1
|
He F, Zheng Y, Elsabagh M, Fan K, Zha X, Zhang B, Wang M, Zhang H. Gut microbiota modulate intestinal inflammation by endoplasmic reticulum stress-autophagy-cell death signaling axis. J Anim Sci Biotechnol 2025; 16:63. [PMID: 40312439 PMCID: PMC12046778 DOI: 10.1186/s40104-025-01196-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
The intestinal tract, a complex organ responsible for nutrient absorption and digestion, relies heavily on a balanced gut microbiome to maintain its integrity. Disruptions to this delicate microbial ecosystem can lead to intestinal inflammation, a hallmark of inflammatory bowel disease (IBD). While the role of the gut microbiome in IBD is increasingly recognized, the underlying mechanisms, particularly those involving endoplasmic reticulum (ER) stress, autophagy, and cell death, remain incompletely understood. ER stress, a cellular response to various stressors, can trigger inflammation and cell death. Autophagy, a cellular degradation process, can either alleviate or exacerbate ER stress-induced inflammation, depending on the specific context. The gut microbiome can influence both ER stress and autophagy pathways, further complicating the interplay between these processes. This review delves into the intricate relationship between ER stress, autophagy, and the gut microbiome in the context of intestinal inflammation. By exploring the molecular mechanisms underlying these interactions, we aim to provide a comprehensive theoretical framework for developing novel therapeutic strategies for IBD. A deeper understanding of the ER stress-autophagy axis, the gut microbial-ER stress axis, and the gut microbial-autophagy axis may pave the way for targeted interventions to restore intestinal health and mitigate the impact of IBD.
Collapse
Affiliation(s)
- Feiyang He
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömermer Halisdemir University, Nigde, 51240, Turkey
| | - Kewei Fan
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, 832000, P. R. China
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China.
| |
Collapse
|
2
|
Romero-Ferreiro V, García-Fernández L, Biscaia JM, Romero C, González-Soltero R, De la Fuente M, Álvarez-Mon MA, Wynn R, Rodriguez-Jimenez R. Effect of probiotics on C-reactive protein levels in schizophrenia: Evidence from a systematic review and meta-analysis. Complement Ther Med 2025; 89:103126. [PMID: 39798817 DOI: 10.1016/j.ctim.2025.103126] [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: 10/16/2024] [Revised: 12/21/2024] [Accepted: 01/02/2025] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Inflammatory markers play a pivotal role in schizophrenia, as they provide insight into the neuroinflammatory processes occurring in the context of the disorder. Elevated levels of these markers, particularly C-reactive protein (CRP), can indicate an underlying immune system dysregulation, potentially influencing symptom severity and progression. Recognizing these markers has led to investigate the use of probiotics as an adjuvant to improve the treatment of schizophrenia. The main objective of this study is to rigorously evaluate the efficacy of probiotics in reducing plasma levels of CRP in patients with schizophrenia. METHODS A systematic search and meta-analysis were conducted to review randomized clinical trials following the PRISMA methodology. The following search strategy ((SCHIZO* OR PSYCHOTIC OR PSYCHOSES) AND (PROBIOTIC* OR BIFIDOBACTER* OR LACTOBACILL*)) was used for searching publications between June-December 2024 on the PubMed, Web of Science, and APA PsycINFO databases. Individual study quality was assessed with the Cochrane risk of bias (RoB2) and the certainty of total evidence was assessed with the GRADE system. RESULTS The primary outcome assessed was the impact of probiotic supplementation on plasma CRP levels. Out of 78 studies initially identified, 4 were finally included in the meta-analysis. Three out four studies found a significant reduction in high-sensitivity C-reactive protein levels in the supplemented compared with the placebo group. The pooled analysis revealed a significant reduction in CRP levels with probiotic supplementation, with a standardized mean difference (SMD) of -0.46, (95 % CI -0.719; -0.201; p = 0.001). CONCLUSIONS The synthesis and meta-analysis of available literature provide evidence for the potential role of probiotics in the reduction of serum CRP in schizophrenia compared with placebo. However, more clinical trials with better control of experimental design are needed before a clear recommendation as adjuvant therapy can be made.
Collapse
Affiliation(s)
- Verónica Romero-Ferreiro
- Universidad Europea de Madrid, Madrid, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; CIBERSAM-ISCIII (Biomedical Research Networking Centre for Mental Health), Spain.
| | - Lorena García-Fernández
- CIBERSAM-ISCIII (Biomedical Research Networking Centre for Mental Health), Spain; Clinical Medicine Department, Universidad Miguel Hernández, Alicante, Spain; Psychiatry Department, Hospital Universitario de San Juan, Alicante, Spain
| | | | - Carmen Romero
- Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain; CIBERESP/ISCIII (Biomedical Research Networking Centre for Epidemiology and PublicHealth/Carlos III Health Institute), Spain
| | | | - Mónica De la Fuente
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; Complutense University of Madrid (UCM), Madrid, Spain
| | - Miguel A Álvarez-Mon
- CIBERSAM-ISCIII (Biomedical Research Networking Centre for Mental Health), Spain; Department of Medicine and Medical Specialities, University of Alcala, Alcala de Henares, Spain; Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, Madrid, Spain; Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Rolf Wynn
- Department of Clinical Medicine, UiT The Arctic University of Norway, Tromsø, Norway; Department of Education, ICT and Learning, Østfold University College, Tromsø, Norway
| | - Roberto Rodriguez-Jimenez
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain; CIBERSAM-ISCIII (Biomedical Research Networking Centre for Mental Health), Spain; Complutense University of Madrid (UCM), Madrid, Spain
| |
Collapse
|
3
|
Di Mattia M, Sallese M, Lopetuso LR. The interplay between gut microbiota and the unfolded protein response: Implications for intestinal homeostasis preservation and dysbiosis-related diseases. Microb Pathog 2025; 200:107279. [PMID: 39761770 DOI: 10.1016/j.micpath.2025.107279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 11/28/2024] [Accepted: 01/03/2025] [Indexed: 01/11/2025]
Abstract
The unfolded protein response (UPR) is a complex intracellular signal transduction system that orchestrates the cellular response during Endoplasmic Reticulum (ER) stress conditions to reestablish cellular proteostasis. If, on one side, prolonged ER stress conditions can lead to programmed cell death and autophagy as a cytoprotective mechanism, on the other, unresolved ER stress and improper UPR activation represent a perilous condition able to trigger or exacerbate inflammatory responses. Notably, intestinal and immune cells experience ER stress physiologically due to their high protein secretory rate. Indeed, there is evidence of UPR's involvement in both physiological and pathological intestinal conditions, while less is known about its bidirectional interaction with gut microbiota. However, gut microbes and their metabolites can influence ER stress and UPR pathways, and, in turn, ER stress conditions can shape gut microbiota composition, with important implications for overall intestinal health. Thus, targeting UPR components is an intriguing strategy for treating ER stress-linked dysbiosis and diseases, particularly intestinal inflammation.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy.
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy; Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
4
|
Di Mattia M, Sallese M, Lopetuso LR. Unfolded protein response: An essential element of intestinal homeostasis and a potential therapeutic target for inflammatory bowel disease. Acta Physiol (Oxf) 2025; 241:e14284. [PMID: 39822064 DOI: 10.1111/apha.14284] [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: 06/11/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Different physiological and pathological situations can produce alterations in the cell's endoplasmic reticulum (ER), leading to a condition known as ER stress, which can trigger an intricate intracellular signal transduction system known as the unfolded protein response (UPR). UPR is primarily tailored to restore proteostasis and ER equilibrium; otherwise, if ER stress persists, it can cause programmed cell death as a cytoprotective mechanism and drive inflammatory processes. Therefore, since intestinal cells strongly rely on UPR for their biological functions and unbalanced UPR has been linked to inflammatory, metabolic, and immune disorders, here we discussed the role of the UPR within the intestinal tract, focusing on the UPR contribution to inflammatory bowel disease development. Importantly, we also highlighted the promising potential of UPR components as therapeutic targets for intestinal inflammatory diseases.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
5
|
Ataollahi F, Amirheidari B, Amirheidari Z, Ataollahi M. Clinical and mechanistic insights into biomedical application of Se-enriched probiotics and biogenic selenium nanoparticles. Biotechnol Lett 2025; 47:18. [PMID: 39826010 DOI: 10.1007/s10529-024-03559-z] [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: 06/14/2024] [Revised: 11/13/2024] [Accepted: 12/16/2024] [Indexed: 01/20/2025]
Abstract
Selenium is an essential element with various industrial and medical applications, hence the current considerable attention towards the genesis and utilization of SeNPs. SeNPs and other nanoparticles could be achieved via physical and chemical methods, but these methods would not only require expensive equipment and specific reagents but are also not always environment friendly. Biogenesis of SeNPs could therefore be considered as a less troublesome alternative, which opens an excellent window to the selenium and nanoparticles' world. bSeNPs have proved to exert higher bioavailability, lower toxicity, and broader utility as compared to their non-bio counterparts. Many researchers have reported promising features of bSeNP such as anti-oxidant and anti-inflammatory, in vitro and in vivo. Considering this, bSeNPs have been tried as effective agents for health disorders, especially as constituents of probiotics. This article briefly reviews selenium, selenium nanoparticles, Se-enriched probiotics, and bSeNPs' usage in an array of health disorders. Obviously, there are very many articles on bSeNPs, but we wanted to summarize studies on prominent bSeNPs features published in the twenty-first century. This review is hoped to give an outlook to researchers for their future investigations, ultimately serving better care of health disorders.
Collapse
Affiliation(s)
- Farshid Ataollahi
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Bagher Amirheidari
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Medical University Campus, Haft-Bagh Highway, Kerman, 76169-13555, Iran.
| | - Zohreh Amirheidari
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutical Sciences and Cosmetic Products Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahshid Ataollahi
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
6
|
Chantanaskul T, Patumcharoenpol P, Roytrakul S, Kingkaw A, Vongsangnak W. Exploring Protein Functions of Gut Bacteriome and Mycobiome in Thai Infants Associated with Atopic Dermatitis Through Metaproteomic and Host Interaction Analysis. Int J Mol Sci 2024; 25:13533. [PMID: 39769296 PMCID: PMC11676981 DOI: 10.3390/ijms252413533] [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: 11/25/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Atopic dermatitis (AD), a prevalent allergic skin condition in children, has been closely associated with imbalances in the gut microbiome. To investigate these microbial alterations and their functional implications, we investigated protein expression, functions and interactions of the gut bacteriome and mycobiome as well as the human proteome in Thai infants with AD using integrative metaproteomic and host interaction analysis. As we observed, probiotic species, such as Lactobacillus acidophilus and Bacteroides salyersiae, were reduced in abundance in the AD group while key pathogenic bacteria and fungi, such as Streptococcus constellatus and Penicillium chrysogenum, increased in abundance. Additionally, the functional analysis of expressed proteins was enriched in response to stress and DNA repair in the bacteriome and ribosome biogenesis-related processes in the mycobiome of the AD group, potentially associated to increased reactive oxygen species (ROS), intestinal inflammation, fungal growth and microbial dysbiosis. Further, a protein-protein interactions (PPIs) network analysis incorporating the human proteome revealed 10 signature proteins related to stress and immune system processes associated with AD. Our findings propose the interactions of the key species and signature protein functions between the gut microbes and the human host in response to AD in Thai infants. To our knowledge, this study serves as the first framework for monitoring bacteriome-mycobiome-human gut studies associated with AD and other allergic diseases in infants.
Collapse
Affiliation(s)
- Thanawit Chantanaskul
- Genetic Engineering and Bioinformatics Program, Graduate School, Kasetsart University, Bangkok 10900, Thailand;
| | | | - Sittirak Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, 144 Thailand Science Park, Phaholyothin Road, Pathum Thani 12120, Thailand;
| | - Amornthep Kingkaw
- Interdisciplinary Graduate Program in Bioscience, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand;
| | - Wanwipa Vongsangnak
- Department of Zoology, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand;
- Omics Center for Agriculture, Bioresources, Food and Health, Kasetsart University (OmiKU), Bangkok 10900, Thailand
| |
Collapse
|
7
|
Kwoji ID, Okpeku M, Aiyegoro OA, Adeleke MA. Metabolic interactions of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 in co-culture: implications for multi-strain probiotics. J Appl Microbiol 2024; 135:lxae264. [PMID: 39510973 DOI: 10.1093/jambio/lxae264] [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: 02/18/2024] [Revised: 08/31/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
AIMS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 are potential probiotic bacteria. The mechanisms of enhanced benefits by muti-strain probiotics are yet fully understood. We elucidated the influence of co-culturing on the metabolite profiles of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 to decipher the impacts of co-culturing on metabolic interactions between the strains. METHODS AND RESULTS Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 were grown in single and co-cultures in defined media. Bacterial cell metabolites were extracted at the mid-stationary growth phase and analysed using two-dimensional gas chromatography-time-of-flight mass spectrometry (GC × GC-TOFMS). Mass-spectral data were preprocessed and analysed using unsupervised and supervised methods based on the group allocations. A total of 1387 metabolites were identified, with 18.31% significant metabolites (P < 0.05) and 10.17% differential metabolites (P < 0.05, variable importance on projection > 1). The differential metabolites identified include arabinofuranose, methyl-galactoside, N-acetylglutamic acid, phosphoric acid, and decanoic acid. The metabolites impacted carbohydrate and amino-sugar metabolism. CONCLUSION Co-culturing of Limosilactobacillus reuteri ZJ625 and Ligilactobacillus salivarius ZJ614 influenced the metabolite profiles of the strains and impacted metabolic/biosynthetic pathways, indicating cell-to-cell interactions between the strains.
Collapse
Affiliation(s)
- Iliya Dauda Kwoji
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Moses Okpeku
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| | - Olayinka Ayobami Aiyegoro
- Unit for Environmental Sciences and Management, Northwest University, Potchefstroom, Northwest 2520, South Africa
| | - Matthew Adekunle Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, 4090 Durban, South Africa
| |
Collapse
|
8
|
Ji M, Wang B, Xie J, Wang G, Yu E, Jiang P, Lu R, Tian J. Effects of low protein feed on hepato-intestinal health and muscle quality of grass carp (Ctenopharyngodon idellus). Comp Biochem Physiol B Biochem Mol Biol 2024; 273:110989. [PMID: 38759883 DOI: 10.1016/j.cbpb.2024.110989] [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: 12/13/2023] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
In this study, grass carp (33.28 ± 0.05 g) were fed three diets for 8 weeks: control (crude protein [CP] 30%, crude lipid [CL] 6%), low protein (LP; CP16%, CL6%), and low protein with high-fat (LPHF; CP16%, CL10%). The final body weight decreased in the LP and LPHF groups compared to the Control (P < 0.05). Liver triglycerides, total cholesterol, and nonesterified fatty acids were higher in the LP group than the Control, whereas these indexes in the LPHF group were higher than those in the LP group (P < 0.05). The LP group had intestinal barrier damage, while the LPHF group had a slight recovery. TNF-α, IL-8, and IL-1β content were lower in the LP group than in the Control (P < 0.05), and even higher in the LPHF group (P < 0.05). The expressions of endoplasmic reticulum stress-related genes Activating transcription factor 6 (ATF-6) and Glucose-regulated protein (GRP78) were higher in the LPHF group against the LP group (P < 0.05). The IL-1β and TNF-α content negatively correlated with intestinal Actinomycetes and Mycobacterium abundance (P < 0.05). The muscle fiber diameter was smaller in both the LP and LPHF groups than the control (P < 0.05), with the LP group showing metabolites related to protein digestion and absorption, and LPHF group exhibiting metabolites related to taste transmission. The results demonstrate reducing dietary protein affects growth, causing liver lipid accumulation, reduced enteritis response, and increased muscle tightness, while increasing fat content accelerates fat accumulation and inflammation.
Collapse
Affiliation(s)
- Mengmeng Ji
- College of Fisheries, Henan Normal University, Xinxiang, Henan 453007, China; Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China
| | - Binbin Wang
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China
| | - Jun Xie
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China
| | - Guangjun Wang
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China
| | - Ermeng Yu
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China
| | - Peng Jiang
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China
| | - Ronghua Lu
- College of Fisheries, Henan Normal University, Xinxiang, Henan 453007, China.
| | - Jingjing Tian
- Key Laboratory of Aquatic Animal Immune Technology of Guangdong Province, Pearl River Fisheries Research Institute Guangzhou, Guangdong 510380, China.
| |
Collapse
|
9
|
Li Z, Peng C, Sun Y, Zhang T, Feng C, Zhang W, Huang T, Yao G, Zhang H, He Q. Both viable Bifidobacterium longum subsp. infantis B8762 and heat-killed cells alleviate the intestinal inflammation of DSS-induced IBD rats. Microbiol Spectr 2024; 12:e0350923. [PMID: 38647334 PMCID: PMC11237488 DOI: 10.1128/spectrum.03509-23] [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: 09/28/2023] [Accepted: 03/21/2024] [Indexed: 04/25/2024] Open
Abstract
In view of the safety concerns of probiotics, more and more attention is paid to the beneficial effects of dead probiotics cells. Herein, we investigated and compared the alleviation effects of viable Bifidobacterium longum subsp. infantis B8762 (B. infantis B8762) and its heat-killed cells on dextran sodium sulfate (DSS)-induced inflammatory bowel disease (IBD) rats. Four groups of rats (n = 12 per group) were included: normal control, DSS-induced colitis rats without bacterial administration (DSS), DSS-induced colitis rats with viable B. infantis B8762 administration (VB8762), and DSS-induced colitis rats with dead B. infantis B8762 administration (DB8762). Our results showed that both VB8762 and DB8762 administration exerted significant protective effects on DSS-induced IBD rats, as evidenced by a reduction in mortality, disease activity index score, body weight loss, as well as decreased histology score, which were companied by a significant decrease in serum pro-inflammatory factors compared with DSS group, and a stronger effect on modulating the fecal microbiota alpha-diversity and beta-diversity compared with DSS group. Additionally, the fecal metabolome results showed that both VB8762 and DB8762 interventions indeed altered the fecal metabolome profile and related metabolic pathways of DSS-induced IBD rats. Therefore, given the alleviation effects on colitis, the DB8762 can be confirmed to be a postbiotic. Overall, our findings suggested that VB8762 and DB8762 had similar ability to alleviate IBD although with some differences. Due to the minimal safety concern of postbiotics, we propose that the postbiotic DB8762 could be a promising alternative to probiotics to be applied in the prevention and treatment of IBDs.IMPORTANCEInflammatory bowel disease (IBD) has emerged as a global disease because of the worldwide spread of western diets and lifestyles during industrialization. Up to now, many probiotic strains are used as a modulator of gut microbiota or an enhancer of gut barrier to alleviate or cure IBD. However, there are still many issues of using probiotics, which were needed to be concerned about, for instance, safety issues in certain groups like neonates and vulnerable populations, and the functional differences between viable and dead microorganisms. Therefore, it is of interest to investigate the beneficial effects of dead probiotics cells. The present study proved that both viable Bifidobacterium longum subsp. infantis B8762 and heat-killed cells could alleviate dextran sodium sulfate-induced colitis in rats. The findings help to support that some heat-killed probiotics cells can also exert relevant biological functions and can be used as a postbiotic.
Collapse
Affiliation(s)
- Zhaojie Li
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
- Qingdao Special Food Research Institute, Qingdao, China
| | - Chuantao Peng
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
- Qingdao Special Food Research Institute, Qingdao, China
| | - Yaru Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Tao Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Cuijiao Feng
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Weiqin Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Tian Huang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Guoqiang Yao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Qiuwen He
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
10
|
Chen P, Lv H, Du M, Liu W, Che C, Zhao J, Liu H. Bacillus subtilis HW2 enhances growth performance and alleviates gut injury via attenuation of endoplasmic reticulum stress and regulation of gut microbiota in broilers under necrotic enteritis challenge. Poult Sci 2024; 103:103661. [PMID: 38547540 PMCID: PMC11000119 DOI: 10.1016/j.psj.2024.103661] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/11/2024] Open
Abstract
This study investigated the effects of Bacillus subtilis HW2 on the growth performance, immune response, endoplasmic reticulum (ER) stress, and intestinal health in broilers with necrotic enteritis. Three hundred 1-day-old male Cobb 500 broilers (33.88 ± 2.34 g) were randomly allocated to 5 groups including non-infected control (NC group), basal diet + necrotic enteritis challenge (NE group), basal diet + 1 × 106 CFU/g B. subtilis HW2 + necrotic enteritis challenge (L-Pro group), basal diet + 5 × 106 CFU/g B. subtilis HW2 + necrotic enteritis challenge (M-Pro group), and basal diet + 1 × 107 CFU/g B. subtilis HW2 + necrotic enteritis challenge (H-Pro group), with 6 replicates per group. All broilers except NC group were orally given with sporulated coccidian oocysts at day 14 and Clostridium perfringens from days 19 to 21. Results showed that L-Pro and M-Pro groups improved growth performance and intestinal morphology in necrotic enteritis-challenged broilers, and L-Pro, M-Pro, and H-Pro groups improved intestinal barrier function and immune response and decreased ER stress in necrotic enteritis-challenged broilers. Analysis of the gut microbiota revealed that L-Pro group increased the abundances of Alistipes, Coprobacter, Barnesiella, and Limosilactobacillus, decreased Erysipelatoclostridium abundance on day 42 in necrotic enteritis-challenged broilers. M-Pro group increased Turicibacter abundance on day 28 and the abundances of Alistipes, Barnesiella, and Limosilactobacillus on day 42 in necrotic enteritis-challenged broilers. H-Pro group decreased Romboutsia abundance on day 28 and unidentified_Clostridia abundance on day 42 in necrotic enteritis-challenged broilers. Analysis of short-chain fatty acids (SCFAs) revealed higher isobutyric acid and isovaleric acid levels in L-Pro and M-Pro groups than NE group. Correlation analysis revealed the correlations between the biochemical parameters and gut microbiota as well as SCFAs, especially Romboutsia, Barnesiella, Coprobacter, isobutyric acid, and isovaleric acid. Overall, our results indicated that B. subtilis HW2 supplementation could ameliorate necrotic enteritis infection-induced gut injury. The optimal dietary supplementation dosage of Bacillus subtilis HW2 was 5 × 106 CFU/g.
Collapse
Affiliation(s)
- Peng Chen
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Huimin Lv
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Mengmeng Du
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Weiyong Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Chuanyan Che
- College of Animal Science and Technology, Anhui Science and Technology University, Fengyang, 233100, China
| | - Jinshan Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China
| | - Huawei Liu
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
11
|
Song X, Liu Y, Zhang X, Weng P, Zhang R, Wu Z. Role of intestinal probiotics in the modulation of lipid metabolism: implications for therapeutic treatments. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
12
|
Choy CT, Siu PLK, Zhou J, Wong CH, Lee YW, Chan HW, Tsui JCC, Lo CJY, Loo SKF, Tsui SKW. Improvements in Gut Microbiome Composition Predict the Clinical Efficacy of a Novel Synbiotics Formula in Children with Mild to Moderate Atopic Dermatitis. Microorganisms 2023; 11:2175. [PMID: 37764019 PMCID: PMC10536305 DOI: 10.3390/microorganisms11092175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease with a significant association with various type-2 inflammation-related comorbidities. Ongoing research suggests the crucial involvement of gut microbiome, especially in childhood onset AD, and hence, probiotics have emerged as a potential non-steroid-based therapeutics option to complement existing AD management plans. In order to delineate the impact of probiotics in the gut microbiome of pediatric AD patients from southern China, targeted 16S rRNA sequencing and thorough bioinformatic analysis were performed to analyze the gut microbiome profiles of 24 AD children after taking an orally administered novel synbiotics formula with triple prebiotics for 8 weeks. A notable improvement in Eczema Area and Severity Index (EASI) (p = 0.008) was observed after taking an 8-week course of probiotics, with no adverse effects observed. The relative abundances of key microbial drivers including Bacteroides fragilis and Lactobacillus acidophilus were significantly increased at week 8. We also found that the positive responsiveness towards an 8-week course of probiotics was associated with improvements in the gut microbiome profile with a higher relative abundance of probiotic species. Over-represented functional abundance pathways related to vitamin B synthesis and peptidoglycan recycling may imply the underlying mechanism. In summary, our study suggests how the gut microbial landscape shifts upon probiotic supplementation in AD children, and provides preliminary evidence to support targeted probiotic supplementation for the management of childhood AD.
Collapse
Affiliation(s)
- Chi Tung Choy
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Pui Ling Kella Siu
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Junwei Zhou
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Chi Ho Wong
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Yuk Wai Lee
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Ho Wang Chan
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | | | - Claudia Jun Yi Lo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
| | - Steven King Fan Loo
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
- Hong Kong Institute of Integrative Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
- Dermatology Centre, CUHK Medical Centre, The Chinese University of Hong Kong, Hong Kong
| | - Stephen Kwok Wing Tsui
- Microbiome Research Centre, BioMed Laboratory Company Limited, Hong Kong
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
- Centre for Microbial Genomics and Proteomics, The Chinese University of Hong Kong, Hong Kong
- Hong Kong Bioinformatics Centre, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
13
|
Woo JS, Hwang SH, Yang S, Lee KH, Lee YS, Choi JW, Park JS, Jhun J, Park SH, Cho ML. Lactobacillus acidophilus and propionate attenuate Sjögren's syndrome by modulating the STIM1-STING signaling pathway. Cell Commun Signal 2023; 21:135. [PMID: 37316856 DOI: 10.1186/s12964-023-01141-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 04/22/2023] [Indexed: 06/16/2023] Open
Abstract
BACKGROUND Sjögren's syndrome (SS) is an autoimmune disease characterized by inflammation of the exocrine gland. An imbalance of gut microbiota has been linked to SS. However, the molecular mechanism is unclear. We investigated the effects of Lactobacillus acidophilus (L. acidophilus) and propionate on the development and progression of SS in mouse model. METHODS We compared the gut microbiomes of young and old mice. We administered L. acidophilus and propionate up to 24 weeks. The saliva flow rate and the histopathology of the salivary glands were investigated, and the effects of propionate on the STIM1-STING signaling pathway were evaluated in vitro. RESULTS Lactobacillaceae and Lactobacillus were decreased in aged mice. SS symptoms were ameliorated by L. acidophilus. The abundance of propionate-producing bacterial was increased by L. acidophilus. Propionate ameliorated the development and progression of SS by inhibiting the STIM1-STING signaling pathway. CONCLUSIONS The findings suggest that Lactobacillus acidophilus and propionate have therapeutic potential for SS. Video Abstract.
Collapse
Affiliation(s)
- Jin Seok Woo
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sun-Hee Hwang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - SeungCheon Yang
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Kun Hee Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Yeon Su Lee
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jeong Won Choi
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jin-Sil Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JooYeon Jhun
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Sung-Hwan Park
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| | - Mi-La Cho
- Rheumatism Research Center, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Lab of Translational ImmunoMedicine, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
14
|
Hajj Hussein I, Dosh L, Al Qassab M, Jurjus R, El Masri J, Abi Nader C, Rappa F, Leone A, Jurjus A. Highlights on two decades with microbiota and inflammatory bowel disease from etiology to therapy. Transpl Immunol 2023; 78:101835. [PMID: 37030558 DOI: 10.1016/j.trim.2023.101835] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 04/08/2023]
Abstract
Inflammatory Bowel diseases (IBDs) constitute a complex panel of disorders characterized with chronic inflammation affecting the alimentary canal along with extra intestinal manifestations. Its exact etiology is still unknown; however, it seems to be the result of uncharacterized environmental insults in the intestine and their immunological consequences along with dysbiosis, in genetically predisposed individuals. It was the main target of our team since 2002 to explore the etiology of IBD and the related role of bacteria. For almost two decades, our laboratory, among others, has been involved in the reciprocal interaction between the host gastrointestinal lining and the homing microbiota. In the first decade, the attention of scientists focused on the possible role of enteropathogenic E. coli and its relationship to the mechanistic pathways involved in IBD induced in both rats and mice by chemicals like Iodoacetamide, Dextran Sodium Sulfate, Trinitrobenzene, thus linking microbial alteration to IBD pathology. A thorough characterization of the various models was the focus of research in addition to exploring how to establish an active homeostatic composition of the commensal microbiota, including its wide diversity by restoration of gut microbiota by probiotics and moving from dysbiosis to eubiosis. In the last six years and in order to effectively translate such findings into clinical practice, it was critical to explore their relationship to colorectal cancer CRC both in solid tumors and chemically induced CRC. It was also critical to explore the degree of intestinal dysbiosis and linking to IBD, CRC and diabetes. Remarkably, the active mechanistic pathways were proposed as well as the role of microbiota or bacterial metabolites involved. This review covers two decades of investigations in our laboratory and sheds light on the different aspects of the relationship between microbiota and IBD with an emphasis on dysbiosis, probiotics and the multiple mechanistic pathways involved.
Collapse
Affiliation(s)
- Inaya Hajj Hussein
- Oakland University William Beaumont School of Medicine, Rochester, MI, USA
| | - Laura Dosh
- Department of Anatomy, Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Biomedicine, Neuroscience and Advanced Diagnostics, Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Mohamad Al Qassab
- Department of Anatomy, Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Rosalyn Jurjus
- Department of Anatomy, Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Jad El Masri
- Department of Anatomy, Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Celine Abi Nader
- Department of Anatomy, Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Francesca Rappa
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Angelo Leone
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, Institute of Human Anatomy and Histology, University of Palermo, Palermo, Italy
| | - Abdo Jurjus
- Department of Anatomy, Biology and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
15
|
Dou Z, Qiu T, Ren Y, Wang X, Wen Q, Shen Y, Wu L, Han L, Jiang T, Xia X. Bilayer Silk Fibroin/Sodium Alginate Scaffold Delivered hUC-MSCs to Enhance Skin Scarless Healing and Hair Follicle Regeneration with the IRE1/XBP1 Pathway Inhibition. ACS Biomater Sci Eng 2023. [PMID: 37256923 DOI: 10.1021/acsbiomaterials.3c00059] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Efficient local delivery of mesenchymal stem cells (MSCs) is a decisive factor for their application in regeneration processes. Here, we prepared a biomimetic bilayer silk fibroin/sodium alginate (SF/SA) scaffold to deliver human umbilical mesenchymal stem cells (hUC-MSCs) for wound healing. An SA membrane was prepared by the casting method on the upper layer of the scaffold to simulate the dense epidermal structure. On the lower layer, porous materials simulating the loose structure of the dermis were formed by the freeze-drying method. In vitro, the scaffold was proven to have a high-density pore structure, good swelling property, and suitable degradation rate. The hUC-MSCs could survive on the scaffold for up to 14 days and maintain cell stemness for at least 7 days. In vivo, SF/SA scaffolds loaded with hUC-MSCs (M-SF/SA) were applied to full-thickness defect wounds and compared with the local injection of hUC-MSCs. The M-SF/SA group showed excellent therapeutic efficacy, characterized by induction of macrophage polarization, regulation of TGF-β expression and collagen components, and enhancement of vascular regeneration, thereby preventing scar formation and promoting hair follicle regeneration. Furthermore, the expression of endoplasmic reticulum stress markers IRE1, XBP1, and CHOP was inhibited significantly in M-SF/SA treatment. In conclusion, the bilayer SF/SA scaffold is an ideal delivery platform for hUC-MSCs, and the M-SF/SA system could locally promote scarless skin healing and hair follicle regeneration by alleviating the IRE1/XBP1 signal pathway.
Collapse
Affiliation(s)
- Zhaona Dou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Tong Qiu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Yimeng Ren
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Quan Wen
- Yinfeng Academy of Life Science, Yichang Laboratory of Hematopoietic Stem Cells, 1109 Gangxing Sanlu, Jinan, Shandong Province 250100, China
| | - Ying Shen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Lin Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Lei Han
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Tao Jiang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan, Hubei 430070, China
| | - Xinke Xia
- Institute of Life Science, Yinfeng Biological Group, High-tech Zone, Jinan, Shandong Province 250100, China
| |
Collapse
|
16
|
Tan YR, Shen SY, Shen HQ, Yi PF, Fu BD, Peng LY. The role of endoplasmic reticulum stress in regulation of intestinal barrier and inflammatory bowel disease. Exp Cell Res 2023; 424:113472. [PMID: 36634742 DOI: 10.1016/j.yexcr.2023.113472] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 01/08/2023] [Indexed: 01/11/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease involving the digestive tract, characterized by abdominal pain, diarrhea, rectal bleeding, and so on, which can make patients physically weakened and live difficultly. Although IBD has been recognized for many years, the pathogenesis of IBD has not yet been established and damage to intestinal barrier is thought to be closely associated with IBD. Intestinal barrier is an innate barrier that maintains the homeostasis of the intestinal environment and impedes pathogenic bacteria and toxins, and the endoplasmic reticulum (ER) has recently been found to be involved in maintaining the integrity of intestinal barrier. Endoplasmic reticulum stress (ERS) is a status of endoplasmic reticulum damaged when unfolded or misfolded proteins accumulate in excess of the degradation systematic clearance limit of the misfolded proteins. The regulation of ERS on protein folding synthesis and maintenance of cellular homeostasis is an important factor in influencing the integrity of the intestinal barrier. This paper mainly discusses the relationship between ERS and the intestinal barrier, aiming to understand the regulatory role of ERS on the intestinal barrier and the mechanism and to improve new solutions and notions for the treatment or prevention of IBD.
Collapse
Affiliation(s)
- Yue-Rong Tan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Si-Yang Shen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Hai-Qing Shen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Peng-Fei Yi
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Ben-Dong Fu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin, 130062, China
| | - Lu-Yuan Peng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, No. 5333 Xi'an Road, Changchun, Jilin, 130062, China.
| |
Collapse
|
17
|
Dou Z, Li B, Wu L, Qiu T, Wang X, Zhang X, Shen Y, Lu M, Yang Y. Probiotic-Functionalized Silk Fibroin/Sodium Alginate Scaffolds with Endoplasmic Reticulum Stress-Relieving Properties for Promoted Scarless Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2023; 15:6297-6311. [PMID: 36700526 DOI: 10.1021/acsami.2c17168] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Bioactive substances such as probiotics are becoming a research hotspot in the field of tissue regeneration due to their excellent regulatory functions. Here, we proposed to load Lactobacillus casei onto a bilayer silk fibroin/sodium alginate (SF/SA) scaffold to endow the scaffold with both antibacterial and regenerative properties. The performance of the scaffold was characterized systemically. The L. casei-loaded scaffolds (L-SF/SA) bring in lactic acid, which has antibacterial and wound healing properties. In vitro, the cell-free supernatant (CFS) of L. casei inhibited the transformation of fibroblasts to myofibroblasts and relieved the endoplasmic reticulum stress (ERS). In vivo, L-SF/SA accelerated the healing of infected wounds in SD rats. The L-SF/SA reduced the bacterial load, induced M2 polarization of macrophages, increased angiogenesis, regulated collagen ratio, and alleviated the ERS, thereby promoting scarless wound healing and increasing hair follicle regeneration. Therefore, probiotic-functionalized silk fibroin/alginate scaffolds showed potential in the infected wound healing.
Collapse
Affiliation(s)
- Zhaona Dou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Lin Wu
- Institute WUT-AMU, Wuhan University of Technology, Wuhan 430070, China
| | - Tong Qiu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Xueqiong Zhang
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Ying Shen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| | - Mengli Lu
- School of Chemistry, Chemical Engineering and Life Sciences, Wuhan University of Technology, Wuhan 430070, China
| | - Yan Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, China
| |
Collapse
|
18
|
Di Stefano M, Santonocito S, Polizzi A, Mauceri R, Troiano G, Lo Giudice A, Romano A, Mascitti M, Isola G. A Reciprocal Link between Oral, Gut Microbiota during Periodontitis: The Potential Role of Probiotics in Reducing Dysbiosis-Induced Inflammation. Int J Mol Sci 2023; 24:1084. [PMID: 36674600 PMCID: PMC9867370 DOI: 10.3390/ijms24021084] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/28/2022] [Indexed: 01/09/2023] Open
Abstract
Human body is colonized by a florid microbial community of bacteria, archaea, fungi, protists, helminths, and viruses, known as microbiota, which co-evolves with the host and influences its health through all stages of its life. It is well known that oral microorganisms form highly structurally and functionally organized multi-species biofilms and establish a network of complex mutual inter-species interactions having a primary function in synergy, signaling, or antagonism. This ecological model allows the microorganisms to increase their resistance to antimicrobial agents and settle a balanced microbes-host symbiotic relationship that ensures oral and global health status in humans. The host-associated microbiome is an important factor in human health and disease. Therefore, to develop novel diagnostic, therapeutic, and preventive strategies, microbiome's functions and the reciprocal interactions every microbiome entertains with other microbial communities in the human body are being investigated. This review provides an analysis of the literature about the close connection between the two largest microbial communities in humans: the oral and the gut microbiomes. Furthermore, it focuses on how the alteration of their microbial and functional characteristics can lead to and reciprocally influence the onset of both oral and intestinal microbiome-associated illness, along with the potential role of probiotics in ameliorating inflammation and microbial dysbiosis.
Collapse
Affiliation(s)
- Mattia Di Stefano
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Simona Santonocito
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Alessandro Polizzi
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Rodolfo Mauceri
- Department of Surgical, Oncological and Oral Sciences (Di.Chir.On.S.), University of Palermo, 90127 Palermo, Italy
| | - Giuseppe Troiano
- Department of Clinical and Experimental Medicine, University of Foggia, Via Rovelli 50, 71122 Foggia, Italy
| | - Antonino Lo Giudice
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Alessandra Romano
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| | - Marco Mascitti
- Department of Clinical Specialistic and Dental Sciences, Marche Polytechnic University, Via Tronto 10/A, 60126 Ancona, Italy
| | - Gaetano Isola
- Department of General Surgery and Surgical-Medical Specialties, School of Dentistry, University of Catania, 95124 Catania, Italy
| |
Collapse
|
19
|
Wang W, Guan J, Feng Y, Nie L, Xu Y, Xu H, Fu F. Polystyrene microplastics induced nephrotoxicity associated with oxidative stress, inflammation, and endoplasmic reticulum stress in juvenile rats. Front Nutr 2023; 9:1059660. [PMID: 36687698 PMCID: PMC9853403 DOI: 10.3389/fnut.2022.1059660] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/19/2022] [Indexed: 01/08/2023] Open
Abstract
Introduction Unintended intake of microplastic particles has been demonstrated to exert adverse health effects, however, studies on relevant nephrotoxicity in juvenile mammals are lacking. Methods Therefore, we investigated the potential nephrotoxicity of oral-exposed polystyrene microplastics (PSMPs) (1,000 nm, 2.0 mg/kg/d) for 28 days in juvenile rats. Levels of oxidative stress, inflammation, and endoplasmic reticulum (ER) stress in kidneys were analyzed. Results and discussion Results revealed that PSMPs noticeably decreased the growth rate of bodyweight, and organ index of the kidney, cardiac, and ovary. The intestinal injury caused by PSMPs exposure was also observed, which was distinctly alleviated with N-acetyl-cysteine (NAC) and Salubrinal (Sal) treatment compared with the single PSMPs group. PSMPs caused histological lesions of the kidney via disrupting the serum blood urea nitrogen (BUN), creatinine (CRE), and pro-inflammatory mediators IL-1β, IL-6, and TNF-α. Furthermore, PSMPs exposure induced ER stress and inflammation presumably potentially mediated by oxidative stress in kidneys of rats. Eventually, PSMPs also promoted renal cells apoptosis, manifested as an obvious increase in the number of positive cells for the dUTP nick end labeling of Terminal deoxynucleotidyl transferase, which also can be confirmed by the elevated expression of genes associated with apoptosis Bcl-2, Bax, Caspase-12, Caspase-9, Caspase-3, and IHC score of Caspase-12 in the PSMPs group. Supplementation of NAC and Sal not only ameliorated the PSMPs-induced oxidative stress and ER stress but also the inflammation and apoptosis in the kidney. Collectively, this study suggested that PSMPs caused nephrotoxicity in juvenile rats potentially through oxidative damage and ER stress, which call for greater efforts to be taken on regulating the PSMPs ingestion in children.
Collapse
Affiliation(s)
- Wanzhen Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, China,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Jiafu Guan
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yueying Feng
- The Second Affiliated Hospital of Nanchang University, Nanchang, China,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Liju Nie
- Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Yuanyuan Xu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China,*Correspondence: Hengyi Xu, ,
| | - Fen Fu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China,Fen Fu,
| |
Collapse
|
20
|
Ganoderma lucidum Ethanol Extraction Promotes Dextran Sulphate Sodium Induced Colitis Recovery and Modulation in Microbiota. Foods 2022; 11:foods11244023. [PMID: 36553765 PMCID: PMC9778072 DOI: 10.3390/foods11244023] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/03/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Popular edible mushrooms Ganoderma lucidum and Gloeostereum incarnatum can improve physical health as a prebiotic and positively alter intestinal microbiota. Our research investigated the prebiotic effects of Ganoderma lucidum and Gloeostereum incarnatum on colon inflammation through G. lucidum water extraction polysaccharides (GLP), G. incarnatum water extraction polysaccharides (GIP), G. lucidum ethanol extraction (GLE), and G. incarnatum ethanol extraction (GIE) administered in mice after 7 days of dextran sulphate sodium (DSS) administration. Among the extracts, GLE showed reduced mortality rates, prevention of weight loss, mitigated colon length shortening, and decreased disease activity indices and histological scores. COX-2, MPO, and iNOS activities and the inflammatory cytokines' expressions were determined to demonstrate the inhibition inflammation by GLE. Meanwhile, GLE upregulated the levels of MUC2, ZO-1, claudin-3, and occluding to protect the intestinal barrier. Furthermore, GLE modulated the composition of gut microbiota disturbed by DSS, as it decreased the abundance of Bacteroides, Staphylococcus, and Escherichia_Shigella, and increased Turicibacter and Bifidobacterium. Through cell experiment, GLE had a positive influence on adherens junction, tight junction, and TRAF6/MyD88/NF-κB signaling pathways. In conclusion, GLE supplementation promotes DSS-induced colitis recovery by regulating inflammatory cytokines, preserving the intestinal mucosal barrier, positively modulating microbiota changes, and positively influences immune response in TRAF6/MyD88/NF-κB signaling pathways.
Collapse
|
21
|
Qin D, Ma Y, Wang Y, Hou X, Yu L. Contribution of Lactobacilli on Intestinal Mucosal Barrier and Diseases: Perspectives and Challenges of Lactobacillus casei. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111910. [PMID: 36431045 PMCID: PMC9696601 DOI: 10.3390/life12111910] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The intestine barrier, the front line of normal body defense, relies on its structural integrity, microbial composition and barrier immunity. The intestinal mucosal surface is continuously exposed to a complex and dynamic community of microorganisms. Although it occupies a relatively small proportion of the intestinal microbiota, Lactobacilli has been discovered to have a significant impact on the intestine tract in previous studies. It is undeniable that some Lactobacillus strains present probiotic properties through maintaining the micro-ecological balance via different mechanisms, such as mucosal barrier function and barrier immunity, to prevent infection and even to solve some neurology issues by microbiota-gut-brain/liver/lung axis communication. Notably, not only living cells but also Lactobacillus derivatives (postbiotics: soluble secreted products and para-probiotics: cell structural components) may exert antipathogenic effects and beneficial functions for the gut mucosal barrier. However, substantial research on specific effects, safety and action mechanisms in vivo should be done. In clinical application of humans and animals, there are still doubts about the precise evaluation of Lactobacilli's safety, therapeutic effect, dosage and other aspects. Therefore, we provide an overview of central issues on the impacts of Lactobacillus casei (L. casei) and their products on the intestinal mucosal barrier and some diseases and highlight the urgent need for further studies.
Collapse
Affiliation(s)
- Da Qin
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yixuan Ma
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yanhong Wang
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xilin Hou
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| | - Liyun Yu
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| |
Collapse
|
22
|
Adıgüzel E, Çiçek B, Ünal G, Aydın MF, Barlak-Keti D. Probiotics and prebiotics alleviate behavioral deficits, inflammatory response, and gut dysbiosis in prenatal VPA-induced rodent model of autism. Physiol Behav 2022; 256:113961. [PMID: 36100109 DOI: 10.1016/j.physbeh.2022.113961] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/07/2022] [Accepted: 09/09/2022] [Indexed: 11/16/2022]
Abstract
Autism spectrum disorders are neuropsychiatric conditions characterized by social interaction and communication disorders and repetitive stereotypical behaviors. These disorders are also accompanied by an inflammatory status. Bidirectional communication between microbiome, gut, and brain has been discovered as a major mechanism influencing core symptoms and biomarkers of autism. Therefore, the modulation of the gut microbiota in autism has recently attracted interest. In this study, probiotic- and prebiotic-mediated modulation of the gut microbiota was compared in terms of different symptoms and findings in an experimental autism model. Valproic acid (VPA) (500 mg/kg) was administered to Wistar rats (on prenatal day 12.5) to induce autistic-like behaviors. Based on the supply of probiotics and prebiotics, animals were grouped as control (saline), autistic-like (prenatal VPA), probiotic (prenatal VPA + 22.5 × 109 cfu/day probiotic), prebiotic (prenatal VPA + 100 mg/day prebiotic), and combined treatment (prenatal VPA + 22.5 × 109 cfu/day probiotic + 100 mg/day prebiotic). After the treatment process, behavioral tests (social behaviors, anxiety, stereotypical behavior, sensorimotor gating, and behavioral despair) and biochemical analyses (serum and brain tissue) were conducted, and the quantities of some phyla and genera were determined in stool samples. Significant positive effects of probiotic and combined treatments were observed on the sociability, social interaction, and anxiety parameters. In addition, all three treatments had positive effects on stereotypical behavior. However, the treatments did not affect sensorimotor gating deficits and behavioral despair. Further, probiotic treatment reversed the VPA-induced increase and decrease in serum IL-6 and IL-10 levels, respectively. Combined treatment also significantly increased the IL-10 levels. Prenatal VPA exposure decreased 5-hydroxytryptamine (5-HT) levels in the prefrontal cortex of the brain; however, combined treatment reversed this decrease. Prenatal VPA exposure also caused a decrease in Bacteroidetes/Firmicutes ratio in the gut microbiota, while the probiotic treatment significantly increased this ratio. These findings indicate that probiotic- and prebiotic-mediated microbial modulation may represent a new therapeutic approach to alleviate autistic-like symptoms.
Collapse
Affiliation(s)
- Emre Adıgüzel
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Karamanoğlu Mehmetbey University, İbrahim Öktem Street, Karaman 70200, Turkey.
| | - Betül Çiçek
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Erciyes University, Kayseri, Turkey.
| | - Gökhan Ünal
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey.
| | - Mehmet Fatih Aydın
- Department of Public Health, Faculty of Health Sciences, Karamanoğlu Mehmetbey University, Karaman, Turkey,.
| | - Didem Barlak-Keti
- Department of Medical Biochemistry, Medical School, Erciyes University, Kayseri, Turkey.
| |
Collapse
|
23
|
Li B, Mao Q, Xiong R, Zhou D, Huang S, Saimaiti A, Shang A, Luo M, Li H, Li H, Li S. Preventive Effects of Different Black and Dark Teas on Obesity and Non-Alcoholic Fatty Liver Disease and Modulate Gut Microbiota in High-Fat Diet Fed Mice. Foods 2022; 11:3457. [PMID: 36360069 PMCID: PMC9658379 DOI: 10.3390/foods11213457] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/27/2022] [Accepted: 10/27/2022] [Indexed: 08/13/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a leading public health challenge and is closely associated with metabolic syndromes, such as obesity. Intestinal microbiota dysbiosis could play a vital role in the pathogenesis and progression of NAFLD. Tea is the second most popular health drink in the world behind water, and exhibits many health-promoting effects. In this study, the protective effects of different black and dark teas on NAFLD induced by long-term high-fat diet (HFD) exposure and their regulation of gut microbiota were evaluated and explored. The results indicated that supplementation with different black and dark tea extracts could significantly suppress the energy intake, alleviate abnormal accumulation of visceral fat, and prevent obesity, hepatic abnormal lipid deposition and liver steatosis in HFD-fed mice at varying degrees. In addition, Dianhong tea and Liupao tea interventions could significantly decrease the ratio of Firmicutes to Bacteroidetes, and selenium-enriched black tea and selenium-enriched dark rea supplementation could remarkably reduce the relative abundance of Actinobacteria compared to the model group. Moreover, these teas could partly shift the relative abundances of Allobaculum, Roseburia and Dubosiella. Taken together, black teas and dark teas could prevent HFD-induced features of obesity and NAFLD, which might partly be due to the modulation of gut microbiota.
Collapse
Affiliation(s)
- Bangyan Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qianqian Mao
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ruogu Xiong
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Dandan Zhou
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Siyu Huang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Adila Saimaiti
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Ao Shang
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Min Luo
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Hangyu Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Huabin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Sha Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
24
|
Huang Y, Lin X, Yu S, Chen R, Chen W. Intestinal Engineered Probiotics as Living Therapeutics: Chassis Selection, Colonization Enhancement, Gene Circuit Design, and Biocontainment. ACS Synth Biol 2022; 11:3134-3153. [PMID: 36094344 DOI: 10.1021/acssynbio.2c00314] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Intestinal probiotics are often used for the in situ treatment of diseases, such as metabolic disorders, tumors, and chronic inflammatory infections. Recently, there has been an increased emphasis on intelligent, customized treatments with a focus on long-term efficacy; however, traditional probiotic therapy has not kept up with this trend. The use of synthetic biology to construct gut-engineered probiotics as live therapeutics is a promising avenue in the treatment of specific diseases, such as phenylketonuria and inflammatory bowel disease. These studies generally involve a series of fundamental design issues: choosing an engineered chassis, improving the colonization ability of engineered probiotics, designing functional gene circuits, and ensuring the safety of engineered probiotics. In this review, we summarize the relevant past research, the progress of current research, and discuss the key issues that restrict the widespread application of intestinal engineered probiotic living therapeutics.
Collapse
Affiliation(s)
- Yan Huang
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Xiaojun Lin
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Siyang Yu
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China
| | - Ruiyue Chen
- Team SZU-China at iGEM 2021, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
| | - Weizhao Chen
- Team SZU-China at iGEM 2021, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518060, China.,Shenzhen Key Laboratory for Microbial Gene Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
25
|
Bucheli JEV, Todorov SD, Holzapfel WH. Role of gastrointestinal microbial populations, a terra incognita of the human body in the management of intestinal bowel disease and metabolic disorders. Benef Microbes 2022; 13:295-318. [PMID: 35866598 DOI: 10.3920/bm2022.0022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intestinal bowel disease (IBD) is a chronic immune-mediated clinical condition that affects the gastrointestinal tract and is mediated by an inflammatory response. Although it has been extensively studied, the multifactorial aetiology of this disorder makes it difficult to fully understand all the involved mechanisms in its development and therefore its treatment. In recent years, the fundamental role played by the human microbiota in the pathogenesis of IBD has been emphasised. Microbial imbalances in the gut bacterial communities and a lower species diversity in patients suffering from inflammatory gastrointestinal disorders compared to healthy individuals have been reported as principal factors in the development of IBD. These served to support scientific arguments for the use of probiotic microorganisms in alternative approaches for the prevention and treatment of IBD. In a homeostatic environment, the presence of bacteria (including probiotics) on the intestinal epithelial surface activates a cascade of processes by which immune responses inhibited and thereby commensal organisms maintained. At the same time these processes may support activities against specific pathogenic bacteria. In dysbiosis, these underlying mechanisms will serve to provoke a proinflammatory response, that, in combination with the use of antibiotics and the genetic predisposition of the host, will culminate in the development of IBD. In this review, we summarised the main causes of IBD, the physiological mechanisms involved and the related bacterial groups most frequently associated with these processes. The intention was to enable a better understanding of the interaction between the intestinal microbiota and the host, and to suggest possibilities by which this knowledge can be useful for the development of new therapeutic treatments.
Collapse
Affiliation(s)
- J E Vazquez Bucheli
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk 37554, Republic of Korea
| | - S D Todorov
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk 37554, Republic of Korea
| | - W H Holzapfel
- Human Effective Microbes, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk 37554, Republic of Korea
| |
Collapse
|
26
|
Probiotics Bring New Hope for Atherosclerosis Prevention and Treatment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3900835. [PMID: 36193065 PMCID: PMC9526629 DOI: 10.1155/2022/3900835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/12/2022] [Accepted: 09/17/2022] [Indexed: 11/26/2022]
Abstract
Cardiovascular disease is the leading cause of human mortality and morbidity worldwide. Atherosclerosis (AS) is the underlying pathological responsible in most acute and severe cardiovascular diseases including myocardial infarction and stroke. However, current drugs applied to the treatment of AS are not clinically effective, and there is a large residual risk of cardiovascular disease and multiple side effects. Increasing evidence supports a close relationship between microorganisms and the incidence of AS. Recent data have shown that probiotics can improve multiple key factors involved in the development and progression of AS, including cholesterol metabolism imbalance, endothelial dysfunction, proinflammatory factor production, macrophage polarization, intestinal flora disturbance, and infection with pathogenic microorganisms, and therefore probiotics have attracted great interest as a novel potential “medicine”. This review is aimed at summarizing the effects of probiotics on various influencing factors, and providing valuable insights in the search for early prevention and potential therapeutic strategies for AS.
Collapse
|
27
|
Kim H, Nam BY, Park J, Song S, Kim WK, Lee K, Nam TW, Park JT, Yoo TH, Kang SW, Ko G, Han SH. Lactobacillus acidophilus KBL409 reduces kidney fibrosis via immune modulatory effects in mice with chronic kidney disease. Mol Nutr Food Res 2022; 66:e2101105. [PMID: 36059191 DOI: 10.1002/mnfr.202101105] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 08/10/2022] [Indexed: 11/10/2022]
Abstract
SCOPE Intestinal dysbiosis has been reported to play an important role in the pathogenesis of various diseases, including chronic kidney disease (CKD). Here, we aimed to evaluate whether probiotic supplements can have protective effects against kidney injury in an animal model of CKD. METHODS AND RESULTS An animal model of CKD was established by feeding C57BL/6 mice a diet containing 0.2% adenine. These model mice were administered Lactobacillus acidophilus KBL409 daily for 4 weeks. Features of adenine-induce CKD (Ade-CKD) mice, such as prominent kidney fibrosis and higher levels of serum creatinine and albuminuria were improved by administration of KBL409. Ade-CKD mice also exhibited a disrupted intestinal barrier and elevated levels of TNF-α, IL-6, and 8-hydroxy-2'-deoxyguanosine. These changes were attenuated by KBL409. Administration of KBL409 significantly reduced macrophage infiltration and promoted a switch to the M2 macrophage phenotype and increasing regulatory T cells. Notably, the NLRP3 inflammasome pathway was activated in the kidneys of Ade-CKD and decreased by KBL409. In primary kidney tubular epithelial cells treated with p-cresyl sulfate, short-chain fatty acids significantly increased M2 macrophage polarization factors and decreased profibrotic markers. CONCLUSIONS These results demonstrate that supplementation with the probiotic KBL409 has beneficial immunomodulating effects and protects against kidney injury. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Hyoungnae Kim
- Division of Nephrology, Soonchunhyang University Seoul Hospital, Seoul, Korea.,Graduate School, Yonsei University College of Medicine, Seoul, Korea
| | - Bo Young Nam
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | - Jimin Park
- Department of Internal Medicine, College of Medicine, Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Institute of Kidney Disease Research, Yonsei University, Seoul, Korea
| | | | - Woon-Ki Kim
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | | | | | - Jung Tak Park
- Yonsei University, Institute of Kidney Disease Research, College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - Tae-Hyun Yoo
- Yonsei University, Institute of Kidney Disease Research, College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - Shin-Wook Kang
- Yonsei University, Institute of Kidney Disease Research, College of Medicine, Department of Internal Medicine, Seoul, Korea
| | - GwangPyo Ko
- KoBiolabs, Inc., Seoul, Korea.,Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Korea
| | - Seung Hyeok Han
- Yonsei University, Institute of Kidney Disease Research, College of Medicine, Department of Internal Medicine, Seoul, Korea
| |
Collapse
|
28
|
Guo H, Hu R, Huang G, Pu W, Chu X, Xing C, Zhang C. Molybdenum and cadmium co-exposure induces endoplasmic reticulum stress-mediated apoptosis by Th1 polarization in Shaoxing duck (Anas platyrhyncha) spleens. CHEMOSPHERE 2022; 298:134275. [PMID: 35278442 DOI: 10.1016/j.chemosphere.2022.134275] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 06/14/2023]
Abstract
Excessive molybdenum (Mo) and cadmium (Cd) are deleterious to animals, but immunotoxicity co-induced by Mo and Cd remains unclear. To ascertain the confederate impacts of Mo and Cd on endoplasmic reticulum (ER) stress-mediated apoptosis by Helper T (Th) cells 1 polarization in the spleen of ducks, we randomly allocated forty 8-day-old Shaoxing ducks (Anas platyrhyncha) into 4 groups and reared them with having different doses of Mo and/or Cd basic diet. At the 16th week of the experiment, serum and spleen tissues were extracted. Data confirmed that Mo and/or Cd strikingly promoted their levels in spleen, caused histological abnormality and trace elements imbalance, and disrupted Th1/Th2 balance to divert toward Th1, then triggered ER stress by increasing three branches PERK/eIF2α/CHOP, IRE1/Caspase-12 and TRAF2/JNK signaling pathways-related genes mRNA and proteins levels, which stimulated apoptosis by elevating Bak-1, Bax, Caspase-9, Caspase-3 mRNA expression, and cleaved-Caspase-9/Caspase-9, cleaved-Caspase-3/Caspase-3 proteins expression as well as apoptosis rate, and decreasing Bcl-xL, Bcl-2 mRNA expression and Bcl-2/Bax ratio. Besides, the variation in combined group was most evident. Briefly, the study indicates that Mo and/or Cd exposure trigger ER stress-induced apoptosis via Th1 polarization in duck spleens, and its mechanism is somehow closely linked with the deposition of Cd and Mo, which may aggravate toxic damage to spleen.
Collapse
Affiliation(s)
- Huiling Guo
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Ruiming Hu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Gang Huang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Wenjing Pu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Xuesheng Chu
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Chenghong Xing
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China
| | - Caiying Zhang
- Jiangxi Provincial Key Laboratory for Animal Health, Institute of Animal Population Health, College of Animal Science and Technology, Jiangxi Agricultural University, Nanchang, Jiangxi, China.
| |
Collapse
|
29
|
KIANI AYSHAKARIM, BONETTI GABRIELE, DONATO KEVIN, BERTELLI MATTEO. Dietary supplements for intestinal inflammation. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2022; 63:E214-E220. [PMID: 36479492 PMCID: PMC9710413 DOI: 10.15167/2421-4248/jpmh2022.63.2s3.2763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Intestinal inflammation leads to various chronic diseases, collectively known as inflammatory bowel disease (IBD). IBD mainly affects the large intestine, but it can also affect the gastrointestinal tract as a whole. Its major symptoms are pain, diarrhea, and weight loss, and it is usually associated with deficiencies of both macro- and micronutrients. Unluckily, after some time the body develops resistance against the already available drugs: thus, many patients fail to maintain remission, which is achieved in less than 50% of cases. Diet is a major determinant of gut inflammation. An unbalanced diet can affect the gut microbiota and cause dysbiosis, which is related to a dysregulated host immune response. The Mediterranean Diet its renowned for its anti-inflammatory effects and for preventing dysbiosis. In order to improve management and treatment of intestinal inflammatory diseases, it should become common practice to integrate the patient's diet with dietary supplements with anti-inflammatory effects (probiotics, butyrate, phosphatidylcholine, lactoferrin, palmitoylethanolamide, silymarin, and omega 3), which maintain the stability of the intestinal microbial cohort and strengthen the mucosal barrier, thus preventing or soothing IBD symptoms. Dietary supplements may help fight the high costs, the adverse side effects, and the recurrent relapses typical of drug use.
Collapse
Affiliation(s)
| | | | | | - MATTEO BERTELLI
- MAGI Euregio, Bolzano, Italy
- MAGI’S LAB, Rovereto (TN), Italy
- MAGISNAT, Peachtree Corners (GA), USA
| |
Collapse
|
30
|
Yang Z, Wei F, Zhang B, Luo Y, Xing X, Wang M, Chen R, Sun G, Sun X. Cellular Immune Signal Exchange From Ischemic Stroke to Intestinal Lesions Through Brain-Gut Axis. Front Immunol 2022; 13:688619. [PMID: 35432368 PMCID: PMC9010780 DOI: 10.3389/fimmu.2022.688619] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
As a vital pivot for the human circulatory system, the brain-gut axis is now being considered as an important channel for many of the small immune molecules’ transductions, including interleukins, interferons, neurotransmitters, peptides, and the chemokines penetrating the mesentery and blood brain barrier (BBB) during the development of an ischemic stroke (IS). Hypoxia-ischemia contributes to pituitary and neurofunctional disorders by interfering with the molecular signal release and communication then providing feedback to the gut. Suffering from such a disease on a long-term basis may cause the peripheral system’s homeostasis to become imbalanced, and it can also lead to multiple intestinal complications such as gut microbiota dysbiosis (GMD), inflammatory bowel disease (IBD), necrotizing enterocolitis (NEC), and even the tumorigenesis of colorectal carcinoma (CRC). Correspondingly, these complications will deteriorate the cerebral infarctions and, in patients suffering with IS, it can even ruin the brain’s immune system. This review summarized recent studies on abnormal immunological signal exchange mediated polarization subtype changes, in both macrophages and microglial cells as well as T-lymphocytes. How gut complications modulate the immune signal transduction from the brain are also elucidated and analyzed. The conclusions drawn in this review could provide guidance and novel strategies to benefit remedies for both IS and relative gut lesions from immune-prophylaxis and immunotherapy aspects.
Collapse
Affiliation(s)
- Zizhao Yang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Fei Wei
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Zhang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun Luo
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Xing
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Wang
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rongchang Chen
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guibo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Xiaobo Sun,
| | - Xiaobo Sun
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Xiaobo Sun,
| |
Collapse
|
31
|
Gao H, He C, Hua R, Guo Y, Wang B, Liang C, Gao L, Shang H, Xu JD. Endoplasmic Reticulum Stress of Gut Enterocyte and Intestinal Diseases. Front Mol Biosci 2022; 9:817392. [PMID: 35402506 PMCID: PMC8988245 DOI: 10.3389/fmolb.2022.817392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum, a vast reticular membranous network from the nuclear envelope to the plasma membrane responsible for the synthesis, maturation, and trafficking of a wide range of proteins, is considerably sensitive to changes in its luminal homeostasis. The loss of ER luminal homeostasis leads to abnormalities referred to as endoplasmic reticulum (ER) stress. Thus, the cell activates an adaptive response known as the unfolded protein response (UPR), a mechanism to stabilize ER homeostasis under severe environmental conditions. ER stress has recently been postulated as a disease research breakthrough due to its significant role in multiple vital cellular functions. This has caused numerous reports that ER stress-induced cell dysfunction has been implicated as an essential contributor to the occurrence and development of many diseases, resulting in them targeting the relief of ER stress. This review aims to outline the multiple molecular mechanisms of ER stress that can elucidate ER as an expansive, membrane-enclosed organelle playing a crucial role in numerous cellular functions with evident changes of several cells encountering ER stress. Alongside, we mainly focused on the therapeutic potential of ER stress inhibition in gastrointestinal diseases such as inflammatory bowel disease (IBD) and colorectal cancer. To conclude, we reviewed advanced research and highlighted future treatment strategies of ER stress-associated conditions.
Collapse
Affiliation(s)
- Han Gao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chengwei He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Rongxuan Hua
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuexin Guo
- Department of Oral Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Boya Wang
- Undergraduate Student of 2018 Eight Program of Clinical Medicine, Peking University Health Science Center, Beijing, China
| | - Chen Liang
- Department of Clinical Medicine, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Lei Gao
- Department of Biomedical Informatics, School of Biomedical Engineering, Capital Medical University, Beijing, China
| | - Hongwei Shang
- Experimental Center for Morphological Research Platform, Capital Medical University, Beijing, China
| | - Jing-Dong Xu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- *Correspondence: Jing-Dong Xu,
| |
Collapse
|
32
|
Little M, Dutta M, Li H, Matson A, Shi X, Mascarinas G, Molla B, Weigel K, Gu H, Mani S, Cui JY. Understanding the physiological functions of the host xenobiotic-sensing nuclear receptors PXR and CAR on the gut microbiome using genetically modified mice. Acta Pharm Sin B 2022; 12:801-820. [PMID: 35256948 PMCID: PMC8897037 DOI: 10.1016/j.apsb.2021.07.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/29/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Pharmacological activation of the xenobiotic-sensing nuclear receptors pregnane X receptor (PXR) and constitutive androstane receptor (CAR) is well-known to increase drug metabolism and reduce inflammation. Little is known regarding their physiological functions on the gut microbiome. In this study, we discovered bivalent hormetic functions of PXR/CAR modulating the richness of the gut microbiome using genetically engineered mice. The absence of PXR or CAR increased microbial richness, and absence of both receptors synergistically increased microbial richness. PXR and CAR deficiency increased the pro-inflammatory bacteria Helicobacteraceae and Helicobacter. Deficiency in both PXR and CAR increased the relative abundance of Lactobacillus, which has bile salt hydrolase activity, corresponding to decreased primary taurine-conjugated bile acids (BAs) in feces, which may lead to higher internal burden of taurine and unconjugated BAs, both of which are linked to inflammation, oxidative stress, and cytotoxicity. The basal effect of PXR/CAR on the gut microbiome was distinct from pharmacological and toxicological activation of these receptors. Common PXR/CAR-targeted bacteria were identified, the majority of which were suppressed by these receptors. hPXR-TG mice had a distinct microbial profile as compared to wild-type mice. This study is the first to unveil the basal functions of PXR and CAR on the gut microbiome.
Collapse
Key Words
- BA, bile acid
- BSH, bile salt hydrolase
- Bile acids
- CA, cholic acid
- CAR
- CAR, constitutive androstane receptor
- CDCA, chenodeoxycholic acid
- CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
- CV, conventional
- CYP, cytochrome P450
- DCA, deoxycholic acid
- EGF, epidermal growth factor
- Feces
- GF, germ free
- GLP-1, glucagon-like peptide-1
- GM-CSF, granulocyte-macrophage colony-stimulating factor
- Gut microbiome
- HDCA, hyodeoxycholic acid
- IBD, inflammatory bowel disease
- IFNγ, interferon-gamma
- IL, interleukin
- IS, internal standards
- Inflammation
- LCA, lithocholic acid
- LC–MS/MS, liquid chromatography–tandem mass spectrometry
- MCA, muricholic acid
- MCP-1, monocyte chemoattractant protein-1
- Mice
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NSAID, non-steroidal anti-inflammatory drug
- Nuclear receptor
- OH, hydroxylated
- OTUs, operational taxonomy units
- PA, indole-3 propionic acid
- PBDEs, polybrominated diphenyl ethers
- PCBs, polychlorinated biphenyls
- PCoA, Principle Coordinate Analysis
- PXR
- PXR, pregnane X receptor
- PiCRUSt, Phylogenetic Investigation of Communities by Reconstruction of Observed States
- QIIME, Quantitative Insights Into Microbial Ecology
- SCFAs, short-chain fatty acids
- SNP, single-nucleotide polymorphism
- SPF, specific-pathogen-free
- T, wild type
- T-, taurine conjugated
- TCPOBOP, 1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene, 3,3′,5,5′-Tetrachloro-1,4-bis(pyridyloxy)benzene
- TGR-5, Takeda G-protein-coupled receptor 5
- TLR4, toll-like receptor 4
- TNF, tumor necrosis factor
- UDCA, ursodeoxycholic acid
- YAP, yes-associated protein
- hPXR-TG, humanized PXR transgenic
Collapse
Affiliation(s)
- Mallory Little
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Hao Li
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Adam Matson
- University of Connecticut, Hartford, CT 06106, USA
| | - Xiaojian Shi
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Gabby Mascarinas
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Bruk Molla
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Kris Weigel
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA
| | - Sridhar Mani
- Department of Medicine, Molecular Pharmacology and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
33
|
Chen L, Li R, Wang Z, Zhang Z, Wang J, Qiao Y, Huang Y, Liu W. Lactate-utilizing bacteria ameliorates DSS-induced colitis in mice. Life Sci 2022; 288:120179. [PMID: 34838850 DOI: 10.1016/j.lfs.2021.120179] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/16/2021] [Accepted: 11/21/2021] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel diseases (IBD) stem from alterations in the intestinal immune system and microbial dysbiosis, but the precise interactions between bacteria and IBD remain obscure. The commensal microbiota have a profound impact on human health and diseases. Here, we developed a selective culture medium for lactate-utilizing bacteria (LUB) that function as candidate probiotics to ameliorate IBD using a mouse model. Firstly, LUB, including Megasphaera, were enriched from human faeces using a selective medium with lactate. LUB efficiently attenuated the pathology of colitis induced by dextran sulphate sodium (DSS). Next, LUB administration counteracted the dysbiosis associated with the intestinal inflammatory process, and elevated the proportion of Escherichia-Shigella in intestines. Moreover, E. coli isolated from healthy faeces downstream recapitulated lactate-utilizing bacterial community to ameliorate the severity of DSS-induced acute colitis. In conclusion, our finding revealed that LUB were sufficient to exert inflammatory protection against colitis in mice, highlighting a novel therapeutic strategy to use LUB as potentially curable probiotics for therapeutic manipulation for IBD.
Collapse
Affiliation(s)
- Lirong Chen
- Department of Medical Laboratory Science, Shanxi Medical University Fenyang College, Shanxi 032200, China.
| | - Rong Li
- College of Basic Medicine, Chengde Medical University, Hebei 067000, China
| | - Ziguang Wang
- First Clinical Medical College, Mudanjiang Medical College; Department of Laboratory Medicine, The Second Affiliated Hospital of Mudanjiang Medical College, Heilongjiang 157000, China
| | - Zhiwei Zhang
- Department of Gastroenterology, Shanxi Fenyang Hospital, Shanxi 032200, China
| | - Jie Wang
- Jingle County People's Hospital, Shanxi 035100, China
| | - Yuebing Qiao
- College of Basic Medicine, Chengde Medical University, Hebei 067000, China
| | - Yongcun Huang
- First Clinical Medical College, Mudanjiang Medical College; Department of Laboratory Medicine, The Second Affiliated Hospital of Mudanjiang Medical College, Heilongjiang 157000, China
| | - Wei Liu
- School of Plant Protection, Anhui Agricultural University; Anhui Province Key Laboratory of Crop Integrated Pest Management; Anhui Province Engineering Laboratory for Green Pesticide Development and Application, Anhui 230036, China; Department of Medical Laboratory Science, Shanxi Medical University Fenyang College, Shanxi 032200, China.
| |
Collapse
|
34
|
Li M, Yu L, Zhai Q, Liu B, Zhao J, Zhang H, Chen W, Tian F. Ganoderma applanatum polysaccharides and ethanol extracts promote the recovery of colitis through intestinal barrier protection and gut microbiota modulations. Food Funct 2021; 13:688-701. [PMID: 34935013 DOI: 10.1039/d1fo03677g] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel disease is associated with intestinal homeostasis dysregulation and gut microbiota dysbiosis. This study aimed to investigate the protective effect of Ganoderma applanatum extracts (G. applanatum polysaccharides (GAP) and 75% ethanol extracts (GAE)) on colon inflammation and elucidate the therapeutic mechanism. GAP and GAE showed considerable protective effects against dextran sodium sulfate (DSS)-induced colitis, as demonstrated by reduced mortality, body weight, disease activity index score, colon length, and histological score. Through GAP and GAE administration, the destroyed intestinal barrier recovered to normal, as did intestinal inflammation. We also confirmed that GAP administration promoted the recovery of colitis in a gut microbiota-dependent manner. The similarity between GAP and GAE administration was that they both altered the disordered gut microbiota damaged by DSS, exhibiting reduced abundance of Escherichia_Shigella, Enterococcus, and Staphylococcus, but the modulation of the gut microbiota was distinct between GAP and GAE.
Collapse
Affiliation(s)
- Miaoyu Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Bingshu Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China.,Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China.,School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
35
|
Ferreira AF, Braga RLL, Andrade MF, Rosa ACDP, Pereira-Manfro WF. SYNERGISTIC IMMUNOMODULATORY ACTIVITY OF PROBIOTICS BIFIDOBACTERIUM ANIMALIS AND LACTOBACILLUS CASEI IN ENTEROAGGREGATIVE ESCHERICHIA COLI (EAEC)-INFECTED CACO-2 CELLS. ARQUIVOS DE GASTROENTEROLOGIA 2021; 58:433-438. [PMID: 34909846 DOI: 10.1590/s0004-2803.202100000-79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/03/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Enteroaggregative Escherichia coli (EAEC) is an E. coli pathotype that presents aggregative adhesion patterns on in vitro cultivated cells, mainly related to persistent diarrhea cases in children. EAEC virulence factors are important for host colonization and pathogeni-city. Intestinal epithelial cells (IECs) recognize pathogen-associated molecular patterns (PAMPs) and initiate an immune response. Several studies using in vivo and in vitro models emphasize the probiotic activity and immunomodulatory capacity of Lactobacillus and Bifidobacterium species. OBJECTIVE To evaluate the modulation of cytokine production by probiotics Bifidobacterium animalis and Lactobacillus casei in human intestinal Caco-2 cells exposed to different strains of EAEC. METHODS Caco-2 cells were incubated with EAEC strains in the presence or absence of probiotics. The production of cytokines IL-8, TNF-α, IL-1β and IL-10 was evaluated in the supernatants by a sandwich enzyme-linked immunosorbent assay (ELISA). RESULTS Cytokine production did not change when uninfected and EAEC-infected Caco-2 cells were exposed to probiotics separately. All EAEC induced a significant increase in IL-8 production by Caco-2 cells, but the probiotics, even together, could not reduce its production. On the other hand, the synergic activity of probiotic strains significantly increased TNF-α production but decreased the basal production of IL-1ß. Also, probiotics induced a significant increase in the production of the anti-inflammatory cytokine IL-10 during EAEC infection. CONCLUSION Our results reinforce the synergistic immunomodulatory activity of probiotics during EAEC infection.
Collapse
Affiliation(s)
- Andréa Fonseca Ferreira
- Universidade do Estado do Rio de Janeiro, Faculdade de Ciências Médicas, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, RJ, Brasil
| | - Ricardo Luís Lopes Braga
- Universidade do Estado do Rio de Janeiro, Faculdade de Ciências Médicas, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, RJ, Brasil
| | - Maysa Ferreira Andrade
- Universidade do Estado do Rio de Janeiro, Faculdade de Ciências Médicas, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, RJ, Brasil
| | - Ana Claudia de Paula Rosa
- Universidade do Estado do Rio de Janeiro, Faculdade de Ciências Médicas, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, RJ, Brasil
| | - Wânia Ferraz Pereira-Manfro
- Universidade do Estado do Rio de Janeiro, Faculdade de Ciências Médicas, Departamento de Microbiologia, Imunologia e Parasitologia, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
36
|
Bakshi HA, Quinn GA, Aljabali AAA, Hakkim FL, Farzand R, Nasef MM, Abuglela N, Ansari P, Mishra V, Serrano-Aroca Á, Tambuwala MM. Exploiting the Metabolism of the Gut Microbiome as a Vehicle for Targeted Drug Delivery to the Colon. Pharmaceuticals (Basel) 2021; 14:ph14121211. [PMID: 34959610 PMCID: PMC8709317 DOI: 10.3390/ph14121211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/10/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022] Open
Abstract
The prevalence of colon-associated diseases has increased significantly over the past several decades, as evidenced by accumulated literature on conditions such as Crohn’s disease, irritable bowel syndrome, colorectal cancer, and ulcerative colitis. Developing therapeutics for these diseases is challenging due to physiological barriers of the colon, systemic side effects, and the intestinal environment. Therefore, in a search for novel methods to overcome some of these problems, researchers discovered that microbial metabolism by gut microbiotia offers a potential method for targeted drug delivery This overview highlights several drug delivery systems used to modulate the microbiota and improve colon-targeted drug delivery. This technology will be important in developing a new generation of therapies which harness the metabolism of the human gut microflora.
Collapse
Affiliation(s)
- Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Sciences, Institute of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (G.A.Q.); (P.A.)
- Correspondence: (H.A.B.); (M.M.T.)
| | - Gerry A. Quinn
- School of Pharmacy and Pharmaceutical Sciences, Institute of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (G.A.Q.); (P.A.)
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 566, Jordan;
| | - Faruck L. Hakkim
- The Hormel Institute, University of Minnesota, Austin, MN 559122, USA;
| | - Rabia Farzand
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, UK; (R.F.); (M.M.N.); (N.A.)
| | - Mohamed M. Nasef
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, UK; (R.F.); (M.M.N.); (N.A.)
| | - Naji Abuglela
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, UK; (R.F.); (M.M.N.); (N.A.)
| | - Prawej Ansari
- School of Pharmacy and Pharmaceutical Sciences, Institute of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (G.A.Q.); (P.A.)
- Department of Pharmacy, Independent University, Dhaka 1229, Bangladesh
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India;
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab., Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, San Vicente Mártir, 46001 Valencia, Spain;
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Institute of Biomedical Sciences, Ulster University, Coleraine BT52 1SA, UK; (G.A.Q.); (P.A.)
- Correspondence: (H.A.B.); (M.M.T.)
| |
Collapse
|
37
|
Zhang Y, Mu T, Yang Y, Zhang J, Ren F, Wu Z. Lactobacillus johnsonii Attenuates Citrobacter rodentium-Induced Colitis by Regulating Inflammatory Responses and Endoplasmic Reticulum Stress in Mice. J Nutr 2021; 151:3391-3399. [PMID: 34383918 DOI: 10.1093/jn/nxab250] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 05/14/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Probiotics are beneficial in intestinal disorders. However, the benefits of Lactobacillus johnsonii in experimental colitis remain unknown. OBJECTIVES This study aimed to investigate the benefits of L. johnsonii against Citrobacter rodentium-induced colitis. METHODS Thirty-six 5-wk-old female C57BL/6J mice were randomly assigned to 3 groups (n = 12): control (Ctrl) group, Citrobacter rodentium treatment (CR) group (2 × 109 CFU C. rodentium), and Lactobacillus johnsonii and Citrobacter rodentium cotreatment (LJ + CR) group (109 CFU L. johnsonii with C. rodentium). Colon length, mucosal thickness, proinflammatory cytokine genes, and endoplasmic reticulum stress were tested. RESULTS The CR group had greater spleen weight, mucosal thickness, and Ki67+ cells (0.4-4.7 times), and a 23.8% shorter colon length than the Ctrl group, which in the LJ + CR group were 22.4%-77.6% lower and 30% greater than in the CR group, respectively. Relative to the Ctrl group, serum proinflammatory cytokines and immune cell infiltration were greater by 0.3-1.6 times and 6.2-8.8 times in the CR group, respectively; relative to the CR group, these were 19.9%-61.9% and 69.5%-84.2% lower in the LJ + CR group, respectively. The mRNA levels of lysozyme (Lyz) and regenerating islet-derived protein III were 22.7%-36.5% lower and 1.5-2.7 times greater in the CR group than in the Ctrl group, respectively, whereas they were 22.2%-25.7% greater and 57.2%-76.9% lower in the LJ + CR group than in the CR group, respectively. Cell apoptosis was 11.9 times greater in the CR group than in the Ctrl group, and 87.4% lower in the LJ + CR group than in the CR group. Consistently, the protein abundances of C/EBP homologous protein (CHOP), cleaved caspase 1 and 3, activating transcription factor 6α (ATF6A), and phospho-inositol-requiring enzyme 1α (P-IRE1A) were 0.3-2.1 times greater in the CR group and 31.1%-60.4% lower in the LJ + CR group. All these indexes did not differ between the Ctrl and LJ + CR groups, except for CD8+ T lymphocytes and CD11b+ and F4/80+ macrophages (1-1.5 times greater in LJ + CR) and mRNA concentration of Lyz2 (20.1% lower in LJ + CR). CONCLUSIONS L. johnsonii supplementation is a promising nutritional strategy for preventing C. rodentium-induced colitis in mice.
Collapse
Affiliation(s)
- Yunchang Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Tianqi Mu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jinhua Zhang
- College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition and Feed Science, College of Animal Science and Technology, China Agricultural University, Beijing, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| |
Collapse
|
38
|
Kim S, Choi S, Dutta M, Asubonteng JO, Polunas M, Goedken M, Gonzalez FJ, Cui JY, Gyamfi MA. Pregnane X receptor exacerbates nonalcoholic fatty liver disease accompanied by obesity- and inflammation-prone gut microbiome signature. Biochem Pharmacol 2021; 193:114698. [PMID: 34303710 PMCID: PMC9135326 DOI: 10.1016/j.bcp.2021.114698] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver disease due to the current epidemics of obesity and diabetes. The pregnane X receptor (PXR) is a xenobiotic-sensing nuclear receptor known for trans-activating liver genes involved in drug metabolism and transport, and more recently implicated in energy metabolism. The gut microbiota can modulate the host xenobiotic biotransformation and contribute to the development of obesity. While the male sex confers a higher risk for NAFLD than women before menopause, the mechanism remains unknown. We hypothesized that the presence of PXR promotes obesity by modifying the gut-liver axis in a sex-specific manner. Male and female C57BL/6 (wild-type/WT) and PXR-knockout (PXR-KO) mice were fed control or high-fat diet (HFD) for 16-weeks. Serum parameters, liver histopathology, transcriptomic profiling, 16S-rDNA sequencing, and bile acid (BA) metabolomics were performed. PXR enhanced HFD-induced weight gain, hepatic steatosis and inflammation especially in males, accompanied by PXR-dependent up-regulation in hepatic genes involved in microbial response, inflammation, oxidative stress, and cancer; PXR-dependent increase in intestinal Firmicutes/Bacteroides ratio (hallmark of obesity) and the pro-inflammatory Lactobacillus, as well as a decrease in the anti-obese Allobaculum and the anti-inflammatory Bifidobacterum, with a PXR-dependent reduction of beneficial BAs in liver. The resistance to NAFLD in females may be explained by PXR-dependent decrease in pro-inflammatory bacteria (Ruminococcus gnavus and Peptococcaceae). In conclusion, PXR exacerbates hepatic steatosis and inflammation accompanied by obesity- and inflammation-prone gut microbiome signature, suggesting that gut microbiome may contribute to PXR-mediated exacerbation of NAFLD.
Collapse
Affiliation(s)
- Sarah Kim
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Sora Choi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA
| | - Moumita Dutta
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA
| | - Jeffrey O Asubonteng
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA
| | - Marianne Polunas
- Office of Research and Economic Development, Research Pathology Services, Rutgers University, Piscataway, NJ, USA
| | - Michael Goedken
- Office of Research and Economic Development, Research Pathology Services, Rutgers University, Piscataway, NJ, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA, USA.
| | - Maxwell A Gyamfi
- Julius L. Chambers Biomedical Biotechnology Research Institute, North Carolina Central University, Durham, NC, USA.
| |
Collapse
|
39
|
Hess JM, Stephensen CB, Kratz M, Bolling BW. Exploring the Links between Diet and Inflammation: Dairy Foods as Case Studies. Adv Nutr 2021; 12:1S-13S. [PMID: 34632478 PMCID: PMC8502778 DOI: 10.1093/advances/nmab108] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 08/04/2021] [Indexed: 11/17/2022] Open
Abstract
Systemic chronic inflammation may be a contributing factor to many noncommunicable diseases, including diabetes, cardiovascular disease, and obesity. With the rapid rise of these conditions, identifying the causes of and treatment for chronic inflammation is an important research priority, especially with regard to modifiable lifestyle factors such as diet. An emerging body of evidence indicates that consuming certain foods, including dairy foods like milk, cheese, and yogurt, may be linked to a decreased risk for inflammation. To discuss both broader research on diet and inflammation as well as research on links between individual foods and inflammation, the National Dairy Council sponsored a satellite session entitled "Exploring the Links between Diet and Inflammation: Dairy Foods as Case Studies" at the American Society for Nutrition's 2020 LIVE ONLINE Conference. This article, a review based on the topics discussed during that session, explores the links between diet and inflammation, focusing most closely on the relations between intake of dairy fat and dairy foods like milk, cheese, and yogurt, and biomarkers of inflammation from clinical trials. While there is currently insufficient evidence to prove an "anti-inflammatory" effect of dairy foods, the substantial body of clinical research discussed in this review indicates that dairy foods do not increase concentrations of biomarkers of chronic systemic inflammation.
Collapse
Affiliation(s)
| | - Charles B Stephensen
- USDA Western Human Nutrition Research Center and Nutrition Department, University of California, Davis, Davis CA, USA
| | - Mario Kratz
- Fred Hutchinson Cancer Research Center, Public Health Sciences Division, Seattle, WA, USA
| | - Bradley W Bolling
- University of Wisconsin-Madison, Department of Food Science, Madison, WI, USA
| |
Collapse
|
40
|
Wu Z, Pan D, Jiang M, Sang L, Chang B. Selenium-Enriched Lactobacillus acidophilus Ameliorates Dextran Sulfate Sodium-Induced Chronic Colitis in Mice by Regulating Inflammatory Cytokines and Intestinal Microbiota. Front Med (Lausanne) 2021; 8:716816. [PMID: 34532332 PMCID: PMC8439139 DOI: 10.3389/fmed.2021.716816] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/05/2021] [Indexed: 12/29/2022] Open
Abstract
Aim: To evaluate the effect of Selenium-enriched Lactobacillus acidophilus (Se-enriched L. acidophilus) on dextran sulfate sodium (DSS)-induced colitis in mice. Methods: Mice were randomly divided into four groups: a control group, a control + Se-enriched L. acidophilus group, a chronic colitis group, and a chronic colitis + Se-enriched L. acidophilus group (n = 10 each group). The mice were sacrificed on the 26th day. The disease activity index, survival rates, and histological injury score were determined. Cytokines produced by lamina propria lymphocytes (LPLs), the selenium (Se) concentrations in serum and colon tissue and the mouse intestinal microbiota were evaluated. Results: Se-enriched L. acidophilus can improve histological injury and the disease activity index in mice with chronic colitis and reduce IL-1β, IL-6, IL-12p70, TNF-α, IL-23, IFN-γ, IL-17A, and IL-21 (P < 0.05) and increase IL-10 (P < 0.05) expression levels. Moreover, Se-enriched L. acidophilus can increase the β diversity of intestinal microbiota in mice with chronic colitis, significantly reduce the relative abundance of Lactobacillus and Romboutsia (P < 0.05), and significantly increase the relative abundance of Parasutterella (P < 0.05). Conclusions: Se-enriched L. acidophilus can improve DSS-induced chronic colitis by regulating inflammatory cytokines and intestinal microbiota.
Collapse
Affiliation(s)
- Zeyu Wu
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dan Pan
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Min Jiang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lixuan Sang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
41
|
Fremder M, Kim SW, Khamaysi A, Shimshilashvili L, Eini-Rider H, Park IS, Hadad U, Cheon JH, Ohana E. A transepithelial pathway delivers succinate to macrophages, thus perpetuating their pro-inflammatory metabolic state. Cell Rep 2021; 36:109521. [PMID: 34380041 DOI: 10.1016/j.celrep.2021.109521] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 02/23/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
The gut metabolite composition determined by the microbiota has paramount impact on gastrointestinal physiology. However, the role that bacterial metabolites play in communicating with host cells during inflammatory diseases is poorly understood. Here, we aim to identify the microbiota-determined output of the pro-inflammatory metabolite, succinate, and to elucidate the pathways that control transepithelial succinate absorption and subsequent succinate delivery to macrophages. We show a significant increase of succinate uptake into pro-inflammatory macrophages, which is controlled by Na+-dependent succinate transporters in macrophages and epithelial cells. Furthermore, we find that fecal and serum succinate concentrations were markedly augmented in inflammatory bowel diseases (IBDs) and corresponded to changes in succinate-metabolizing gut bacteria. Together, our results describe a succinate production and transport pathway that controls the absorption of succinate generated by distinct gut bacteria and its delivery into macrophages. In IBD, this mechanism fails to protect against the succinate surge, which may result in chronic inflammation.
Collapse
Affiliation(s)
- Moran Fremder
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Seung Won Kim
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Ahlam Khamaysi
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Liana Shimshilashvili
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Hadar Eini-Rider
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - I Seul Park
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea
| | - Uzi Hadad
- The Ilse Katz Institute for Nanoscale Science and Technology Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Jae Hee Cheon
- Department of Internal Medicine and Institute of Gastroenterology, Yonsei University College of Medicine, Seoul, Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Korea; Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea.
| | - Ehud Ohana
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| |
Collapse
|
42
|
Yao M, Lu Y, Zhang T, Xie J, Han S, Zhang S, Fei Y, Ling Z, Wu J, Hu Y, Ji S, Chen H, Berglund B, Li L. Improved functionality of Ligilactobacillus salivarius Li01 in alleviating colonic inflammation by layer-by-layer microencapsulation. NPJ Biofilms Microbiomes 2021; 7:58. [PMID: 34244520 PMCID: PMC8270932 DOI: 10.1038/s41522-021-00228-1] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 06/02/2021] [Indexed: 02/04/2023] Open
Abstract
The low viability during gastrointestinal transit and poor mucoadhesion considerably limits the effectiveness of Ligilactobacillus salivarius Li01 (Li01) in regulating gut microbiota and alleviating inflammatory bowel disease (IBD). In this study, a delivery system was designed through layer-by-layer (LbL) encapsulating a single Li01cell with chitosan and alginate. The layers were strengthened by cross-linking to form a firm and mucoadhesive shell (~10 nm thickness) covering the bacterial cell. The LbL Li01 displayed improved viability under simulated gastrointestinal conditions and mucoadhesive function. Almost no cells could be detected among the free Li01 after 2 h incubation in digestive fluids, while for LbL Li01, the total reduction was around 3 log CFU/mL and the viable number of cells remained above 6 log CFU/mL. Besides, a 5-fold increase in the value of rupture length and a two-fold increase in the number of peaks were found in the (bacteria-mucin) adhesion curves of LbL Li01, compared to those of free Li01. Oral administration with LbL Li01 on colitis mice facilitated intestinal barrier recovery and restoration of the gut microbiota. The improved functionality of Li01 by LbL encapsulation could increase the potential for the probiotic to be used in clinical applications to treat IBD; this should be explored in future studies.
Collapse
Affiliation(s)
- Mingfei Yao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yanmeng Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ting Zhang
- Department of Bone marrow, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaojiao Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shengyi Han
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Shuobo Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yiqiu Fei
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zongxin Ling
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jingjing Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yue Hu
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China
| | - Shouling Ji
- College of Computer Science and Technology, Zhejiang University, Hangzhou, China
| | - Hao Chen
- Center for molecular Imaging Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Björn Berglund
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
43
|
Sun M, Liu Y, Song Y, Gao Y, Zhao F, Luo Y, Qian F, Mu G, Tuo Y. The ameliorative effect of Lactobacillus plantarum-12 on DSS-induced murine colitis. Food Funct 2021; 11:5205-5222. [PMID: 32458908 DOI: 10.1039/d0fo00007h] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Some strains of lactobacilli can exert beneficial effects on a host when ingested in an adequate dose, such as immunoregulation and anti-inflammatory activities. In this study, the survival abilities under simulated gastrointestinal conditions, adhesion abilities on HT-29 cell monolayers, and hemolytic activities of four Lactobacillus plantarum strains were assessed. Among the four strains, L. plantarum-12 showed the higher survival rate under simulated gastrointestinal conditions and adhesion index on the HT-29 cell monolayers, exhibited γ-haemolytic activity and had no biological amine producing ability. L. plantarum-12 was administered to dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) Balb/c mice by oral gavage for 10 days. It was observed that the UC Balb/c mice showed symptoms of colonic atrophy, intestinal histopathological change, gut microbial disturbance, and pro-inflammatory cytokine expression. L. plantarum-12 administration remarkably attenuated DSS-induced UC in mice. L. plantarum-12 administration could restore gut microbiota by increasing beneficial bacteria such as Lactobacillus and decreasing intestinal pathogenic bacteria like Proteobacteria. L. plantarum-12 administration could improve immunity via activating the janus kinase-signal transducer and the activator of the transcription (JAK-STAT) pathway and up-regulating adenosine deaminase (ADA) and interferon-induced protein with tetratricopeptide repeats 1 protein (IFIT1), and enforce the intestinal barrier function by up-regulating mucin 2 (MUC2) protein expression. In conclusion, L. plantarum-12 could attenuate DSS-induced UC in Balb/c mice by ameliorating intestinal inflammation, and restoring the disturbed gut microbiota. L. plantarum-12 could be used as promising probiotics to ameliorate colitis.
Collapse
Affiliation(s)
- Mengying Sun
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China.
| | - Yujun Liu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China.
| | - Yinglong Song
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China.
| | - Yuan Gao
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China.
| | - Fujunzhu Zhao
- Food Science Department, Pennsylvania State University, Pennsylvania, USA
| | - Yanghe Luo
- Institute of Food Research, Hezhou University, Hezhou 542899, P. R. China
| | - Fang Qian
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China.
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China. and Dalian probiotics function research key laboratory, Dalian Polytechnic University, Dalian 116034, P. R. China
| | - Yanfeng Tuo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, P. R. China.
| |
Collapse
|
44
|
Wu H, Zheng S, Zhang J, Xu S, Miao Z. Cadmium induces endoplasmic reticulum stress-mediated apoptosis in pig pancreas via the increase of Th1 cells. Toxicology 2021; 457:152790. [PMID: 33891997 DOI: 10.1016/j.tox.2021.152790] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/31/2021] [Accepted: 04/18/2021] [Indexed: 12/11/2022]
Abstract
Cadmium (Cd), an environmental pollutant, causes several adverse reactions in animals. High dose of Cd has serious cytotoxicities, including the induction of programmed cell necrosis, autophagy and apoptosis, which has aroused wide public concern. The balance of cytokine network is affected by Th1/Th2 balance which is closely related to immune response and the occurrence, development, treatment and outcome of various diseases. Cd can induce severe apoptosis, but the relationship between Cd induced apoptosis and Th1/Th2 balance has not been clarified. In this study, we established a pig Cd poisoning model, exposing to CdCl2 for 40 days (20 mg Cd/kg diet). Firstly, deviation of Th1/Th2 balance was observed by fluorescence staining, and apoptosis was observed by TUNEL staining. Then, real-time fluorescence quantitative analysis and Western blot were used to detect the expression of related proteins. The results show that Cd can interfere with the balance of Th1/Th2 and shift the balance towards Th1. In addition, through the experiments, we found that Cd exposure can increase the expression of glucose-regulated protein 94 (GRP94) and glucose-regulated protein 78 (GRP78), marker proteins of unfolded protein response (UPR). Cd exposure can increase the expression of pancreatic endoplasmic reticulum kinase (PERK), CCAAT-enhancer-binding protein homologous protein (CHOP), inositol-requiring enzyme 1 (IRE-1), activating transcription factor 6 (ATF-6), cysteinyl aspartate specific proteinase (Caspase12), indicating the three branches (ATF6, PERK and IRE-1) of endoplasmic reticulum stress (ER-stress) were activated. Moreover, we found that the expression of pro-apoptosis genes in the downstream pathway of ER-stress increased. In summary, our results indicated that Cd exposure upregulated the expression of pro-apoptosis related genes and caused apoptosis via the activation of the ER-stress signaling pathways in pancreas cells. And these negative effects were correlated with the equilibrium drift of Th1/Th2, increase in the expression and secretion of Th1 cytokines.
Collapse
Affiliation(s)
- Hao Wu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shufang Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jinxi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Zhiruo Miao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
45
|
Patel F, Parwani K, Patel D, Mandal P. Metformin and Probiotics Interplay in Amelioration of Ethanol-Induced Oxidative Stress and Inflammatory Response in an In Vitro and In Vivo Model of Hepatic Injury. Mediators Inflamm 2021; 2021:6636152. [PMID: 33953643 PMCID: PMC8064785 DOI: 10.1155/2021/6636152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/26/2021] [Accepted: 03/14/2021] [Indexed: 02/07/2023] Open
Abstract
Alcohol-induced liver injury implicates inflammation and oxidative stress as important mediators. Despite rigorous research, there is still no Food and Drug Administration (FDA) approved therapies for any stage of alcoholic liver disease (ALD). Interestingly, metformin (Met) and several probiotic strains possess the potential of inhibiting alcoholic liver injury. Therefore, we investigated the effectiveness of combination therapy using a mixture of eight strains of lactic acid-producing bacteria, commercialized as Visbiome® (V) and Met in preventing the ethanol-induced hepatic injury using in vitro and in vivo models. Human HepG2 cells and male Wistar rats were exposed to ethanol and simultaneously treated with probiotic V or Met alone as well as in combination. Endoplasmic reticulum (ER) stress markers, inflammatory markers, lipid metabolism, reactive oxygen species (ROS) production, and oxidative stress were evaluated, using qRT-PCR, Oil red O staining, fluorimetry, and HPLC. In vitro, probiotic V and Met in combination prevented ethanol-induced cellular injury, ER stress, oxidative stress, and regulated lipid metabolism as well as inflammatory response in HepG2 cells. Probiotic V and Met also promoted macrophage polarization towards the M2 phenotype in ethanol-exposed RAW 264.7 macrophage cells. In vivo, combined administration of probiotic V and Met ameliorated the histopathological changes, inflammatory response, hepatic markers (liver enzymes), and lipid metabolism induced by ethanol. It also improved the antioxidant markers (HO-1 and Nrf-2), as seen by their protein levels in both HepG2 cells as well as liver tissue using ELISA. Hence, probiotic V may act, in addition to the Met, as an effective preventive treatment against ethanol-induced hepatic injury.
Collapse
Affiliation(s)
- Farhin Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, 388421 Anand, Gujarat, India
| | - Kirti Parwani
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, 388421 Anand, Gujarat, India
| | - Dhara Patel
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, 388421 Anand, Gujarat, India
| | - Palash Mandal
- Department of Biological Sciences, P. D. Patel Institute of Applied Sciences, Charotar University of Science and Technology, Changa, 388421 Anand, Gujarat, India
| |
Collapse
|
46
|
Yin S, Li L, Tao Y, Yu J, Wei S, Liu M, Li J. The Inhibitory Effect of Artesunate on Excessive Endoplasmic Reticulum Stress Alleviates Experimental Colitis in Mice. Front Pharmacol 2021; 12:629798. [PMID: 33767628 PMCID: PMC7985062 DOI: 10.3389/fphar.2021.629798] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 01/14/2021] [Indexed: 12/13/2022] Open
Abstract
Endoplasmic reticulum (ER) stress may contribute to the pathogenesis and perpetuation of ulcerative colitis (UC). Previous studies have shown artesuante (ARS) has the protective effect on experimental UC. Therefore, it can be assumed that ARS can regulate ER stress and its related reactions. Dextran sulfate sodium (DSS) induced UC model in mice was used to testify this hypothesis. The results clearly showed that DSS exposure caused excessive ER stress evidenced by a markedly increase of GRP78 and CHOP expression, and then activated the ER stress sensors PERK, IRE1, ATF6 and their respective signaling pathways, followed by upregulated caspases12 and lowered Bcl-2/Bax ratio. However, ARS treatment significantly inhibited the occurrence of ER stress via preventing the activation of PERK-eIF2α-ATF4-CHOP and IRE1α-XBP1 signaling pathways, concurrently ER-stress-associated apoptosis in colon tissues. Moreover, ARS treatment remarkably inhibited the activation of NF-κB and the expression levels of pro-inflammatory cytokines, improved the clinical and histopathological alterations as well as maintained the expression of claudin-1 and Muc2 in mucosal layer of colon. Notably, the classic ER stress inhibitor 4-phenyhlbutyric acid enhanced the beneficial effects of ARS; in contrast, the ER stress inducer 2-deoxy-d-glucose substantially abrogated the above-mentioned effects, uncovering the involvement of ER stress in the response. These findings indicated the protection of ARS on UC is associated with its suppressing excessive ER stress mediated intestinal barrier damage and inflammatory response. This study provides a novel aspect to understand the mechanism of ARS against UC.
Collapse
Affiliation(s)
- Shaojie Yin
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Liuhui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Ya Tao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jie Yu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Simin Wei
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Mingjiang Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Jingui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
47
|
Spangler JR, Caruana JC, Medintz IL, Walper SA. Harnessing the potential of Lactobacillus species for therapeutic delivery at the lumenal-mucosal interface. Future Sci OA 2021; 7:FSO671. [PMID: 33815818 PMCID: PMC8015674 DOI: 10.2144/fsoa-2020-0153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lactobacillus species have been studied for over 30 years in their role as commensal organisms in the human gut. Recently there has been a surge of interest in their abilities to natively and recombinantly stimulate immune activities, and studies have identified strains and novel molecules that convey particular advantages for applications as both immune adjuvants and immunomodulators. In this review, we discuss the recent advances in Lactobacillus-related activity at the gut/microbiota interface, the efforts to probe the boundaries of the direct and indirect therapeutic potential of these bacteria, and highlight the continued interest in harnessing the native capacity for the production of biogenic compounds shown to influence nervous system activity. Taken together, these aspects underscore Lactobacillus species as versatile therapeutic delivery vehicles capable of effector production at the lumenal-mucosal interface, and further establish a foundation of efficacy upon which future engineered strains can expand.
Collapse
Affiliation(s)
- Joseph R Spangler
- National Research Council Postdoctoral Fellow sited in US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Julie C Caruana
- American Society for Engineering Education Postdoctoral Fellow sited in US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Igor L Medintz
- US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| | - Scott A Walper
- US Naval Research Laboratory, Code 6900, Center for Bio/Molecular Science & Engineering, 4555 Overlook Ave SW, Washington DC, 20375, USA
| |
Collapse
|
48
|
Eugene SP, Reddy VS, Trinath J. Endoplasmic Reticulum Stress and Intestinal Inflammation: A Perilous Union. Front Immunol 2020; 11:543022. [PMID: 33324392 PMCID: PMC7723926 DOI: 10.3389/fimmu.2020.543022] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
The intestinal tract encompasses the largest mucosal surface fortified with a fine layer of intestinal epithelial cells along with highly sophisticated network of the lamina propria immune cells that are indispensable to sustain gut homeostasis. However, it can be challenging to uphold homeostasis when these cells in the intestine are perpetually exposed to insults of both endogenous and exogenous origin. The complex networking and dynamic microenvironment in the intestine demand highly functional cells ultimately burdening the endoplasmic reticulum (ER) leading to ER stress. Unresolved ER stress is one of the primary contributors to the pathogenesis of inflammatory bowel diseases (IBD). Studies also suggest that ER stress can be the primary cause of inflammation and/or the consequence of inflammation. Therefore, understanding the patterns of expression of ER stress regulators and deciphering the intricate interplay between ER stress and inflammatory pathways in intestinal epithelial cells in association with lamina propria immune cells contribute toward the development of novel therapies to tackle IBD. This review provides imperative insights into the molecular markers involved in the pathogenesis of IBD by potentiating ER stress and inflammation and briefly describes the potential pharmacological intervention strategies to mitigate ER stress and IBD. In addition, genetic mutations in the biomarkers contributing to abnormalities in the ER stress signaling pathways further emphasizes the relevance of biomarkers in potential treatment for IBD.
Collapse
Affiliation(s)
- Sanchez Preethi Eugene
- Department of Biological Sciences, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, India
| | | | - Jamma Trinath
- Department of Biological Sciences, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad, India
| |
Collapse
|
49
|
Zong S, Ye Z, Zhang X, Chen H, Ye M. Protective effect of Lachnum polysaccharide on dextran sulfate sodium-induced colitis in mice. Food Funct 2020; 11:846-859. [PMID: 31934694 DOI: 10.1039/c9fo02719j] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Inflammatory bowel disease (IBD) has been gradually considered as a public health challenge worldwide. This study determined the protective effect of Lachnum polysaccharide (LEP) on dextran sulfate sodium (DSS)-induced experimental colitis in mice and explored the underlying mechanism. Results showed that dietary LEP reduced DSS-induced disease activity index (DAI), colon shortening and colonic tissue damage. LEP treatment restored intestinal barrier integrity by regulating the expression of tight junction proteins and mucus layer protecting proteins. Moreover, pro-inflammatory cytokine production was inhibited by LEP through regulating PPARγ/NF-κB and IL-6/STAT3 pathways and inhibiting inflammatory cell infiltration. In addition, LEP also inhibited (NOD)-like receptor family pyrin domain containing 3 (NLRP3) inflammasome activation, endoplasmic reticulum (ER) stress and oxidative/nitrosative stress induced by DSS. These results provided a scientific basis for LEP as a potential natural agent for protecting mice from DSS-induced IBD.
Collapse
Affiliation(s)
- Shuai Zong
- School of Food Science and Engineering, Hefei University of Technology, Hefei, Anhui 230009, P.R. China.
| | | | | | | | | |
Collapse
|
50
|
Yang GY, Xia B, Su JH, He T, Liu X, Guo L, Zhang S, Zhu YH, Wang JF. Anti-inflammatory effects of Lactobacillus johnsonii L531 in a pig model of Salmonella Infantis infection involves modulation of CCR6 + T cell responses and ER stress. Vet Res 2020; 51:26. [PMID: 32093767 PMCID: PMC7041187 DOI: 10.1186/s13567-020-00754-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 02/03/2020] [Indexed: 12/13/2022] Open
Abstract
Probiotic pretreatment is an effective non-antibiotic strategy for preventing or controlling Salmonella infections. We found that Lactobacillus johnsonii L531, isolated from the colon of a clinically healthy weaned piglet, effectively prevented infection with Salmonella enterica serovar Infantis in a pig model. Newly weaned piglets were intragastrically administered Lactobacillus johnsonii L531 at 1.0 × 1010 CFU/day for 1 week before S. Infantis challenge. Pretreatment with L. johnsonii L531 lessened the severity of diarrhea and ileal inflammation in S. Infantis-infected piglets. Lactobacilli were more abundant in the ileum than jejunum after L. johnsonii L531 pretreatment. Treatment with L. johnsonii L531 reduced the abundance of total bacteria in the ileal mucosa and the production of lipocalin 2 in the jejunum of piglets challenged with Salmonella. Both intestinal morphology and transmission electron microscopy results indicated that L. johnsonii L531 alleviated intestinal tissue damage following S. Infantis challenge, especially in the villus and endoplasmic reticulum (ER). ER stress induced by S. Infantis was attenuated by L. johnsonii L531 treatment. The number of CD4- CCR6+ T cells decreased following S. Infantis challenge, but the percentage of CCR6- IFNγ+ T cells in peripheral blood increased. In intestinal mesenteric lymph nodes, S. Infantis increased the proportion of CCR6+ IFNγ+ T cells, whereas L. johnsonii L531 induced an increase in the proportion of CD4+ CCR6+ T cells in response to S. Infantis infection. Our data thus suggest that L. johnsonii L531 contributes to the maintenance of intestinal homeostasis by modulating T-cell responses and ER stress.
Collapse
Affiliation(s)
- Gui-Yan Yang
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Bing Xia
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jin-Hui Su
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Ting He
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xiao Liu
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Liang Guo
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Shuai Zhang
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yao-Hong Zhu
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Jiu-Feng Wang
- College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|