1
|
Li W, Zeng Y, Zhong J, Hu Y, Xiong X, Zhou Y, Fu L. Probiotics Exert Gut Immunomodulatory Effects by Regulating the Expression of Host miRNAs. Probiotics Antimicrob Proteins 2025:10.1007/s12602-024-10443-9. [PMID: 39754704 DOI: 10.1007/s12602-024-10443-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2024] [Indexed: 01/06/2025]
Abstract
Probiotics exert a diverse range of immunomodulatory effects on the human gut immune system. These mechanisms encompass strengthening the intestinal mucosal barrier, inhibiting pathogen adhesion and colonization, stimulating immune modulation, and fostering the production of beneficial substances. As a result, probiotics hold significant potential in the prevention and treatment of various conditions, including inflammatory bowel disease and colorectal cancer. A pivotal mechanism by which probiotics achieve these effects is through modulating the expression of host miRNAs. miRNAs, non-coding RNA molecules, are vital regulators of fundamental biological processes like cell growth, differentiation, and apoptosis. By interacting with mRNAs, miRNAs can either promote their degradation or repress their translation, thereby regulating gene expression post-transcriptionally and modulating the immune system. This review provides a comprehensive overview of how probiotics modulate gut immune responses by altering miRNA expression levels, both upregulating and downregulating specific miRNAs. It further delves into how this modulation impacts the host's resistance to pathogens and susceptibility to diseases, offering a theoretical foundation and practical insights for the clinical utilization of probiotics in disease prevention and therapy.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Yongwei Zeng
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Jiayu Zhong
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Youyu Hu
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China
| | - Xia Xiong
- Department of Dermatology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yingshun Zhou
- Department of Pathogenic Biology, School of Basic Medical Sciences, Southwest Medical University, Luzhou, 646000, China.
- Public Center of Experimental Technology of Pathogen Biology Technology Platform, Southwest Medical University, Luzhou, 646000, China.
| | - Li Fu
- Department of Reproductive Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
2
|
Guo J, Zhong L, Momeni MR. MicroRNA-155 and its exosomal form: Small pieces in the gastrointestinal cancers puzzle. Cell Biol Toxicol 2024; 40:77. [PMID: 39283408 PMCID: PMC11405467 DOI: 10.1007/s10565-024-09920-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/04/2024] [Indexed: 09/22/2024]
Abstract
Gastrointestinal (GI) cancers are common cancers that are responsible for a large portion of global cancer fatalities. Due to this, there is a pressing need for innovative strategies to identify and treat GI cancers. MicroRNAs (miRNAs) are short ncRNAs that can be considered either cancer-causing or tumor-inhibiting molecules. MicroRNA-155, also known as miR-155, is a vital regulator in various cancer types. This miRNA has a carcinogenic role in a variety of gastrointestinal cancers, including pancreatic, colon, and gastric cancers. Since the abnormal production of miR-155 has been detected in various malignancies and has a correlation with increased mortality, it is a promising target for future therapeutic approaches. Moreover, exosomal miR-155 associated with tumors have significant functions in communicating between cells and establishing the microenvironment for cancer in GI cancers. Various types of genetic material, such as specifically miR-155 as well as proteins found in cancer-related exosomes, have the ability to be transmitted to other cells and have a function in the advancement of tumor. Therefore, it is critical to conduct a review that outlines the diverse functions of miR-155 in gastrointestinal malignancies. As a result, we present a current overview of the role of miR-155 in gastrointestinal cancers. Our research highlighted the role of miR-155 in GI cancers and covered critical issues in GI cancer such as pharmacologic inhibitors of miRNA-155, miRNA-155-assosiated circular RNAs, immune-related cells contain miRNA-155. Importantly, we discussed miRNA-155 in GI cancer resistance to chemotherapy, diagnosis and clinical trials. Furthermore, the function of miR-155 enclosed in exosomes that are released by cancer cells or tumor-associated macrophages is also covered.
Collapse
Affiliation(s)
- Jinbao Guo
- Department of Thoracic Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Li Zhong
- Department of Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | | |
Collapse
|
3
|
Bárcenas-Preciado V, Mata-Haro V. Probiotics in miRNA-Mediated Regulation of Intestinal Immune Homeostasis in Pigs: A Physiological Narrative. Microorganisms 2024; 12:1606. [PMID: 39203448 PMCID: PMC11356641 DOI: 10.3390/microorganisms12081606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
The microbiota plays a crucial role in maintaining the host's intestinal homeostasis, influencing numerous physiological functions. Various factors, including diet, stress, and antibiotic use, can lead to such imbalances. Probiotics have been shown to restore the microbiota, contributing to maintaining this balance. For instance, the weaning stage in piglets is crucial; this transition can cause unfavorable changes that may contribute to the onset of diarrhea. Probiotic supplementation has increased due to its benefits. However, its mechanism of action is still controversial; one involves the regulation of intestinal immunity. When recognized by immune system cells through membrane receptors, probiotics activate intracellular signaling pathways that lead to changes in gene expression, resulting in an anti-inflammatory response. This complex regulatory system involves transcriptional and post-transcriptional mechanisms, including the modulation of various molecules, emphasizing microRNAs. They have emerged as important regulators of innate and adaptive immune responses. Analyzing these mechanisms can enhance our understanding of probiotic-host microbiota interactions, providing insights into their molecular functions. This knowledge can be applied not only in the swine industry, but also in studying microbiota-related disorders. Moreover, these studies serve as animal models, helping to understand better conditions such as inflammatory bowel disease and other related disorders.
Collapse
Affiliation(s)
| | - Verónica Mata-Haro
- Laboratorio de Microbiología e Inmunología, Centro de Investigación en Alimentación y Desarrollo, AC (CIAD) Carretera Gustavo E. Astiazarán 46, Col. La Victoria, Hermosillo 83304, Mexico;
| |
Collapse
|
4
|
Şahin TÖ, Yılmaz B, Yeşilyurt N, Cicia D, Szymanowska A, Amero P, Ağagündüz D, Capasso R. Recent insights into the nutritional immunomodulation of cancer-related microRNAs. Phytother Res 2023; 37:4375-4397. [PMID: 37434291 DOI: 10.1002/ptr.7937] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 06/21/2023] [Accepted: 06/24/2023] [Indexed: 07/13/2023]
Abstract
Cancer is the most common cause of death worldwide, following cardiovascular diseases. Cancer is a multifactorial disease and many reasons such as physical, chemical, biological, and lifestyle-related factors. Nutrition, which is one of the various factors that play a role in the prevention, development, and treatment of many types of cancer, affects the immune system, which is characterized by disproportionate pro-inflammatory signaling in cancer. Studies investigating the molecular mechanisms of this effect have shown that foods rich in bioactive compounds, such as green tea, olive oil, turmeric, and soybean play a significant role in positively changing the expression of miRNAs involved in the regulation of genes associated with oncogenic/tumor-suppressing pathways. In addition to these foods, some diet models may change the expression of specific cancer-related miRNAs in different ways. While Mediterranean diet has been associated with anticancer effects, a high-fat diet, and a methyl-restricted diet are considered to have negative effects. This review aims to discuss the effects of specific foods called "immune foods," diet models, and bioactive components on cancer by changing the expression of miRNAs in the prevention and treatment of cancer.
Collapse
Affiliation(s)
| | - Birsen Yılmaz
- Department of Nutrition and Dietetics, Cukurova University, Adana, Turkey
| | | | - Donatella Cicia
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Paola Amero
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Gazi University, Ankara, Turkey
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Napoli, Italy
| |
Collapse
|
5
|
Pös O, Styk J, Buglyó G, Zeman M, Lukyova L, Bernatova K, Hrckova Turnova E, Rendek T, Csók Á, Repiska V, Nagy B, Szemes T. Cross-Kingdom Interaction of miRNAs and Gut Microbiota with Non-Invasive Diagnostic and Therapeutic Implications in Colorectal Cancer. Int J Mol Sci 2023; 24:10520. [PMID: 37445698 DOI: 10.3390/ijms241310520] [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: 05/17/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Colorectal cancer (CRC) has one of the highest incidences among all types of malignant diseases, affecting millions of people worldwide. It shows slow progression, making it preventable. However, this is not the case due to shortcomings in its diagnostic and management procedure and a lack of effective non-invasive biomarkers for screening. Here, we discuss CRC-associated microRNAs (miRNAs) and gut microbial species with potential as CRC diagnostic and therapy biomarkers. We provide rich evidence of cross-kingdom miRNA-mediated interactions between the host and gut microbiome. miRNAs have emerged with the ability to shape the composition and dynamics of gut microbiota. Intestinal microbes can uptake miRNAs, which in turn influence microbial growth and provide the ability to regulate the abundance of various microbial species. In the context of CRC, targeting miRNAs could aid in manipulating the balance of the microbiota. Our findings suggest the need for correlation analysis between the composition of the gut microbiome and the miRNA expression profile.
Collapse
Affiliation(s)
- Ondrej Pös
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Geneton Ltd., 841 04 Bratislava, Slovakia
| | - Jakub Styk
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Geneton Ltd., 841 04 Bratislava, Slovakia
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Gergely Buglyó
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Michal Zeman
- Comenius University Science Park, 841 04 Bratislava, Slovakia
| | - Lydia Lukyova
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 842 05 Bratislava, Slovakia
| | - Kamila Bernatova
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 842 05 Bratislava, Slovakia
| | - Evelina Hrckova Turnova
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Slovgen Ltd., 841 04 Bratislava, Slovakia
| | - Tomas Rendek
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
| | - Ádám Csók
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Vanda Repiska
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, 811 08 Bratislava, Slovakia
- Medirex Group Academy, n.p.o., 949 05 Nitra, Slovakia
| | - Bálint Nagy
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Department of Human Genetics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Tomas Szemes
- Comenius University Science Park, 841 04 Bratislava, Slovakia
- Geneton Ltd., 841 04 Bratislava, Slovakia
- Department of Molecular Biology, Faculty of Natural Sciences, Comenius University, 842 05 Bratislava, Slovakia
| |
Collapse
|
6
|
Guedes BFS, Cardoso SM, Esteves AR. The Impact of microRNAs on Mitochondrial Function and Immunity: Relevance to Parkinson's Disease. Biomedicines 2023; 11:biomedicines11051349. [PMID: 37239020 DOI: 10.3390/biomedicines11051349] [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: 03/23/2023] [Revised: 04/21/2023] [Accepted: 04/28/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's Disease (PD), the second most common neurodegenerative disorder, is characterised by the severe loss of dopaminergic neurons in the Substantia Nigra pars compacta (SNpc) and by the presence of Lewy bodies. PD is diagnosed upon the onset of motor symptoms, such as bradykinesia, resting tremor, rigidity, and postural instability. It is currently accepted that motor symptoms are preceded by non-motor features, such as gastrointestinal dysfunction. In fact, it has been proposed that PD might start in the gut and spread to the central nervous system. Growing evidence reports that the gut microbiota, which has been found to be altered in PD patients, influences the function of the central and enteric nervous systems. Altered expression of microRNAs (miRNAs) in PD patients has also been reported, many of which regulate key pathological mechanisms involved in PD pathogenesis, such as mitochondrial dysfunction and immunity. It remains unknown how gut microbiota regulates brain function; however, miRNAs have been highlighted as important players. Remarkably, numerous studies have depicted the ability of miRNAs to modulate and be regulated by the host's gut microbiota. In this review, we summarize the experimental and clinical studies implicating mitochondrial dysfunction and immunity in PD. Moreover, we gather recent data on miRNA involvement in these two processes. Ultimately, we discuss the reciprocal crosstalk between gut microbiota and miRNAs. Studying the bidirectional interaction of gut microbiome-miRNA might elucidate the aetiology and pathogenesis of gut-first PD, which could lead to the application of miRNAs as potential biomarkers or therapeutical targets for PD.
Collapse
Affiliation(s)
- Beatriz F S Guedes
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra Morais Cardoso
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Ana Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology and CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC-Institute for Interdisciplinary Research, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
7
|
Comparison of the effects of probiotic-based formulations on growth, feed utilization, blood constituents, cecal fermentation, and duodenal morphology of rabbits reared under hot environmental conditions. ANNALS OF ANIMAL SCIENCE 2023. [DOI: 10.2478/aoas-2023-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Abstract
The present study aimed to assess the effects of three probiotic-supplemented diets on growth, cecal fermentation, blood biochemical, and intestinal morphological features in growing rabbits reared under summer conditions. Rabbits were allotted into four groups: G1 rabbits were fed the basal diet (control), G2 rabbits received Enterococcus faecium (EF) and Clostridium butyricum (CB) complexes (1 × 108 and 2.5 × 106 cfu/kg diet, respectively), G3 rabbits were given CB (2.5 × 106 cfu/kg diet) and yeast complexes (1 g/kg diet), and G4 rabbits received EF (2 × 108 cfu/kg diet) and yeast (1 g/kg diet). G2 rabbits exhibited the highest performances in terms of enhanced body weight and weight gain, protein efficiency ratio and feed conversion ratio (P<0.05). Serum total protein, globulin, immunoglobulin M, and high-density lipoprotein concentrations were higher in probiotic-fed rabbits than those in controls. Additionally, lipid profile parameters were significantly reduced in the probiotic-fed rabbits, with the lowest concentrations measured in G4 rabbits (P<0.05). Rabbits given EF and CB had the highest total volatile fatty acid (VFA) and propionic acid levels and the lowest ammonia concentrations. Increased villi length and muscular layer thickness and reduced crypt depth were observed in rabbits receiving EF and CB compared with the values obtained in controls (P<0.05). In summary, supplementing fattening rabbit diets with EF and CB, as a novel formulation, might be a promising and easy method to enhance growth performance under hot climate conditions by improving the feed utilization, immune response, serum lipid profile, cecal VFA production, and duodenal morphology.
Collapse
|
8
|
Kumar N, Sahoo NK, Mehan S, Verma B. The importance of gut-brain axis and use of probiotics as a treatment strategy for multiple sclerosis. Mult Scler Relat Disord 2023; 71:104547. [PMID: 36805171 DOI: 10.1016/j.msard.2023.104547] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 01/16/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023]
Abstract
It has been shown that the dysbiosis of the gut's microbes substantially impacts CNS illnesses, including Alzheimer's, Parkinson's, autism, and autoimmune diseases like multiple sclerosis (MS). MS is a CNS-affected autoimmune demyelination condition. Through a two-way communication pathway known as the gut-brain axis, gut microbes communicate with the CNS. When there is a disruption in the gut microbiome, cytokines and other immune cells are secreted, which affects the BBB and gastrointestinal permeability. Recent research using animal models has revealed that the gut microbiota may greatly influence the pathophysiology of EAE/MS. Any change in the gut might increase inflammatory cytokinesand affect the quantity of SCFAs, and other metabolites that cause neuroinflammation and demyelination. In- vivo and in-vitro studies have concluded that probiotics affect the immune system and can be utilized to treat gastrointestinal dysbiosis. Any alteration in the gut microbial composition caused by probiotic intake may serve as a preventive and treatment strategy for MS. The major goal of this review is to emphasize an overview of recent research on the function of gut microbiota in the onset of MS and how probiotics have a substantial impact on gastrointestinal disruption in MS and other neuro disorders. It will be easier to develop new therapeutic approaches, particularly probiotic-based supplements, for treating multiple sclerosis (MS) if we know the link between the gut and CNS.
Collapse
Affiliation(s)
- Nitish Kumar
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India.
| | - Nalini Kanta Sahoo
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, (An Autonomous College), Moga, Punjab 142001, India
| | - Bharti Verma
- SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology (Deemed to be University), Delhi-NCR Campus, Modinagar, Ghaziabad, Uttar Pradesh 201204, India
| |
Collapse
|
9
|
Yu W, Nan X, Schroyen M, Wang Y, Zhou M, Tang X, Xiong B. Effect of inulin on small extracellular vesicles microRNAs in milk from dairy cows with subclinical mastitis. J Anim Sci 2023; 101:skae366. [PMID: 39656780 DOI: 10.1093/jas/skae366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024] Open
Abstract
Milk contains microRNAs (miRNA) that are shielded by small extracellular vesicles (sEVs). Beyond variations among individuals, many factors including nutrition play a role in shaping miRNA expression profiles. This study is to explore milk-derived sEVs-miRNA variations induced by inulin supplementation in subclinical mastitis-suffering cows. Fourteen lactating cows diagnosed with subclinical mastitis were equally assigned to either an inulin or a control group. Apart from total mixed rations, cows in the inulin group were provided with 300 g/d inulin during the morning feeding, while the control group did not receive any supplement. Following 1 wk of adaptation and 5 wk of treatment, sEVs-miRNA were isolated from the milk of each cow. RNA is subjected to high-throughput sequencing and differentially expressed (DE) miRNA (P < 0.05 and ∣ log2FC∣> 1) were detected through bioinformatics analysis. Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were conducted to examine the target genes of DE miRNA. A sum of 350 miRNA was discovered, including 332 in the control group and 249 in the inulin group. Among these, 9 miRNA showed differential expression within the 2 groups, including 3 upregulated and 6 downregulated in the inulin group. The DE miRNA participates in regulating organismal systems, cellular processes, and signal transduction, which may affect inflammatory response and milk production. Overall, our study provides insight into the micromolecular-level mechanism of inulin in alleviating subclinical mastitis in dairy cows.
Collapse
Affiliation(s)
- Wanjie Yu
- Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Xuemei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Martine Schroyen
- Precision Livestock and Nutrition Laboratory, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, Gembloux 5030, Belgium
| | - Yue Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Mengting Zhou
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Xiangfang Tang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| | - Benhai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, P. R. China
| |
Collapse
|
10
|
Effect of Niacin on Growth Performance, Intestinal Morphology, Mucosal Immunity and Microbiota Composition in Weaned Piglets. Animals (Basel) 2021; 11:ani11082186. [PMID: 34438645 PMCID: PMC8388363 DOI: 10.3390/ani11082186] [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: 06/21/2021] [Revised: 07/12/2021] [Accepted: 07/20/2021] [Indexed: 01/24/2023] Open
Abstract
Simple Summary The protective effect of niacin on growth performance and gut health of weaned piglets and the underlying mechanism remains unclear despite it being a common additive in pig diets. The present study aimed to investigate the effect of niacin on growth performance, intestinal morphology, intestinal mucosal immunity, and colonic microbiota in weaned piglets. Our results show that niacin supplementation significantly improved the growth performance in piglets as compared with those given a niacin receptor antagonist. Niacin also significantly improved the relative abundance of beneficial bacteria in the colon and alleviate the inflammatory response in the intestinal mucosa as compared with control piglets and those given a niacin receptor antagonist. These results provide new insight into the beneficial effects of niacin on growth performance and gut health in weaned piglets. Abstract This study aimed to investigate the effects of niacin on growth performance, intestinal morphology, intestinal mucosal immunity, and colonic microbiota in weaned piglets. A total of 96 weaned piglets (Duroc × (Landrace × Yorkshire), 21-d old, 6.65 ± 0.02 kg body weight (BW)) were randomly allocated into 3 treatment groups (8 replicate pens per treatment, each pen containing 4 males; n = 32/treatment) for 14 d. Piglets were fed a control diet (CON) or the CON diet supplemented with 20.4 mg/kg niacin (NA) or an antagonist for the niacin receptor GPR109A (MPN). The results showed that NA or MPN had no effect on ADG, ADFI, G/F or diarrhea incidence compared with the CON diet. However, compared with piglets in the NA group, piglets in the MPN group had lower ADG (p = 0.042) and G/F (p = 0.055). In comparison with the control and MPN group, niacin supplementation increased the villus height and the ratio of villus height to crypt depth (p < 0.05), while decreasing the crypt depth in the duodenum (p < 0.05). Proteomics analysis of cytokines showed that niacin supplementation increased the expression of duodenal transforming growth factor-β (TGF-β), jejunal interleukin-10 (IL-10) and ileal interleukin-6 (IL-6) (p < 0.05), and reduced the expression of ileal interleukin-8 (IL-8) (p < 0.05) compared with the control diet. Piglets in the MPN group had significantly increased expression of ileal IL-6, and jejunal IL-8 and interleukin-1β (IL-1β) (p < 0.05) compared with those in the control group. Piglets in the MPN group had lower jejunal IL-10 level and higher jejunal IL-8 level than those in the NA group (p < 0.05). The mRNA abundance of duodenal IL-8 and ileal granulocyte-macrophage colony-stimulating factor (GM-CSF) genes were increased (p < 0.05), and that of ileal IL-10 transcript was decreased (p < 0.05) in the MPN group compared with both the control and NA groups. Additionally, niacin increased the relative abundance of Dorea in the colon as compared with the control and MPN group (p < 0.05), while decreasing that of Peptococcus compared with the control group (p < 0.05) and increasing that of Lactobacillus compared with MPN supplementation (p < 0.05). Collectively, the results indicated that niacin supplementation efficiently ensured intestinal morphology and attenuated intestinal inflammation of weaned piglets. The protective effects of niacin on gut health may be associated with increased Lactobacillus and Dorea abundance and butyrate content and decreased abundances of Peptococcus.
Collapse
|
11
|
He Y, Liu X, Dong Y, Lei J, Ito K, Zhang B. Enterococcus faecium PNC01 isolated from the intestinal mucosa of chicken as an alternative for antibiotics to reduce feed conversion rate in broiler chickens. Microb Cell Fact 2021; 20:122. [PMID: 34182992 PMCID: PMC8240220 DOI: 10.1186/s12934-021-01609-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 06/09/2021] [Indexed: 12/23/2022] Open
Abstract
Background The development and utilization of probiotics had many environmental benefits for replacing antibiotics in animal production. Bacteria in the intestinal mucosa have better adhesion to the host intestinal epithelial cells compared to bacteria in the intestinal contents. In this study, lactic acid bacteria were isolated from the intestinal mucosa of broiler chickens and investigated as the substitution to antibiotic in broiler production. Results In addition to acid resistance, high temperature resistance, antimicrobial sensitivity tests, and intestinal epithelial cell adhesion, Enterococcus faecium PNC01 (E. faecium PNC01) was showed to be non-cytotoxic to epithelial cells. Draft genome sequence of E. faecium PNC01 predicted that it synthesized bacteriocin to perform probiotic functions and bacteriocin activity assay showed it inhibited Salmonella typhimurium from invading intestinal epithelial cells. Diet supplemented with E. faecium PNC01 increased the ileal villus height and crypt depth in broiler chickens, reduced the relative length of the cecum at day 21, and reduced the relative length of jejunum and ileum at day 42. Diet supplemented with E. faecium PNC01 increased the relative abundance of Firmicutes and Lactobacillus, decreased the relative abundance of Bacteroides in the cecal microbiota. Conclusion E. faecium PNC01 replaced antibiotics to reduce the feed conversion rate. Furthermore, E. faecium PNC01 improved intestinal morphology and altered the composition of microbiota in the cecum to reduce feed conversion rate. Thus, it can be used as an alternative for antibiotics in broiler production to avoid the adverse impact of antibiotics by altering the gut microbiota. Graphic Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12934-021-01609-z.
Collapse
Affiliation(s)
- Yang He
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China.,College of Veterinary Medicine, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Xuan Liu
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Yuanyang Dong
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Jiaqi Lei
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China
| | - Koichi Ito
- Department of Food and Physiological Models, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 3145 Ago, Kasama, Ibaraki, 319-0206, Japan
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition, Department of Animal Nutrition & Feed Science, College of Animal Science & Technology, China Agricultural University, Haidian District, Beijing, 100193, China.
| |
Collapse
|
12
|
Zhao L, Ye Y, Gu L, Jian Z, Stary CM, Xiong X. Extracellular vesicle-derived miRNA as a novel regulatory system for bi-directional communication in gut-brain-microbiota axis. J Transl Med 2021; 19:202. [PMID: 33975607 PMCID: PMC8111782 DOI: 10.1186/s12967-021-02861-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 11/27/2020] [Indexed: 02/08/2023] Open
Abstract
The gut-brain-microbiota axis (GBMAx) coordinates bidirectional communication between the gut and brain, and is increasingly recognized as playing a central role in physiology and disease. MicroRNAs are important intracellular components secreted by extracellular vesicles (EVs), which act as vital mediators of intercellular and interspecies communication. This review will present current advances in EV-derived microRNAs and their potential functional link with GBMAx. We propose that EV-derived microRNAs comprise a novel regulatory system for GBMAx, and a potential novel therapeutic target for modifying GBMAx in clinical therapy.
Collapse
Affiliation(s)
- Liang Zhao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Creed M Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
13
|
Wang Q, Sun Q, Wang J, Qiu X, Qi R, Huang J. Lactobacillus Plantarum 299v Changes miRNA Expression in the Intestines of Piglets and Leads to Downregulation of LITAF by Regulating ssc-miR-450a. Probiotics Antimicrob Proteins 2021; 13:1093-1105. [PMID: 33486700 DOI: 10.1007/s12602-021-09743-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 11/26/2022]
Abstract
Lactiplantibacillus plantarum subsp. plantarum 299v (L. plantarum 299v) is one of the most important probiotic strains in animal health, but the molecular mechanisms of how it exerts health benefits remain unclear. The purpose of this study was to explore the changes in miRNA expression profiles in the intestinal tissues of piglets by L. plantarum 299v and to explore its possible molecular regulatory mechanism in intestinal function. Neonatal piglets were orally administered L. plantarum 299v daily from 1 to 20 days old, and high-throughput sequencing was conducted to analyse the changes in miRNA expression in the jejunum and ileum. The results showed that 370 known porcine miRNAs were identified from eight libraries. Five miRNAs (ssc-miR-21-5p, -143-3p, -194b-5p, -192, and -126-3p) were highly expressed in the intestinal tissues. There were 15 differentially expressed miRNAs between the control group and the L. plantarum group, and only miR-450a was expressed differentially in both intestinal tissues. KEGG analysis revealed that the target genes of the 15 differentially expressed miRNAs were involved in 37 significantly enriched pathways (P < 0.01). Then, quantitative polymerase chain reaction confirmed that the miRNA expression was corresponded well with those from the sequencing. Luciferase reporter assays verified that lipopolysaccharide-induced TNF-α factor is a target of miR-450a. Our results also showed L. plantarum 299v could influence intestinal function by changing the levels of cytokines via miRNA expression. This is the first study to analyse differential expression miRNA profiles in intestinal tissue after L. plantarum 299v treatment and investigate the molecular regulatory mechanism of functional miRNA.
Collapse
Affiliation(s)
- Qi Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China
| | - Qian Sun
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
| | - Jing Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China
| | - Xiaoyu Qiu
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
| | - Renli Qi
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China
| | - Jinxiu Huang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China.
- Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China.
| |
Collapse
|
14
|
Davoodvandi A, Marzban H, Goleij P, Sahebkar A, Morshedi K, Rezaei S, Mahjoubin-Tehran M, Tarrahimofrad H, Hamblin MR, Mirzaei H. Effects of therapeutic probiotics on modulation of microRNAs. Cell Commun Signal 2021; 19:4. [PMID: 33430873 PMCID: PMC7798223 DOI: 10.1186/s12964-020-00668-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022] Open
Abstract
Probiotics are beneficial bacteria that exist within the human gut, and which are also present in different food products and supplements. They have been investigated for some decades, due to their potential beneficial impact on human health. Probiotics compete with pathogenic microorganisms for adhesion sites within the gut, to antagonize them or to regulate the host immune response resulting in preventive and therapeutic effects. Therefore, dysbiosis, defined as an impairment in the gut microbiota, could play a role in various pathological conditions, such as lactose intolerance, gastrointestinal and urogenital infections, various cancers, cystic fibrosis, allergies, inflammatory bowel disease, and can also be caused by antibiotic side effects. MicroRNAs (miRNAs) are short non-coding RNAs that can regulate gene expression in a post-transcriptional manner. miRNAs are biochemical biomarkers that play an important role in almost all cellular signaling pathways in many healthy and disease states. For the first time, the present review summarizes current evidence suggesting that the beneficial properties of probiotics could be explained based on the pivotal role of miRNAs. Video Abstract.
Collapse
Affiliation(s)
| | - Havva Marzban
- Department of Veterinary Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Pouya Goleij
- Department of Genetics, Faculty of Biology,Sana Institute of Higher Education, Sari, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Korosh Morshedi
- Faculty of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Samaneh Rezaei
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahjoubin-Tehran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Michael R. Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, Boston, MA 02114 USA
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
15
|
Bassiony SS, Al-Sagheer AA, El-Kholy MS, Elwakeel EA, Helal AA, Alagawany M. Evaluation of Enterococcus faecium NCIMB 11181 and Clostridium butyricum probiotic supplements in post-weaning rabbits reared under thermal stress conditions. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1941334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Samar S. Bassiony
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Adham A. Al-Sagheer
- Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Mohamed S. El-Kholy
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Eman A. Elwakeel
- Department of Animal and Fish Production, Faculty of Agriculture (El-Shatby), Alexandria University, Alexandria, Egypt
| | - Amera A. Helal
- Department of Animal Production, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Mahmoud Alagawany
- Poultry Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| |
Collapse
|
16
|
Wang Q, Sun Q, Wang J, Qiu X, Qi R, Huang J. Identification of differentially expressed miRNAs after Lactobacillus reuteri treatment in the ileum mucosa of piglets. Genes Genomics 2020; 42:1327-1338. [PMID: 32980994 DOI: 10.1007/s13258-020-00998-6] [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: 07/21/2020] [Accepted: 09/10/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND Lactobacillus reuteri I5007 possesses many excellent probiotic characteristics in piglets. miRNA plays important role in host-microbiota interactions, but the mechanism by which L. reuteri I5007 regulates intestinal function through its influence on miRNA expression is unknown. OBJECTIVE This study analyzed the miRNA expression patterns in the ileum mucosa tissue of piglets by L. reuteri I5007 treatment, aim to clarify its molecular mechanism for regulating intestinal function through miRNA. METHODS Neonatal piglets were orally administered L. reuteri I5007 or a placebo daily starting on day 1, and differential expression of ileal miRNAs was analyzed at 10 and 20 days of age by small RNA sequencing. RESULTS 361 known porcine miRNAs were identified, and ten miRNAs were highly expressed in the ileum mucosa in both treatments. Nineteen differentially expressed (DE) miRNAs were identified in response to L. reuteri treatment, and four DE miRNAs (ssc-miR-196a, -196b-5p, -1285 and -10386) were differentially expressed at both time points. The KEGG pathway analyses showed the targets of 19 DE miRNAs were involved in 63 significantly enriched pathways, including the PI3K-Akt and MAPK pathways, which were confirmed to play important roles in probiotic-host communication. L. reuteri I5007 exerted anti-inflammatory effects by influencing the levels of inflammatory cytokines. Suppressor of cytokine signalling 4 gene was the target gene of ssc-miR-196a/-196b-5p, overexpression of ssc-miR-196a/-196b-5p downregulated the mRNA expression of IL-1β and TNFα in IPEC-J2 cells. CONCLUSION Our study provides new insight into the role of miRNAs in the intestinal function of piglets after L. reuteri I5007 treatment.
Collapse
Affiliation(s)
- Qi Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing, 402460, China
| | - Qian Sun
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
| | - Jing Wang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing, 402460, China
| | - Xiaoyu Qiu
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China
| | - Renli Qi
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China.,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China.,Chongqing Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing, 402460, China
| | - Jinxiu Huang
- Chongqing Academy of Animal Sciences, Rongchang, Chongqing, 402460, China. .,Key Laboratory of Pig Industry Sciences, Ministry of Agriculture, Rongchang, Chongqing, 402460, China. .,Chongqing Key Laboratory of Pig Industry Sciences, Rongchang, Chongqing, 402460, China.
| |
Collapse
|
17
|
Zhao Y, Zeng D, Wang H, Sun N, Xin J, Yang H, Lei L, Khalique A, Rajput DS, Pan K, Shu G, Jing B, Ni X. Analysis of miRNA Expression in the Ileum of Broiler Chickens During Bacillus licheniformis H2 Supplementation Against Subclinical Necrotic Enteritis. Probiotics Antimicrob Proteins 2020; 13:356-366. [PMID: 32975724 DOI: 10.1007/s12602-020-09709-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2020] [Indexed: 01/20/2023]
Abstract
Subclinical necrotic enteritis (SNE) is one of the serious threats to the poultry industry. Probiotics have been proven to exert beneficial effects in controlling SNE. However, their exact mechanisms have not been fully elucidated. Moreover, few studies have focused on their impact on microRNAs (miRNAs). Therefore, the present study aimed to explore the miRNA expression profiles in the ileum of broiler chickens during probiotic supplementation for controlling SNE. A total of 180 newly hatched male broilers were randomly allocated into three groups, including a negative control group, an SNE infection group, and a Bacillus licheniformis H2 pretreatment group. Illumina high-throughput sequencing was conducted to identify the miRNA expression of the three groups. Results showed that 628 miRNAs, including 582 known miRNAs and 46 novel miRNAs, were detected in the miRNA libraries. The target genes of 57 significantly differentially expressed miRNAs were predicted and annotated. Moreover, they were found to be partly enriched in pathways related to immunity and inflammation such as tumor necrosis factor receptor binding, immune response-regulating signaling pathway, Toll-like receptor 2 signaling pathway, interleukin-15 production, activation of NF-κB-inducing kinase activity, and MAP kinase tyrosine/serine/threonine phosphatase activity. Some of the target genes of 57 miRNAs were related to the MAPK signaling pathway. Furthermore, the expression of several miRNAs, which may be involved in the MAPK signaling pathway, was significantly affected by SNE induction and showed no significant difference in the presence of H2. All these findings provide comprehensive miRNA expression profiles of three different treatment groups. They further suggest that H2 could exert beneficial effects in controlling SNE through immune and inflammatory response associated with altered miRNA expression, such as the MAPK signaling pathway.
Collapse
Affiliation(s)
- Ying Zhao
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Dong Zeng
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hesong Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Institute of Gastroenterology of Guangdong Province, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Sun
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Jinge Xin
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Hanbo Yang
- Chengdu Slan Biotechnology Co., Ltd, Chengdu, China
| | - Lei Lei
- Chengdu Slan Biotechnology Co., Ltd, Chengdu, China
| | - Abdul Khalique
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Danish Sharafat Rajput
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Kangcheng Pan
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Gang Shu
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Bo Jing
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Xueqin Ni
- Animal Microecology Institute, College of Veterinary, Sichuan Agricultural University, Chengdu, Sichuan, China.
| |
Collapse
|
18
|
Heydari Z, Rahaie M, Alizadeh AM, Agah S, Khalighfard S, Bahmani S. Effects of Lactobacillus acidophilus and Bifidobacterium bifidum Probiotics on the Expression of MicroRNAs 135b, 26b, 18a and 155, and Their Involving Genes in Mice Colon Cancer. Probiotics Antimicrob Proteins 2020; 11:1155-1162. [PMID: 30311185 DOI: 10.1007/s12602-018-9478-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A wide range of sources supports that the link between diet and colorectal cancer may be due to an imbalance of the intestinal microflora. In this case, it seems that the probiotics may have a possible molecular mechanism via microRNAs (miRNAs). The present study is aimed to evaluate the effects of Lactobacillus acidophilus and Bifidobacterium bifidum probiotics on the expression of miRNAs 135b, 26b, 18a, and 155 and their target genes, including APC, PTEN, KRAS, and PU.1 in mouse azoxymethane (AOM)-induced colon cancer. Thirty-eight male BALB/c mice were randomly divided into four groups: the control, AOM, Lactobacillus acidophilus, and Bifidobacterium bifidum to deliberate the effects of the probiotics on the miRNAs and their target genes. Except for the control group, the rest groups were weekly given AOM (15 mg/kg, s.c) in three consecutive weeks to induce mouse colon cancer. The animals were given 1.5 g powders of L. acidophilus (1 × 109 cfu/g) and B. bifidum (1 × 109 cfu/g) in 30 cc drinking water in the related groups for 5 months. At the end of the study, the animals were sacrificed and their blood and colon samples were removed for the molecular analyses. The results showed that the expression of the miR-135b, miR-155, and KRAS was increased in the AOM group compared to the control group in both the plasma and the colon tissue samples, and the consumption of the probiotics decreased their expression. Moreover, the miR-26b, miR-18a, APC, PU.1, and PTEN expressions were decreased in the AOM group compared to the control group and the consumption of the probiotics increased their expressions. It seems that Lactobacillus acidophilus and Bifidobacterium bifidum though increasing the expression of the tumor suppressor miRNAs and their target genes and decreasing the oncogenes can improve colon cancer treatment.
Collapse
Affiliation(s)
- Zahra Heydari
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mahdi Rahaie
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Ali Mohammad Alizadeh
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, 1419733141, Iran.
- Cancer Biology Research Center, Tehran University of Medical Science, Tehran, Iran.
| | - Shahram Agah
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran.
| | - Solmaz Khalighfard
- Cancer Research Center, Tehran University of Medical Sciences, Tehran, 1419733141, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Bahmani
- Research and Development Department, Zist Takhmir Company, Tehran, Iran
| |
Collapse
|
19
|
Behrouzi A, Ashrafian F, Mazaheri H, Lari A, Nouri M, Riazi Rad F, Hoseini Tavassol Z, Siadat SD. The importance of interaction between MicroRNAs and gut microbiota in several pathways. Microb Pathog 2020; 144:104200. [PMID: 32289465 DOI: 10.1016/j.micpath.2020.104200] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
The human gut harbors diverse microbes that play a fundamental role in the well-being of their host. Microbiota disruption affects the immune function, metabolism, and causes several diseases. Therefore, understanding how the microbiome is adjusted, and identifying methods for manipulating it is critical. Studies have found that there is an inverse association between MicroRNAs (miRNAs) abundance and microbe abundance. miRNAs are known to be engaged in post-transcription regulation of cell-autonomous gene expression. Recently, they have gained great attention for their proposed roles in cell-to-cell communication, and as biomarkers for human disease. Here, we review recent studies on the role of miRNAs as a component of outer membrane vesicles (OMVs) in the composition of gut microbiota and their significance in the human situation of health and diseases and discuss their effect on inflammatory responses and dysbiosis. Further, we explain how probiotics exert influence on the expression of miRNAs.
Collapse
Affiliation(s)
- Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Ashrafian
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hoora Mazaheri
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Arezou Lari
- Systems Biomedicine Unit, Pasteur Institute of Iran, Tehran, Iran
| | - Matineh Nouri
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Farhad Riazi Rad
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Hoseini Tavassol
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran; Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran; Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
20
|
Quan SY, Nan XM, Wang K, Zhao YG, Jiang LS, Yao JH, Xiong BH. Replacement of forage fiber with non-forage fiber sources in dairy cow diets changes milk extracellular vesicle-miRNA expression. Food Funct 2020; 11:2154-2162. [PMID: 32073015 DOI: 10.1039/c9fo03097b] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Milk is a dynamic source of nutrients and bioactive factors, varying with the nutrition status of the cattle. We partly replaced alfalfa hay with whole cotton seed and soybean hull (non-forage fiber source, NFFS) in the feed formula of treated cows and evaluated the effects on milk extracellular vesicles (EVs). The NFFS supplement did not affect the shape of milk EVs observed using a transmission electron microscope. Nanoparticle tracking analysis revealed that the EV concentration increased significantly in treated cows (P = 0.019), with the peak diameter unaffected by the treatment. The EV-RNA concentration and small RNA content, particularly rRNAs and tRNAs, significantly increased in the treated cows (P < 0.05). The other small RNAs, i.e. miRNAs, cis-regulatory elements, snRNAs, and other Rfam RNAs showed no significant difference between the two groups. Totally 276 milk EV-miRNAs were identified. Thirteen miRNAs, accounting for 76%, in the highly expressed top 20, were immune-related. In addition, 9 differently expressed miRNAs (4 up-regulated and 5 down-regulated) were identified (P < 0.05). Kyoto Encyclopedia of Genes and Genomes pathway analysis showed that the differently expressed miRNAs were related to the citrate cycle, fat digestion and absorption process, taurine and hypo-taurine metabolism, and glycosphingolipid biosynthesis. This study documents the milk nutrition assessment from macromolecules, especially EVs.
Collapse
Affiliation(s)
- Su-Yu Quan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China. and College of Animal Science and Technology, Northwest A&F University, Yanglin 712100, China.
| | - Xue-Mei Nan
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Kun Wang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Yi-Guang Zhao
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Lin-Shu Jiang
- Beijing Key Laboratory for Dairy Cow Nutrition, Beijing University of Agriculture, Beijing 102206, China
| | - Jun-Hu Yao
- College of Animal Science and Technology, Northwest A&F University, Yanglin 712100, China.
| | - Ben-Hai Xiong
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
21
|
Shin D, Chang SY, Bogere P, Won K, Choi JY, Choi YJ, Lee HK, Hur J, Park BY, Kim Y, Heo J. Beneficial roles of probiotics on the modulation of gut microbiota and immune response in pigs. PLoS One 2019; 14:e0220843. [PMID: 31461453 PMCID: PMC6713323 DOI: 10.1371/journal.pone.0220843] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
The importance of probiotics in swine production is widely acknowledged as crucial. However, gaps still remain in the exact roles played by probiotics in modulation of gut microbiota and immune response. This study determined the roles of probiotic Lactobacillus plantarum strain JDFM LP11in gut microbiota modulation and immune response in weaned piglets. L. plantarum JDFM LP11 increased the population of lactic acid bacteria in feces and enhanced the development of villi in the small intestine. Metagenome analysis showed that microbial diversity and richness (Simpson, Shannon, ACE, Chao1) and the relative abundance of the Firmicutes were higher in weaned piglets fed probiotics. Five bacterial families were different in the relative abundance, especially; Prevotellaceae occupied the largest part of microbial community showed the most difference between two groups. Transcriptome analysis identified 25 differentially expressed genes using RNA-sequencing data of the ileum. Further gene ontology and immune DB analysis determined 8 genes associated with innate defense response and cytokine production. BPI, RSAD2, SLPI, LUM, OLFM4, DMBT1 and C6 genes were down-regulated by probiotic supplementation except PLA2G2A. PICRUSt analysis predicting functional profiling of microbial communities indicated branched amino acid biosynthesis and butyrate metabolism promoting gut development and health were increased by probiotics. Altogether, our data suggest that L. plantarum JDFM LP11 increases the diversity and richness in the microbial community, and attenuates the ileal immune gene expression towards gut inflammation, promoting intestinal development in weaned piglets.
Collapse
Affiliation(s)
- Donghyun Shin
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, Republic of Korea
| | - Sung Yong Chang
- Department of Animal Science and Institute of Milk Genomics, Chonbuk National University, Jeonju, Republic of Korea
| | - Paul Bogere
- Department of Agricultural Convergence Technology, Chonbuk National University, Jeonju, Republic of Korea
| | - KyeongHye Won
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, Republic of Korea
| | - Jae-Young Choi
- The Animal Molecular Genetics and Breeding Center, Chonbuk National University, Jeonju, Republic of Korea
| | - Yeon-Jae Choi
- International Agricultural Development and Cooperation Center, Chonbuk National University, Jeonju, Republic of Korea
| | - Hak Kyo Lee
- Department of Animal Biotechnology, Chonbuk National University, Jeonju, Republic of Korea
- The Animal Molecular Genetics and Breeding Center, Chonbuk National University, Jeonju, Republic of Korea
| | - Jin Hur
- College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Byung-Yong Park
- College of Veterinary Medicine, Chonbuk National University, Iksan, Republic of Korea
| | - Younghoon Kim
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Science, Seoul National University, Seoul, Republic of Korea
| | - Jaeyoung Heo
- International Agricultural Development and Cooperation Center, Chonbuk National University, Jeonju, Republic of Korea
- * E-mail:
| |
Collapse
|
22
|
Heydari Z, Rahaie M, Alizadeh AM. Different anti-inflammatory effects of Lactobacillus acidophilus and Bifidobactrum bifidioum in hepatocellular carcinoma cancer mouse through impact on microRNAs and their target genes. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2019. [DOI: 10.1016/j.jnim.2019.100096] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
23
|
Wang J, Teng Y, Zhao G, Li F, Hou A, Sun B, Kong W, Gao F, Cai L, Jiang C. Exosome-Mediated Delivery of Inducible miR-423-5p Enhances Resistance of MRC-5 Cells to Rabies Virus Infection. Int J Mol Sci 2019; 20:ijms20071537. [PMID: 30934732 PMCID: PMC6479321 DOI: 10.3390/ijms20071537] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/19/2022] Open
Abstract
The human diploid cell line Medical Research Council -5 (MRC-5) is commonly utilized for vaccine development. Although a rabies vaccine developed in cultured MRC-5 cells exists, the poor susceptibility of MRC-5 cells to the rabies virus (RABV) infection limits the potential yield of this vaccine. The underlying mechanism of MRC-5 cell resistance to RABV infection remains unknown. In this study, we demonstrate that viral infection increased exosomal release from MRC-5 cells; conversely, blocking exosome release promoted RABV infection in MRC-5 cells. Additionally, RABV infection up-regulated microRNA (miR)-423-5p expression in exosomes, resulting in feedback inhibition of RABV replication by abrogating the inhibitory effect of suppressor of cytokine signaling 3 (SOCS3) on type I interferon (IFN) signaling. Furthermore, intercellular delivery of miR-423-5p by exosomes inhibited RABV replication in MRC-5 cells. We also show that RABV infection increased IFN-β production in MRC-5 cells and that blocking the type I IFN receptor promoted RABV infection. In conclusion, MRC-5 cells were protected from RABV infection by the intercellular delivery of exosomal miR-423-5p and the up-regulation of IFN-β. These findings reveal novel antiviral mechanisms in MRC-5 cells against RABV infection. miR-423-5p, exosomes, and IFN signaling pathways may therefore be potential targets for improving MRC-5 cell-based rabies vaccine production.
Collapse
Affiliation(s)
- Jingyu Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Yawei Teng
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Guanshu Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Fang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
| | - Ali Hou
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Bo Sun
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Feng Gao
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Linjun Cai
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun 130012, China.
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Science, Jilin University, Changchun 130012, China.
| |
Collapse
|
24
|
Wu Y, Zhen W, Geng Y, Wang Z, Guo Y. Effects of dietary Enterococcus faecium NCIMB 11181 supplementation on growth performance and cellular and humoral immune responses in broiler chickens. Poult Sci 2019; 98:150-163. [PMID: 30137622 DOI: 10.3382/ps/pey368] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 12/26/2022] Open
Abstract
This study evaluated the effects of dietary Enterococcus faecium NCIMB 11181 on growth performance and immune response in broiler chickens. A total of 360 1-day-old Arbor Acres male birds were randomly assigned to 4 treatments that administered different dosages of E. faecium (0, 5 × 107, 1 × 108, and 2 × 108 CFU E. faecium/kg diet). The results revealed that average daily gain (ADG) changed quadratically, while feed conversion rate (FCR) increased linearly from day 22 to 35 and day 1 to 35 (P < 0.05). Supplementation of E. faecium at 5 × 107CFU/kg diet resulted in increased ADG (P < 0.05) compared with the other groups. Birds fed with 2 × 108 CFU/kg E. faecium exhibited increased peripheral blood lymphocyte proliferation in response to concanavalin A (Con A) (P < 0.05) at day 35 and enhanced skin responses following phytohemagglutinin (PHA) injection (P < 0.05) at 12 h. Serum lysozyme activity at day 21 increased linearly with dietary E. faecium concentration (P < 0.05), the highest activity was observed in the 1 × 108 and the 2 × 108 CFU E. faecium groups (P < 0.01). Serum levels of proinflammatory cytokines IL-1β, IL-2, IL-6, IFN-γ, and anti-inflammatory IL-4, IL-10 changed linearly or quadratically both at the initial and final phases (P < 0.05). In addition, BSA antibody titers were significantly increased following both primary and secondary inoculation when birds were fed with 1 × 108 or 2 × 108 CFU/kg E. faecium (P < 0.05). In comparison with other groups, birds received 5 × 107 CFU E. faecium exhibited the highest levels of serum IgG (P < 0.05) at day 35. Together, our results revealed that broiler diet supplemented with 5 × 107 CFU/kg E. faecium NCIMB 11181 was appropriate in relation to growth performance under normal conditions. Upon administration with higher dosages of E. faecium NCIMB 11181, obvious immune-stimulatory effects were observed following both cell-mediated and humoral immunity.
Collapse
Affiliation(s)
- Yuanyuan Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, 100093 Beijing, China
| | - Wenrui Zhen
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, 100093 Beijing, China
| | - Yanqiang Geng
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, 100093 Beijing, China
| | - Zhong Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, 100093 Beijing, China
| | - Yuming Guo
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, 100093 Beijing, China
| |
Collapse
|
25
|
Llewellyn A, Foey A. Probiotic Modulation of Innate Cell Pathogen Sensing and Signaling Events. Nutrients 2017; 9:E1156. [PMID: 29065562 PMCID: PMC5691772 DOI: 10.3390/nu9101156] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/06/2017] [Accepted: 10/09/2017] [Indexed: 02/07/2023] Open
Abstract
There is a growing body of evidence documenting probiotic bacteria to have a beneficial effect to the host through their ability to modulate the mucosal immune system. Many probiotic bacteria can be considered to act as either immune activators or immune suppressors, which have appreciable influence on homeostasis, inflammatory- and suppressive-immunopathology. What is becoming apparent is the ability of these probiotics to modulate innate immune responses via direct or indirect effects on the signaling pathways that drive these activatory or suppressive/tolerogenic mechanisms. This review will focus on the immunomodulatory role of probiotics on signaling pathways in innate immune cells: from positive to negative regulation associated with innate immune cells driving gut mucosal functionality. Research investigations have shown probiotics to modulate innate functionality in many ways including, receptor antagonism, receptor expression, binding to and expression of adaptor proteins, expression of negative regulatory signal molecules, induction of micro-RNAs, endotoxin tolerisation and finally, the secretion of immunomodulatory proteins, lipids and metabolites. The detailed understanding of the immunomodulatory signaling effects of probiotic strains will facilitate strain-specific selective manipulation of innate cell signal mechanisms in the modulation of mucosal adjuvanticity, immune deviation and tolerisation in both healthy subjects and patients with inflammatory and suppressive pathology.
Collapse
Affiliation(s)
- Amy Llewellyn
- School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine & Dentistry, Drake Circus, Plymouth PL4 8AA, UK.
- Menzies School of Health Research, John Mathews Building (Building 58), Royal Darwin Hospital Campus, PO Box 41096, Casuarina NT0811, Australia.
| | - Andrew Foey
- School of Biomedical & Healthcare Sciences, Plymouth University Peninsula Schools of Medicine & Dentistry, Drake Circus, Plymouth PL4 8AA, UK.
| |
Collapse
|
26
|
Taibi A, Singh N, Chen J, Arioli S, Guglielmetti S, Comelli EM. Time- and strain-specific downregulation of intestinal EPAS1
via miR-148a by Bifidobacterium bifidum. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201600596] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 10/07/2016] [Accepted: 11/18/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Amel Taibi
- Department of Nutritional Sciences; University of Toronto; ON Canada
| | - Natasha Singh
- Department of Nutritional Sciences; University of Toronto; ON Canada
| | - Jianmin Chen
- Department of Nutritional Sciences; University of Toronto; ON Canada
| | - Stefania Arioli
- Department of Nutritional Sciences; University of Toronto; ON Canada
- Department of Food; Environmental and Nutritional Sciences (DeFENS); University of Milan; Milan Italy
| | - Simone Guglielmetti
- Department of Food; Environmental and Nutritional Sciences (DeFENS); University of Milan; Milan Italy
| | - Elena M. Comelli
- Department of Nutritional Sciences; University of Toronto; ON Canada
- Centre for Child Nutrition and Health; Faculty of Medicine; University of Toronto; ON Canada
| |
Collapse
|