigating OUD impacts. This comprehensive review explores the intricate bidirectional link through the gut-brain axis, focusing on how opiates influence microbial composition, functional changes, and gut mucosal integrity. By synthesizing current findings, the review aims to inspire new strategies to combat the opioid crisis and leverage microbiome-centred interventions for preventing and treating OUD.
Collapse
Affiliation(s)
- Negin Kazemian
- School of EngineeringUniversity of British ColumbiaKelownaBritish ColumbiaCanada
| | - Sepideh Pakpour
- School of EngineeringUniversity of British ColumbiaKelownaBritish ColumbiaCanada
| |
Collapse
|
27
|
Kropp C, Tambosco K, Chadi S, Langella P, Claus SP, Martin R. Christensenella minuta protects and restores intestinal barrier in a colitis mouse model by regulating inflammation. NPJ Biofilms Microbiomes 2024; 10:88. [PMID: 39294159 PMCID: PMC11411060 DOI: 10.1038/s41522-024-00540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 07/23/2024] [Indexed: 09/20/2024] Open
Abstract
Christensenella minuta DSM 22607 has recently been suggested as a potential microbiome-based therapy for inflammatory bowel disease (IBD) because it displays strong anti-inflammatory effects both in vitro and in vivo. Here, we aimed to decipher the mechanism(s) underlying the DSM 22607-mediated beneficial effects on the host in a mouse model of chemically induced acute colitis. We observed that C. minuta plays a key role in the preservation of the epithelial barrier and the management of DNBS-induced inflammation by inhibiting interleukin (IL)-33 and Tumor necrosis factor receptor superfamily member 8 (Tnfrsf8) gene expression. We also showed that DSM 22607 abundance was positively correlated with Akkermansia sp. and Dubosiella sp. and modulated microbial metabolites in the cecum. These results offer new insights into the biological and molecular mechanisms underlying the beneficial effects of C. minuta DSM 22607 by protecting the intestinal barrier integrity and regulating inflammation.
Collapse
Affiliation(s)
- Camille Kropp
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 68350, Jouy-en-Josas, France
- YSOPIA Bioscience, 33076, Bordeaux, France
| | - Kevin Tambosco
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 68350, Jouy-en-Josas, France
| | - Sead Chadi
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 68350, Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 68350, Jouy-en-Josas, France
| | | | - Rebeca Martin
- Micalis Institute, AgroParisTech, INRAE, Université Paris-Saclay, 68350, Jouy-en-Josas, France.
| |
Collapse
|
28
|
Matthews K, Cavagnaro T, Weinstein P, Stanhope J. Health by design; optimising our urban environmental microbiomes for human health. ENVIRONMENTAL RESEARCH 2024; 257:119226. [PMID: 38797467 DOI: 10.1016/j.envres.2024.119226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/13/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
Humans have evolved in direct and intimate contact with their environment and the microbes that it contains, over a period of 2 million years. As a result, human physiology has become intrinsically linked to environmental microbiota. Urbanisation has reduced our exposure to harmful pathogens, however there is now increasing evidence that these same health-protective improvements in our environment may also be contributing to a hidden disease burden: immune dysregulation. Thoughtful and purposeful design has the potential to ameliorate these health concerns by providing sources of microbial diversity for human exposure. In this narrative review, we highlight the role of environmental microbiota in human health and provide insights into how we can optimise human health through well-designed cities, urban landscapes and buildings. The World Health Organization recommends there should be at least one public green space of least 0.5 ha in size within 300m of a place of residence. We argue that these larger green spaces are more likely to permit functioning ecosystems that deliver ecosystem services, including the provision of diverse aerobiomes. Urban planning must consider the conservation and addition of large public green spaces, while landscape design needs to consider how to maximise environmental, social and public health outcomes, which may include rewilding. Landscape designers need to consider how people use these spaces, and how to optimise utilisation, including for those who may experience challenges in access (e.g. those living with disabilities, people in residential care). There are also opportunities to improve health via building design that improves access to diverse environmental microbiota. Considerations include having windows that open, indoor plants, and the relationship between function, form and organization. We emphasise possibilities for re-introducing potentially health-giving microbial exposures into urban environments, particularly where the benefits of exposure to biodiverse environments may have been lost.
Collapse
Affiliation(s)
- Kate Matthews
- College of Science and Engineering, Flinders University, Bedford Park, SA, Australia; School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia
| | - Timothy Cavagnaro
- College of Science and Engineering, Flinders University, Bedford Park, SA, Australia; School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, SA, Australia; Environment Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Philip Weinstein
- Environment Institute, The University of Adelaide, Adelaide, SA, Australia; School of Public Health, The University of Adelaide, Adelaide, SA, Australia; South Australian Museum, Adelaide, SA, Australia
| | - Jessica Stanhope
- Environment Institute, The University of Adelaide, Adelaide, SA, Australia; School of Allied Health Science and Practice, The University of Adelaide, Adelaide, SA, Australia.
| |
Collapse
|
29
|
Toto F, Marangelo C, Scanu M, De Angelis P, Isoldi S, Abreu MT, Cucchiara S, Stronati L, Del Chierico F, Putignani L. A Novel Microbial Dysbiosis Index and Intestinal Microbiota-Associated Markers as Tools of Precision Medicine in Inflammatory Bowel Disease Paediatric Patients. Int J Mol Sci 2024; 25:9618. [PMID: 39273567 PMCID: PMC11395508 DOI: 10.3390/ijms25179618] [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: 08/06/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
Recent evidence indicates that the gut microbiota (GM) has a significant impact on the inflammatory bowel disease (IBD) progression. Our aim was to investigate the GM profiles, the Microbial Dysbiosis Index (MDI) and the intestinal microbiota-associated markers in relation to IBD clinical characteristics and disease state. We performed 16S rRNA metataxonomy on both stools and ileal biopsies, metabolic dysbiosis tests on urine and intestinal permeability and mucosal immunity activation tests on the stools of 35 IBD paediatric patients. On the GM profile, we assigned the MDI to each patient. In the statistical analyses, the MDI was correlated with clinical parameters and intestinal microbial-associated markers. In IBD patients with high MDI, Gemellaceae and Enterobacteriaceae were increased in stools, and Fusobacterium, Haemophilus and Veillonella were increased in ileal biopsies. Ruminococcaceae and WAL_1855D were enriched in active disease condition; the last one was also positively correlated to MDI. Furthermore, the MDI results correlated with PUCAI and Matts scores in ulcerative colitis patients (UC). Finally, in our patients, we detected metabolic dysbiosis, intestinal permeability and mucosal immunity activation. In conclusion, the MDI showed a strong association with both severity and activity of IBD and a positive correlation with clinical scores, especially in UC. Thus, this evidence could be a useful tool for the diagnosis and prognosis of IBD.
Collapse
Affiliation(s)
- Francesca Toto
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Chiara Marangelo
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Paola De Angelis
- Digestive Endoscopy and Surgery Unit, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Sara Isoldi
- Pediatric Gastroenterology and Hepatology Unit, Santobono-Pausilipon Children's Hospital, 80122 Naples, Italy
| | - Maria Teresa Abreu
- Crohn's and Colitis Center, Division of Digestive Health and Liver Diseases, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Salvatore Cucchiara
- Maternal Child Health Department, Pediatric Gastroenterology and Liver Unit, Sapienza University of Rome, 00185 Rome, Italy
| | - Laura Stronati
- Department of Molecular Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Federica Del Chierico
- Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics and Immunology, Rheumatology and Infectious Diseases Research Area, Unit of Microbiome, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| |
Collapse
|
30
|
van Lingen E, Nooij S, Terveer EM, Crossette E, Prince AL, Bhattarai SK, Watson A, Galazzo G, Menon R, Szabady RL, Bucci V, Norman JM, van der Woude CJ, van der Marel S, Verspaget HW, van der Meulen-de Jong AE, Keller JJ. Faecal Microbiota Transplantation Engraftment After Budesonide or Placebo in Patients With Active Ulcerative Colitis Using Pre-selected Donors: A Randomized Pilot Study. J Crohns Colitis 2024; 18:1381-1393. [PMID: 38572716 PMCID: PMC11369067 DOI: 10.1093/ecco-jcc/jjae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/30/2023] [Indexed: 04/05/2024]
Abstract
BACKGROUND Faecal microbiota transplantation [FMT] shows some efficacy in treating patients with ulcerative colitis [UC], although variability has been observed among donors and treatment regimens. We investigated the effect of FMT using rationally selected donors after pretreatment with budesonide or placebo in active UC. METHODS Patients ≥18 years old with mild to moderate active UC were randomly assigned to 3 weeks of budesonide [9 mg] or placebo followed by 4-weekly infusions of a donor faeces suspension. Two donors were selected based on microbiota composition, regulatory T cell induction and short-chain fatty acid production in mice. The primary endpoint was engraftment of donor microbiota after FMT. In addition, clinical efficacy was assessed. RESULTS In total, 24 patients were enrolled. Pretreatment with budesonide did not increase donor microbiota engraftment [p = 0.56] nor clinical response, and engraftment was not associated with clinical response. At week 14, 10/24 [42%] patients achieved [partial] remission. Remarkably, patients treated with FMT suspensions from one donor were associated with clinical response [80% of responders, p < 0.05] but had lower overall engraftment of donor microbiota. Furthermore, differences in the taxonomic composition of the donors and the engraftment of certain taxa were associated with clinical response. CONCLUSION In this small study, pretreatment with budesonide did not significantly influence engraftment or clinical response after FMT. However, clinical response appeared to be donor-dependent. Response to FMT may be related to transfer of specific strains instead of overall engraftment, demonstrating the need to characterize mechanisms of actions of strains that maximize therapeutic benefit in UC.
Collapse
Affiliation(s)
- Emilie van Lingen
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sam Nooij
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Elisabeth M Terveer
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Shakti K Bhattarai
- University of Massachusetts Chan Medical School, Department of Microbiology and Physiological Systems, Worcester, MA, USA
| | | | | | | | - Rose L Szabady
- Vedanta Biosciences, Cambridge, MA, USA
- Ferring Pharmaceuticals, San Diego, CA, USA
| | - Vanni Bucci
- University of Massachusetts Chan Medical School, Department of Microbiology and Physiological Systems, Worcester, MA, USA
| | | | - C Janneke van der Woude
- Department of Gastroenterology and Hepatology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sander van der Marel
- Department of Gastroenterology and Hepatology, Haaglanden Medisch Centrum, den Haag, The Netherlands
| | - Hein W Verspaget
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Josbert J Keller
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Gastroenterology and Hepatology, Haaglanden Medisch Centrum, den Haag, The Netherlands
| |
Collapse
|
31
|
Bertolini A, Nguyen M, Zehra SA, Taleb SA, Bauer-Pisani T, Palm N, Strazzabosco M, Fiorotto R. Prominent role of gut dysbiosis in the pathogenesis of cystic fibrosis-related liver disease in mice. J Hepatol 2024; 81:429-440. [PMID: 38554847 PMCID: PMC11347101 DOI: 10.1016/j.jhep.2024.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND & AIMS Cystic fibrosis-related liver disease (CFLD) is a chronic cholangiopathy that increases morbidity and mortality in patients with CF. Current treatments are unsatisfactory, and incomplete understanding of CFLD pathogenesis hampers therapeutic development. We have previously shown that mouse CF cholangiocytes respond to lipopolysaccharide with excessive inflammation. Thus, we investigated the role of the gut-liver axis in the pathogenesis of CFLD. METHODS Wild-type (WT), whole-body Cftr knockout (CFTR-KO) and gut-corrected (CFTR-KO-GC) mice were studied. Liver changes were assessed by immunohistochemistry and single-cell transcriptomics (single-cell RNA sequencing), inflammatory mediators were analysed by proteome array, faecal microbiota by 16S ribosomal RNA sequencing and gut permeability by FITC-dextran assay. RESULTS The livers of CFTR-KO mice showed ductular proliferation and periportal inflammation, whereas livers of CFTR-KO-GC mice had no evident pathology. Single-cell RNA sequencing analysis of periportal cells showed increased presence of neutrophils, macrophages and T cells, and activation of pro-inflammatory and pathogen-mediated immune pathways in CFTR-KO livers, consistent with a response to gut-derived stimuli. CFTR-KO mice exhibited gut dysbiosis with enrichment of Enterobacteriaceae and Enterococcus spp., which was associated with increased intestinal permeability and mucosal inflammation, whereas gut dysbiosis and inflammation were absent in CFTR-KO-GC mice. Treatment with nonabsorbable antibiotics ameliorated intestinal permeability and liver inflammation in CFTR-KO mice. Faecal microbiota transfer from CFTR-KO to germ-free WT mice did not result in dysbiosis nor liver pathology, indicating that defective intestinal CFTR is required to maintain dysbiosis. CONCLUSION Defective CFTR in the gut sustains a pathogenic microbiota, creates an inflammatory milieu, and alters intestinal permeability. These changes are necessary for the development of cholangiopathy. Restoring CFTR in the intestine or modulating the microbiota could be a promising strategy to prevent or attenuate liver disease. IMPACT AND IMPLICATIONS Severe cystic fibrosis-related liver disease (CFLD) affects 10% of patients with cystic fibrosis (CF) and contributes to increased morbidity and mortality. Treatment options remain limited due to a lack of understanding of disease pathophysiology. The cystic fibrosis transmembrane conductance regulator (CFTR) mediates Cl- and HCO3- secretion in the biliary epithelium and its defective function is thought to cause cholestasis and excessive inflammatory responses in CF. However, our study in Cftr-knockout mice demonstrates that microbial dysbiosis, combined with increased intestinal permeability caused by defective CFTR in the intestinal mucosa, acts as a necessary co-factor for the development of CFLD-like liver pathology in mice. These findings uncover a major role for the gut microbiota in CFLD pathogenesis and call for further investigation and clinical validation to develop targeted therapeutic strategies acting on the gut-liver axis in CF.
Collapse
Affiliation(s)
- Anna Bertolini
- Department of Internal Medicine, Section of Digestive Diseases, Yale Liver Center, Yale School of Medicine, New Haven, USA
| | - Mytien Nguyen
- Department of Immunobiology, Yale School of Medicine, New Haven, USA
| | - Syeda Andleeb Zehra
- Department of Internal Medicine, Section of Digestive Diseases, Yale Liver Center, Yale School of Medicine, New Haven, USA
| | - Shakila Afroz Taleb
- Department of Internal Medicine, Section of Digestive Diseases, Yale Liver Center, Yale School of Medicine, New Haven, USA
| | - Tory Bauer-Pisani
- Department of Internal Medicine, Section of Digestive Diseases, Yale Liver Center, Yale School of Medicine, New Haven, USA
| | - Noah Palm
- Department of Immunobiology, Yale School of Medicine, New Haven, USA
| | - Mario Strazzabosco
- Department of Internal Medicine, Section of Digestive Diseases, Yale Liver Center, Yale School of Medicine, New Haven, USA
| | - Romina Fiorotto
- Department of Internal Medicine, Section of Digestive Diseases, Yale Liver Center, Yale School of Medicine, New Haven, USA.
| |
Collapse
|
32
|
Gormley A, Garavito-Duarte Y, Kim SW. The Role of Milk Oligosaccharides in Enhancing Intestinal Microbiota, Intestinal Integrity, and Immune Function in Pigs: A Comparative Review. BIOLOGY 2024; 13:663. [PMID: 39336091 PMCID: PMC11428639 DOI: 10.3390/biology13090663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 08/22/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024]
Abstract
The objective of this review was to identify the characteristics and functional roles of milk coproducts from human, bovine, and porcine sources and their impacts on the intestinal microbiota and intestinal immunity of suckling and nursery pigs. Modern pig production weans piglets at 3 to 4 weeks of age, which is earlier than pigs would naturally be weaned outside of artificial rearing. As a result, the immature intestines of suckling and nursery pigs face many challenges associated with intestinal dysbiosis, which can be caused by weaning stress or the colonization of the intestines by enteric pathogens. Milk oligosaccharides are found in sow milk and function as a prebiotic in the intestines of pigs as they cannot be degraded by mammalian enzymes and are thus utilized by intestinal microbial populations. The consumption of milk oligosaccharides during suckling and through the nursery phase can provide benefits to young pigs by encouraging the proliferation of beneficial microbial populations, preventing pathogen adhesion to enterocytes, and through directly modulating immune responses. Therefore, this review aims to summarize the specific functional components of milk oligosaccharides from human, bovine, and porcine sources, and identify potential strategies to utilize milk oligosaccharides to benefit young pigs through the suckling and nursery periods.
Collapse
Affiliation(s)
| | | | - Sung Woo Kim
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA; (A.G.); (Y.G.-D.)
| |
Collapse
|
33
|
Liu D, Mueed A, Ma H, Wang T, Su L, Wang Q. Pleurocinus ostreatus Polysaccharide Alleviates Cyclophosphamide-Induced Immunosuppression through the Gut Microbiome, Metabolome, and JAK/STAT1 Signaling Pathway. Foods 2024; 13:2679. [PMID: 39272445 PMCID: PMC11394083 DOI: 10.3390/foods13172679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
This study investigated the structure of Pleurocinus ostreatus polysaccharide (POP-1) and its effect on immunocompromised mice induced by cyclophosphamide (CY). Novel POP-1 was α- and β-glucopyranose, its molecular weight was 4.78 × 104 Da, it was mainly composed of glucose (88.9%), and it also contained galactose (2.97%), mannose (5.02%), fucose (0.3%), arabinose (0.21%), ribose (0.04%), galactose acid (0.17%), and glucose acid (1.45%). After POP-1 was administered to immunosuppressed mice, results showed that POP-1 increased the body weight, spleen, and thymus index and enhanced T lymphocyte proliferation in mice. POP-1 up-regulated the expression of CD3+, CD4+, and CD8+ lymphocytes and the ratio of CD4+/CD8+ in the mouse spleen to increase immunoglobulin (IgM, IgG, and IgA) and secrete cytokines (IL-2, IL-6, TNF-α, and IFN-γ) through activation of the JAK/STAT1 signaling pathway. Moreover, POP-1 remarkably reversed the gut-microbiota dysbiosis in immunosuppressed mice by increasing the abundance of Muribaculaceae, Lactobacillaceae, Blautia, and Ligilactobacillus and altered the fecal metabolites by increasing hexahomomethionine, DG(8:0/20:4(5Z, 8Z, 11Z, 14Z)-OH(20)/0:0, 2-((3-aminopyridin-2-yl)methylene)hydrazinecarbothioamide, Ginkgoic acid, and carboxy-ethyl-hydroxychroman, which is closely related to the immunity function. This study indicates that P. ostreatus polysaccharide effectively restores immunosuppressive activity and can be a functional ingredient in food and pharmaceutical products.
Collapse
Affiliation(s)
- Daiyao Liu
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Abdul Mueed
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - He Ma
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Tianci Wang
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Ling Su
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| | - Qi Wang
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun 130118, China
- College of Plant Protection, Jilin Agricultural University, Changchun 130118, China
| |
Collapse
|
34
|
Yu J, Tang H, Zhou N, Wang Z, Huang W, Chen Y, Wang D, Ni J, Lu J, Yao YF. Dietary L-arabinose-induced gut dysbiosis exacerbates Salmonella infection outcome. mSystems 2024; 9:e0052224. [PMID: 38980058 PMCID: PMC11334454 DOI: 10.1128/msystems.00522-24] [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: 04/10/2024] [Accepted: 06/18/2024] [Indexed: 07/10/2024] Open
Abstract
The gut microbiota is essential for providing colonization resistance against pathogens. Dietary sugars markedly shift the composition of the intestinal microbiota and alter host susceptibility to enteric infections. Here, we demonstrate the effect of L-arabinose on bacterial infection by using a mouse infection model with Salmonella enterica serovar Typhimurium (S. Tm). In the presence of microbiota, L-arabinose induces a dramatic expansion of Enterobacteriaceae, thereby decreasing the microbiota diversity and causing more severe systemic infection. However, L-arabinose supplementation does not alter the disease progression of Salmonella infection in a microbiota-depleted mouse model. More importantly, short-term supplementation of L-arabinose fails to exert anti-diabetic effects in Salmonella-infected hyperglycemia mice and still promotes infection. Overall, our work reveals that a high intake of dietary L-arabinose supports a bloom of Enterobacteriaceae in Salmonella-infected gut, further accelerating the process of systemic infection.IMPORTANCEL-arabinose is a promising natural sweetener and food additive for the regulation of hyperglycemia. Since diabetic subjects are more susceptible to infections, the safety of dietary L-arabinose in diabetic patients experiencing infection remains a concern. Our findings reveal that L-arabinose exacerbates Salmonella infection outcome by inducing gut microbiota dysbiosis in mice. High dietary intake of L-arabinose may be deleterious for diabetic individuals undergoing infection.
Collapse
Affiliation(s)
- Jingchen Yu
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huang Tang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Zhou
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zuoqiang Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqiu Huang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yana Chen
- Department of Pediatrics, Anhui Provincial Hospital, The First Affiliated Hospital of USTC, Hefei, Anhui, China
| | - Danni Wang
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinjing Ni
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Lu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-Feng Yao
- Laboratory of Bacterial Pathogenesis, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, China
| |
Collapse
|
35
|
Zhao X, Ying J, Wang Z, Wang Y, Li Z, Gu T, Liu S, Li Y, Liu B, Xin F, Wen B. In vitro digestive properties and the bioactive effect of walnut green husk on human gut microbiota. Front Microbiol 2024; 15:1392774. [PMID: 39224223 PMCID: PMC11367867 DOI: 10.3389/fmicb.2024.1392774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Walnut green husk (WGH) is a waste byproduct from walnut industry. However, it is not well-known about its bioactive effect on human gut health. Methods This study conducted in vitro digestion and fermentation experiments to study the bioactive effect of WGH. Results Microbial fermentation was the primary mechanism to efficiently release phenolics and flavonoids, resulting in more excellent antioxidant capacities (DPPH, ABTS, and FRAP assays), which reached a highest value with 14.82 ± 0.01 mg VcE/g DW, 3.47 ± 0.01 mmol TE/g DW, and 0.96 ± 0.07 mmol FeSO4·7H2O/g DW, respectively. The surface microstructure of WGH became loose and fragmented after microbial fermentation. The analytical results of gut microbiota demonstrated that WGH could significantly increase the relative abundance of Proteobacteria in phylum level and Phascolarctobacterium in genus level while certain pro-inflammatory bacteria (such as Clostridium_sensu_stricto_1, Dorea, Alistipes, and Bilophila) was inhibited. Additionally, 1,373 differential metabolites were identified and enriched in 283 KEGG pathways. Of which some metabolites were significantly upregulated including ferulic acid, chlorogenic acid, umbelliferone, scopolin, muricholic acid, and so forth. Discussion These results indicated that WGH could have antioxidant and anti-inflammatory activities in the human gut, which could improve the economical value of WGH in the food industry.
Collapse
Affiliation(s)
- Xiaolan Zhao
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Jiabao Ying
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Zhuochen Wang
- Institute of Agro-Products Processing, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Yulu Wang
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Zhen Li
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Tianyi Gu
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Shujun Liu
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Yulong Li
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
| | - Bing Liu
- Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin, China
| | - Fengjiao Xin
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Laboratory of Biomanufacturing and Food Engineering, Institute of Agricultural Product Processing and Nutritional Health, Chinese Academy of Agricultural Sciences (CAAS), Cangzhou, China
| | - Boting Wen
- Laboratory of Biomanufacturing and Food Engineering, Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing, China
- Laboratory of Biomanufacturing and Food Engineering, Institute of Agricultural Product Processing and Nutritional Health, Chinese Academy of Agricultural Sciences (CAAS), Cangzhou, China
| |
Collapse
|
36
|
Francis D, Sun F. A comparative analysis of mutual information methods for pairwise relationship detection in metagenomic data. BMC Bioinformatics 2024; 25:266. [PMID: 39143554 PMCID: PMC11323399 DOI: 10.1186/s12859-024-05883-7] [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: 08/02/2023] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Construction of co-occurrence networks in metagenomic data often employs correlation to infer pairwise relationships between microbes. However, biological systems are complex and often display qualities non-linear in nature. Therefore, the reliance on correlation alone may overlook important relationships and fail to capture the full breadth of intricacies presented in underlying interaction networks. It is of interest to incorporate metrics that are not only robust in detecting linear relationships, but non-linear ones as well. RESULTS In this paper, we explore the use of various mutual information (MI) estimation approaches for quantifying pairwise relationships in biological data and compare their performances against two traditional measures-Pearson's correlation coefficient, r, and Spearman's rank correlation coefficient, ρ. Metrics are tested on both simulated data designed to mimic pairwise relationships that may be found in ecological systems and real data from a previous study on C. diff infection. The results demonstrate that, in the case of asymmetric relationships, mutual information estimators can provide better detection ability than Pearson's or Spearman's correlation coefficients. Specifically, we find that these estimators have elevated performances in the detection of exploitative relationships, demonstrating the potential benefit of including them in future metagenomic studies. CONCLUSIONS Mutual information (MI) can uncover complex pairwise relationships in biological data that may be missed by traditional measures of association. The inclusion of such relationships when constructing co-occurrence networks can result in a more comprehensive analysis than the use of correlation alone.
Collapse
Affiliation(s)
- Dallace Francis
- Quantitative and Computational Biology Department, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Fengzhu Sun
- Quantitative and Computational Biology Department, University of Southern California, Los Angeles, CA, 90089, USA
| |
Collapse
|
37
|
Wang Z, Wang P, Zhou Y, Zhuang S. Quercetin Supplementation Improves Intestinal Digestive and Absorptive Functions and Microbiota in Rats Fed Protein-Oxidized Soybean Meal: Transcriptomics and Microbiomics Insights. Animals (Basel) 2024; 14:2326. [PMID: 39199859 PMCID: PMC11350852 DOI: 10.3390/ani14162326] [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: 06/22/2024] [Revised: 08/01/2024] [Accepted: 08/10/2024] [Indexed: 09/01/2024] Open
Abstract
To clarify the nutritional mechanisms of quercetin mitigation in the digestive and absorptive functions in rats fed protein-oxidized soybean meal, 48 three-week-old male SD rats were randomly allocated into a 2 × 2 factorial design with two soybean meal types (fresh soybean meal or protein-oxidized soybean meal) and two quercetin levels (0 or 400 mg/kg) for a 28-day feeding trial. The protein-oxidized soybean meal treatment decreased (p < 0.05) the relative weights of the pancreas, stomach, and cecum, duodenal villus height, pancreatic and jejunal lipase activities, apparent ileal digestibility of amino acids, and apparent total tract digestibility of dry matter, crude protein, and ether extract. The supplementation of quercetin in the protein-oxidized soybean meal diet reversed (p < 0.05) the decreases in the duodenal length, ileal villus height, lipase activity, apparent ileal digestibility of amino acids, and apparent total tract digestibility of dry matter, crude protein, and ether extract. Transcriptomics revealed that the "alanine transport" and "lipid digestion and absorption" pathways were downregulated by the protein-oxidized soybean meal compared with fresh soybean meal, while the "basic amino acid transmembrane transporter activity" and "lipid digestion and absorption" pathways were upregulated by the quercetin supplementation. Microbiomics revealed that the protein-oxidized soybean meal increased the protein-degrading and inflammation-triggering bacteria in the cecum, while the relative abundances of beneficial bacteria were elevated by the quercetin supplementation.
Collapse
Affiliation(s)
| | | | | | - Su Zhuang
- College of Animal Science and Technology, Nanjing Agricultural University, No. 1 Weigang, Nanjing 210095, China; (Z.W.); (P.W.); (Y.Z.)
| |
Collapse
|
38
|
De Bruyn F, James K, Cottenet G, Dominick M, Katja J. Combining Bifidobacterium longum subsp. infantis and human milk oligosaccharides synergistically increases short chain fatty acid production ex vivo. Commun Biol 2024; 7:943. [PMID: 39098939 PMCID: PMC11298527 DOI: 10.1038/s42003-024-06628-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 07/24/2024] [Indexed: 08/06/2024] Open
Abstract
To enhance health benefits, a probiotic can be co-administered with a metabolizable prebiotic forming a synergistic synbiotic. We assessed the synergies resulting from combining Bifidobacterium longum subsp. infantis LMG 11588 and an age-adapted blend of six human milk oligosaccharides (HMOs) in ex vivo colonic incubation bioreactors seeded with fecal background microbiota from infant and toddler donors. When HMOs were combined with B. infantis LMG 11588, they were rapidly and completely consumed. This resulted in increased short chain fatty acid (SCFA) production compared to the summed SCFA production from individual ingredients (synergy). Remarkably, HMOs were partially consumed for specific infant donors in the absence of B. infantis LMG 11588, yet all donors showed increased SCFA production upon B. infantis LMG 11588 supplementation. We found specific bacterial taxa associated with the differential response pattern to HMOs. Our study shows the importance of carefully selecting pre- and probiotic into a synergistic synbiotic that could benefit infants.
Collapse
Affiliation(s)
- Florac De Bruyn
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland.
| | - Kieran James
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland
| | - Geoffrey Cottenet
- Nestlé Institute of Food Safety and Analytical Science, Nestlé Research, Route du Jorat 57, CH-1000, Lausanne, Switzerland
| | - Maes Dominick
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland
| | - Johnson Katja
- Nestlé Research and Development, Nestléstrasse 3, CH-3510, Konolfingen, Switzerland
| |
Collapse
|
39
|
Alexandrescu L, Nicoara AD, Tofolean DE, Herlo A, Nelson Twakor A, Tocia C, Trandafir A, Dumitru A, Dumitru E, Aftenie CF, Preotesoiu I, Dina E, Tofolean IT. Healing from Within: How Gut Microbiota Predicts IBD Treatment Success-A Systematic Review. Int J Mol Sci 2024; 25:8451. [PMID: 39126020 PMCID: PMC11313389 DOI: 10.3390/ijms25158451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Recent research indicates that the microbiome has a significant impact on the progression of inflammatory bowel disease (IBD) and that creating therapies that change its composition could positively impact the outcomes of IBD treatment. This review summarizes the results of extensive studies that examined IBD patients undergoing several therapies, including anti-TNF medication, vedolizumab, ustekinumab, probiotics, and fecal microbiota transplantation (FMT), and the alterations in their gut microbiota's composition and function. The objective was to investigate the variety and effectiveness of microbial species in order to discover new biomarkers or therapeutic targets that could improve the outcome of treatment for these patients. This research aimed to offer useful insights into personalized medicine techniques for managing IBD. Beneficial bacteria such as Faecalibacterium prausnitzii and Roseburia have been consistently linked to favorable clinical outcomes, whereas pathogenic bacteria such as Escherichia coli and Clostridioides difficile are associated with worsening disease conditions. Although many studies have examined the role of gut microbiota in IBD, there is still a need for more targeted research on the connection between specific microbial communities and treatment outcomes. This study sought to address this gap by exploring the intricate relationship between the gut microbiota composition and the effectiveness of IBD medications.
Collapse
Affiliation(s)
- Luana Alexandrescu
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
| | - Alina Doina Nicoara
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
- Internal Medicine Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Doina Ecaterina Tofolean
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
- Pneumology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Alexandra Herlo
- Department XIII, Discipline of Infectious Diseases, “Victor Babes” University of Medicine and Pharmacy Timisoara, 2 Eftimie Murgu Square, 300041 Timisoara, Romania;
| | - Andreea Nelson Twakor
- Internal Medicine Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania;
| | - Cristina Tocia
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
| | - Anamaria Trandafir
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
| | - Andrei Dumitru
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (E.D.); (I.T.T.)
| | - Eugen Dumitru
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
- Academy of Romanian Scientist, 3 Ilfov Street, 050044 Bucharest, Romania
| | - Cristian Florentin Aftenie
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
| | - Ionela Preotesoiu
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
| | - Elena Dina
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (E.D.); (I.T.T.)
| | - Ioan Tiberiu Tofolean
- Gastroenterology Department, “Sf. Apostol Andrei” Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania; (L.A.); (C.T.); (A.D.); (E.D.); (E.D.); (I.T.T.)
- Medicine Faculty, “Ovidius” University of Constanta, 1 Universitatii Street, 900470 Constanta, Romania; (D.E.T.); (A.T.); (C.F.A.); (I.P.)
| |
Collapse
|
40
|
Moncada E, Bulut N, Li S, Johnson T, Hamaker B, Reddivari L. Dietary Fiber's Physicochemical Properties and Gut Bacterial Dysbiosis Determine Fiber Metabolism in the Gut. Nutrients 2024; 16:2446. [PMID: 39125327 PMCID: PMC11314264 DOI: 10.3390/nu16152446] [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: 06/25/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
A fiber-rich diet is considered beneficial for gut health. An inflamed gut with a dysbiotic bacterial community can result in altered fiber metabolism depending on the fiber's physicochemical properties. This study examined the effect of fiber's physicochemical properties on fiber fermentation in the presence of healthy and colitis-associated bacteria. Sixteen fibers with different levels of solubility, complexity, and fermentation rate were used in in vitro fermentation with healthy human gut bacteria. Resistant maltodextrins (RMD), pectin (HMP), inulin (ChIn), and wheat bran (WB) were selected for fermentation using ulcerative colitis (UC)-associated bacteria to assess bacterial dysbiosis effect. UC-associated gut microbiota showed a significant reduction in α-and β-diversity indices compared to healthy-associated microbiota. The differences in the gut microbiota composition and diversity between the donors resulted in decreased fermentation rates with UC-associated bacteria. Fiber fermentation metabolites, short-chain fatty acids (SCFA) and gas production were significantly lower in the presence of UC-associated bacteria for all four fibers tested. Overall, we conclude that dietary fiber properties and microbial dysbiosis are influential in fiber fermentation and metabolite production in the gut.
Collapse
Affiliation(s)
- Edward Moncada
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (E.M.); (N.B.); (S.L.); (B.H.)
| | - Nuseybe Bulut
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (E.M.); (N.B.); (S.L.); (B.H.)
| | - Shiyu Li
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (E.M.); (N.B.); (S.L.); (B.H.)
| | - Timothy Johnson
- Department of Animal Science, Purdue University, West Lafayette, IN 47907, USA;
| | - Bruce Hamaker
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (E.M.); (N.B.); (S.L.); (B.H.)
| | - Lavanya Reddivari
- Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; (E.M.); (N.B.); (S.L.); (B.H.)
| |
Collapse
|
41
|
Xu H, Luo Y, Li Q, Zhu H. Acupuncture influences multiple diseases by regulating gut microbiota. Front Cell Infect Microbiol 2024; 14:1371543. [PMID: 39040602 PMCID: PMC11260648 DOI: 10.3389/fcimb.2024.1371543] [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: 01/16/2024] [Accepted: 06/13/2024] [Indexed: 07/24/2024] Open
Abstract
Acupuncture, an important green and side effect-free therapy in traditional Chinese medicine, is widely use both domestically and internationally. Acupuncture can interact with the gut microbiota and influence various diseases, including metabolic diseases, gastrointestinal diseases, mental disorders, nervous system diseases, and other diseases. This review presents a thorough analysis of these interactions and their impacts and examines the alterations in the gut microbiota and the potential clinical outcomes following acupuncture intervention to establish a basis for the future utilization of acupuncture in clinical treatments.
Collapse
Affiliation(s)
- Huimin Xu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yingzhe Luo
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qiaoqi Li
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Zhu
- Department of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
42
|
Guan Y, Zhao S, Li J, Zhang W, Guo Z, Luo Y, Jiang X, Li J, Liu J, Chen X, Zhao Z, Zhang Z. Insights from metagenomics into gut microbiome associated with acute coronary syndrome therapy. Front Microbiol 2024; 15:1369478. [PMID: 39035441 PMCID: PMC11258018 DOI: 10.3389/fmicb.2024.1369478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024] Open
Abstract
Acute coronary syndrome (ACS) is a predominant cause of mortality, and the prompt and precise identification of this condition is crucial to minimize its impact. Recent research indicates that gut microbiota is associated with the onset, progression, and treatment of ACS. To investigate its role, we sequenced the gut microbiota of 38 ACS patients before and after percutaneous coronary intervention and statin therapy at three time points, examining differential species and metabolic pathways. We observed a decrease in the abundance of Parabacteroides, Escherichia, and Blautia in patients after treatment and an increase in the abundance of Gemalla, Klebsiella variicola, Klebsiella pneumoniae, and others. Two pathways related to sugar degradation were more abundant in patients before treatment, possibly correlated with disorders of sugar metabolism and risk factors, such as hyperglycemia, insulin resistance, and insufficient insulin secretion. Additionally, seven pathways related to the biosynthesis of vitamin K2 and its homolog were reduced after treatment, suggesting that ACS patients may gradually recover after therapy. The gut microbiota of patients treated with different statins exhibited notable differences after treatment. Rosuvastatin appeared to promote the growth of anti-inflammatory bacteria while reducing pro-inflammatory bacteria, whereas atorvastatin may have mixed effects on pro-inflammatory and anti-inflammatory bacteria while increasing the abundance of Bacteroides. Our research will provide valuable insights and enhance comprehension of ACS, leading to better patient diagnosis and therapy.
Collapse
Affiliation(s)
- Yuee Guan
- Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Shuru Zhao
- Shenzhen Byoryn Technology Co., Ltd., Shenzhen, China
| | - Jing Li
- University of Science and Technology of China, Hefei, China
| | - Wenqian Zhang
- Department of Cardiology, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi'an, China
- Department of Computer Science, City University of Hong Kong, Kowloon Tong, China
| | - Zhonghao Guo
- School of Medicine, Xi’an Jiaotong University, Xi’an, China
| | - Yi Luo
- Department of Cardiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaofei Jiang
- Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Jun Li
- Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Jianxiong Liu
- Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Xi Chen
- Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Zicheng Zhao
- Shenzhen Byoryn Technology Co., Ltd., Shenzhen, China
| | - Zhe Zhang
- Department of Cardiology, Zhuhai People’s Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
- Department of Cardiology, The Zhuhai National Hi-tech Industrial Development District People’s Hospital (Zhuhai People’s Hospital Medical Group, High-tech Zone), Zhuhai, China
| |
Collapse
|
43
|
Hwang CH, Kim SH, Lee CH. Bacterial Growth Modulatory Effects of Two Branched-Chain Hydroxy Acids and Their Production Level by Gut Microbiota. J Microbiol Biotechnol 2024; 34:1314-1321. [PMID: 38938006 PMCID: PMC11239411 DOI: 10.4014/jmb.2404.04009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 04/25/2024] [Indexed: 06/29/2024]
Abstract
Branched-chain hydroxy acids (BCHAs), produced by lactic acid bacteria, have recently been suggested as bioactive compounds contributing to the systemic metabolism and modulation of the gut microbiome. However, the relationship between BCHAs and gut microbiome remains unclear. In this study, we investigated the effects of BCHAs on the growth of seven different families in the gut microbiota. Based on in vitro screening, both 2-hydroxyisovaleric acid (HIVA) and 2-hydroxyisocaproic acid (HICA) stimulated the growth of Lactobacillaceae and Bifidobacteriaceae, with HIVA showing a significant growth promotion. Additionally, we observed not only the growth promotion of probiotic Lactobacillaceae strains but also growth inhibition of pathogenic B. fragilis in a dosedependent manner. The production of HIVA and HICA varied depending on the family of the gut microbiota and was relatively high in case of Lactobacillaceae and Lachnosporaceae. Furthermore, HIVA and HICA production by each strain positively correlated with their growth variation. These results demonstrated gut microbiota-derived BCHAs as active metabolites that have bacterial growth modulatory effects. We suggest that BCHAs can be utilized as active metabolites, potentially contributing to the treatment of diseases associated with gut dysbiosis.
Collapse
Affiliation(s)
- Chan Hyuk Hwang
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| | - Su-Hyun Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
- MetaMass Corp., Seoul 05029, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
- MetaMass Corp., Seoul 05029, Republic of Korea
| |
Collapse
|
44
|
Panyod S, Wu WK, Chang CT, Wada N, Ho HC, Lo YL, Tsai SP, Chen RA, Huang HS, Liu PY, Chen YH, Chuang HL, Shen TCD, Tang SL, Ho CT, Wu MS, Sheen LY. Common dietary emulsifiers promote metabolic disorders and intestinal microbiota dysbiosis in mice. Commun Biol 2024; 7:749. [PMID: 38902371 PMCID: PMC11190199 DOI: 10.1038/s42003-024-06224-3] [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: 03/24/2023] [Accepted: 04/22/2024] [Indexed: 06/22/2024] Open
Abstract
Dietary emulsifiers are linked to various diseases. The recent discovery of the role of gut microbiota-host interactions on health and disease warrants the safety reassessment of dietary emulsifiers through the lens of gut microbiota. Lecithin, sucrose fatty acid esters, carboxymethylcellulose (CMC), and mono- and diglycerides (MDG) emulsifiers are common dietary emulsifiers with high exposure levels in the population. This study demonstrates that sucrose fatty acid esters and carboxymethylcellulose induce hyperglycemia and hyperinsulinemia in a mouse model. Lecithin, sucrose fatty acid esters, and CMC disrupt glucose homeostasis in the in vitro insulin-resistance model. MDG impairs circulating lipid and glucose metabolism. All emulsifiers change the intestinal microbiota diversity and induce gut microbiota dysbiosis. Lecithin, sucrose fatty acid esters, and CMC do not impact mucus-bacterial interactions, whereas MDG tends to cause bacterial encroachment into the inner mucus layer and enhance inflammation potential by raising circulating lipopolysaccharide. Our findings demonstrate the safety concerns associated with using dietary emulsifiers, suggesting that they could lead to metabolic syndromes.
Collapse
Affiliation(s)
- Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Kai Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC
- Bachelor Program of Biotechnology and Food Nutrition, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Ting Chang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Naohisa Wada
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Han-Chen Ho
- Department of Anatomy, Tzu Chi University, Hualien, Taiwan, ROC
| | - Yi-Ling Lo
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Sing-Ping Tsai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Huai-Syuan Huang
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC
| | - Po-Yu Liu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan, ROC
| | - Yi-Hsun Chen
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan, ROC
| | - Ting-Chin David Shen
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sen-Lin Tang
- Biodiversity Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ, USA
| | - Ming-Shiang Wu
- Department of Internal Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, ROC.
- Center for Food and Biomolecules, National Taiwan University, Taipei, Taiwan, ROC.
- National Center for Food Safety Education and Research, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
45
|
Hong D, Kim HK, Yang W, Yoon C, Kim M, Yang CS, Yoon S. Integrative analysis of single-cell RNA-seq and gut microbiome metabarcoding data elucidates macrophage dysfunction in mice with DSS-induced ulcerative colitis. Commun Biol 2024; 7:731. [PMID: 38879692 PMCID: PMC11180211 DOI: 10.1038/s42003-024-06409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 06/03/2024] [Indexed: 06/19/2024] Open
Abstract
Ulcerative colitis (UC) is a significant inflammatory bowel disease caused by an abnormal immune response to gut microbes. However, there are still gaps in our understanding of how immune and metabolic changes specifically contribute to this disease. Our research aims to address this gap by examining mouse colons after inducing ulcerative colitis-like symptoms. Employing single-cell RNA-seq and 16 s rRNA amplicon sequencing to analyze distinct cell clusters and microbiomes in the mouse colon at different time points after induction with dextran sodium sulfate. We observe a significant reduction in epithelial populations during acute colitis, indicating tissue damage, with a partial recovery observed in chronic inflammation. Analyses of cell-cell interactions demonstrate shifts in networking patterns among different cell types during disease progression. Notably, macrophage phenotypes exhibit diversity, with a pronounced polarization towards the pro-inflammatory M1 phenotype in chronic conditions, suggesting the role of macrophage heterogeneity in disease severity. Increased expression of Nampt and NOX2 complex subunits in chronic UC macrophages contributes to the inflammatory processes. The chronic UC microbiome exhibits reduced taxonomic diversity compared to healthy conditions and acute UC. The study also highlights the role of T cell differentiation in the context of dysbiosis and its implications in colitis progression, emphasizing the need for targeted interventions to modulate the inflammatory response and immune balance in colitis.
Collapse
Affiliation(s)
- Dawon Hong
- RNA Cell Biology Laboratory, Graduate Department of Bioconvergence Engineering, Dankook University, Yongin, Republic of Korea
| | - Hyo Keun Kim
- Dept of Molecular and Life Science and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan-si, Korea
| | - Wonhee Yang
- Department of AI-based Convergence, Dankook University, Yongin, Republic of Korea
| | - Chanjin Yoon
- Dept of Molecular and Life Science and Institute of Natural Science and Technology, Hanyang University, Ansan-si, Korea
| | - Minsoo Kim
- Department of Computer Science, College of SW Convergence, Dankook University, Yongin, Republic of Korea
| | - Chul-Su Yang
- Dept of Medicinal and Life Science and Center for Bionano Intelligence Education and Research, Hanyang University, Ansan-si, Korea.
| | - Seokhyun Yoon
- Department of Electronics & Electrical Engineering, College of Engineering, Dankook University, Yongin, Republic of Korea.
| |
Collapse
|
46
|
Modrackova N, Horvathova K, Mekadim C, Splichal I, Splichalova A, Amin A, Mrazek J, Vlkova E, Neuzil-Bunesova V. Defined Pig Microbiota Mixture as Promising Strategy against Salmonellosis in Gnotobiotic Piglets. Animals (Basel) 2024; 14:1779. [PMID: 38929398 PMCID: PMC11200913 DOI: 10.3390/ani14121779] [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: 05/14/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Probiotics are a potential strategy for salmonellosis control. A defined pig microbiota (DPM) mixture of nine bacterial strains previously exhibited probiotic and anti-Salmonella properties in vitro. Therefore, we evaluated its gut colonization ability and protection effect against S. typhimurium LT2-induced infection in the gnotobiotic piglet model. The DPM mixture successfully colonized the piglet gut and was stable and safe until the end of the experiment. The colon was inhabited by about 9 log CFU g-1 with a significant representation of bifidobacteria and lactobacilli compared to ileal levels around 7-8 log CFU g-1. Spore-forming clostridia and bacilli seemed to inhabit the environment only temporarily. The bacterial consortium contributed to the colonization of the gut at an entire length. The amplicon profile analysis supported the cultivation trend with a considerable representation of lactobacilli with bacilli in the ileum and bifidobacteria with clostridia in the colon. Although there was no significant Salmonella-positive elimination, it seems that the administered bacteria conferred the protection of infected piglets because of the slowed delayed infection manifestation without translocations of Salmonella cells to the blood circulation. Due to its colonization stability and potential protective anti-Salmonella traits, the DPM mixture has promising potential in pig production applications. However, advanced immunological tests are needed.
Collapse
Affiliation(s)
- Nikol Modrackova
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic; (K.H.); (A.A.); (E.V.); (V.N.-B.)
| | - Kristyna Horvathova
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic; (K.H.); (A.A.); (E.V.); (V.N.-B.)
| | - Chahrazed Mekadim
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (C.M.); (J.M.)
| | - Igor Splichal
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Doly 183, 549 22 Novy Hradek, Czech Republic; (I.S.); (A.S.)
| | - Alla Splichalova
- Laboratory of Gnotobiology, Institute of Microbiology, Czech Academy of Sciences, Doly 183, 549 22 Novy Hradek, Czech Republic; (I.S.); (A.S.)
| | - Ahmad Amin
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic; (K.H.); (A.A.); (E.V.); (V.N.-B.)
| | - Jakub Mrazek
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic; (C.M.); (J.M.)
| | - Eva Vlkova
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic; (K.H.); (A.A.); (E.V.); (V.N.-B.)
| | - Vera Neuzil-Bunesova
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Kamycka 129, 165 00 Prague, Czech Republic; (K.H.); (A.A.); (E.V.); (V.N.-B.)
| |
Collapse
|
47
|
Leung HKM, Lo EKK, Zhang F, Felicianna, Ismaiah MJ, Chen C, El-Nezami H. Modulation of Gut Microbial Biomarkers and Metabolites in Cancer Management by Tea Compounds. Int J Mol Sci 2024; 25:6348. [PMID: 38928054 PMCID: PMC11203446 DOI: 10.3390/ijms25126348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Cancers are causing millions of deaths and leaving a huge clinical and economic burden. High costs of cancer drugs are limiting their access to the growing number of cancer cases. The development of more affordable alternative therapy could reach more patients. As gut microbiota plays a significant role in the development and treatment of cancer, microbiome-targeted therapy has gained more attention in recent years. Dietary and natural compounds can modulate gut microbiota composition while providing broader and more accessible access to medicine. Tea compounds have been shown to have anti-cancer properties as well as modulate the gut microbiota and their related metabolites. However, there is no comprehensive review that focuses on the gut modulatory effects of tea compounds and their impact on reshaping the metabolic profiles, particularly in cancer models. In this review, the effects of different tea compounds on gut microbiota in cancer settings are discussed. Furthermore, the relationship between these modulated bacteria and their related metabolites, along with the mechanisms of how these changes led to cancer intervention are summarized.
Collapse
Affiliation(s)
- Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Marsena Jasiel Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
48
|
Arron HE, Marsh BD, Kell DB, Khan MA, Jaeger BR, Pretorius E. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: the biology of a neglected disease. Front Immunol 2024; 15:1386607. [PMID: 38887284 PMCID: PMC11180809 DOI: 10.3389/fimmu.2024.1386607] [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: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a chronic, debilitating disease characterised by a wide range of symptoms that severely impact all aspects of life. Despite its significant prevalence, ME/CFS remains one of the most understudied and misunderstood conditions in modern medicine. ME/CFS lacks standardised diagnostic criteria owing to variations in both inclusion and exclusion criteria across different diagnostic guidelines, and furthermore, there are currently no effective treatments available. Moving beyond the traditional fragmented perspectives that have limited our understanding and management of the disease, our analysis of current information on ME/CFS represents a significant paradigm shift by synthesising the disease's multifactorial origins into a cohesive model. We discuss how ME/CFS emerges from an intricate web of genetic vulnerabilities and environmental triggers, notably viral infections, leading to a complex series of pathological responses including immune dysregulation, chronic inflammation, gut dysbiosis, and metabolic disturbances. This comprehensive model not only advances our understanding of ME/CFS's pathophysiology but also opens new avenues for research and potential therapeutic strategies. By integrating these disparate elements, our work emphasises the necessity of a holistic approach to diagnosing, researching, and treating ME/CFS, urging the scientific community to reconsider the disease's complexity and the multifaceted approach required for its study and management.
Collapse
Affiliation(s)
- Hayley E. Arron
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Benjamin D. Marsh
- MRCPCH Consultant Paediatric Neurodisability, Exeter, Devon, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - M. Asad Khan
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | - Beate R. Jaeger
- Long COVID department, Clinic St Georg, Bad Aibling, Germany
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
49
|
Prins FM, Collij V, Groot HE, Björk JR, Swarte JC, Andreu-Sánchez S, Jansen BH, Fu J, Harmsen HJM, Zhernakova A, Lipsic E, van der Harst P, Weersma RK, Gacesa R. The gut microbiome across the cardiovascular risk spectrum. Eur J Prev Cardiol 2024; 31:935-944. [PMID: 38060843 DOI: 10.1093/eurjpc/zwad377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/03/2023] [Accepted: 12/05/2023] [Indexed: 06/04/2024]
Abstract
AIMS Despite treatment advancements, cardiovascular disease remains a leading cause of death worldwide. Identifying new targets is crucial for enhancing preventive and therapeutic strategies. The gut microbiome has been associated with coronary artery disease (CAD), however our understanding of specific changes during CAD development remains limited. We aimed to investigate microbiome changes in participants without clinically manifest CAD with different cardiovascular risk levels and in patients with ST-elevation myocardial infarction (STEMI). METHODS AND RESULTS In this cross-sectional study, we characterized the gut microbiome using metagenomics of 411 faecal samples from individuals with low (n = 130), intermediate (n = 130), and high (n = 125) cardiovascular risk based on the Framingham score, and STEMI patients (n = 26). We analysed diversity, and differential abundance of species and functional pathways while accounting for confounders including medication and technical covariates. Collinsella stercoris, Flavonifractor plautii, and Ruthenibacterium lactatiformans showed increased abundances with cardiovascular risk, while Streptococcus thermophilus was negatively associated. Differential abundance analysis revealed eight species and 49 predicted metabolic pathways that were differently abundant among the groups. In the gut microbiome of STEMI patients, there was a depletion of pathways linked to vitamin, lipid, and amino acid biosynthesis. CONCLUSION We identified four microbial species showing a gradual trend in abundance from low-risk individuals to those with STEMI, and observed differential abundant species and pathways in STEMI patients compared to those without clinically manifest CAD. Further investigation is warranted to gain deeper understanding of their precise role in CAD progression and potential implications, with the ultimate goal of identifying novel therapeutic targets.
Collapse
Affiliation(s)
- Femke M Prins
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Valerie Collij
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Hilde E Groot
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Johannes R Björk
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - J Casper Swarte
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Sergio Andreu-Sánchez
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Bernadien H Jansen
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
| | - Jingyuan Fu
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Pediatrics, Groningen, The Netherlands
| | - Hermie J M Harmsen
- University of Groningen, University Medical Center Groningen, Department of Medical Microbiology and Infection prevention, Groningen, The Netherlands
| | - Alexandra Zhernakova
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Erik Lipsic
- University of Groningen, University Medical Center Groningen, Department of Cardiology, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rinse K Weersma
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| | - Ranko Gacesa
- University of Groningen, University Medical Center Groningen, Department of Gastroenterology and Hepatology, Groningen, The Netherlands
- University of Groningen, University Medical Center Groningen, Department of Genetics, Groningen, The Netherlands
| |
Collapse
|
50
|
Filippo D, Guardone L, Listorti V, Elisabetta R. Microbiome in cancer: A comparative analysis between humans and dogs. Vet J 2024; 305:106145. [PMID: 38788999 DOI: 10.1016/j.tvjl.2024.106145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 04/22/2024] [Accepted: 05/21/2024] [Indexed: 05/26/2024]
Abstract
Cancer is a major cause of death in humans and animals worldwide. While cancer survival rates have increased over recent decades, further research to identify risk factors for the onset and progression of disease, and safe and highly efficacious treatments, is needed. Spontaneous tumours in pets represent an excellent model for neoplastic disease in humans. In this regard, dogs are an interesting species, as the divergence between the dog and human genome is low, humans and dogs have important similarities in the development and functioning of the immune system, and both species often share the same physical environment. There is also a higher homology between the canine and human microbiome than murine model. This review aims to describe and organize recently published information on canine microbiome assemblages and their relationship with the onset and progression of colorectal cancer, breast cancer and lymphoma, and to compare this with human disease. In both species, dysbiosis can induce variations in the gut microbiota that strongly influence shifts in status between health and disease. This can produce an inflammatory state, potentially leading to neoplasia, especially in the intestine, thus supporting canine studies in comparative oncology. Intestinal dysbiosis can also alter the efficacy and side effects of cancer treatments. Fewer published studies are available on changes in the relevant microbiomes in canine lymphoma and mammary cancer, and further research in this area could improve our understanding of the role of microbiota in the development of these cancers.
Collapse
Affiliation(s)
- Dell'Anno Filippo
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy; Department of Public Health, Experimental and Forensic Medicine, Section of Biostatistics and Clinical Epidemiology, University of Pavia, Pavia, Italy
| | - Lisa Guardone
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy
| | - Valeria Listorti
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy
| | - Razzuoli Elisabetta
- National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle d'Aosta, Genova 16129, Italy.
| |
Collapse
|