1
|
Yasavoli‐Sharahi H, Shahbazi R, Alsadi N, Robichaud S, Kambli D, Izadpanah A, Mohsenifar Z, Matar C. Edodes Cultured Extract Regulates Immune Stress During Puberty and Modulates MicroRNAs Involved in Mammary Gland Development and Breast Cancer Suppression. Cancer Med 2024; 13:e70277. [PMID: 39382253 PMCID: PMC11462599 DOI: 10.1002/cam4.70277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/09/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Immune stressors, such as lipopolysaccharides (LPS), profoundly affect microbiota balance, leading to gut dysbiosis. This imbalance disrupts the metabolic phenotype and structural integrity of the gut, increasing intestinal permeability. During puberty, a critical surge in estrogen levels is crucial for mammary gland development. However, inflammation originating from the gut in this period may interfere with this development, potentially heightening breast cancer risk later. The long-term effects of pubertal inflammation on mammary development and breast cancer risk are underexplored. Such episodes can dysregulate cytokine levels and microRNA expression, altering mammary cell gene expression, and predisposing them to tumorigenesis. METHODS This study hypothesizes that prebiotics, specifically Lentinula edodes Cultured Extract (AHCC), can counteract LPS's adverse effects. Using BALB/c mice, an acute LPS dose was administered at puberty, and breast cancer predisposition was assessed at 13 weeks. Cytokine and tumor-related microRNA levels, tumor development, and cancer stem cells were explored through immunoassays and qRT-PCR. RESULTS Results show that LPS induces lasting effects on cytokine and microRNA expression in mammary glands and tumors. AHCC modulates cytokine expression, including IL-1β, IL-17A/F, and IL-23, and mitigates LPS-induced IL-6 in mammary glands. It also regulates microRNA expression linked to tumor progression and suppression, particularly counteracting the upregulation of oncogenic miR-21, miR-92, and miR-155. Although AHCC slightly alters some tumor-suppressive microRNAs, these changes are modest, highlighting a complex regulatory role that warrants further study. CONCLUSION These findings underscore the potential of dietary interventions like AHCC to mitigate pubertal LPS-induced inflammation on mammary gland development and tumor formation, suggesting a preventive strategy against breast cancer.
Collapse
Affiliation(s)
- Hamed Yasavoli‐Sharahi
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Roghayeh Shahbazi
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Nawal Alsadi
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Samuel Robichaud
- Department of PathologyUniversity of MontrealMontrealQuebecCanada
| | - Darshan Babu Kambli
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
| | - Amirhossein Izadpanah
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Zhaleh Mohsenifar
- Department of PathologySchool of Medicine, Shahid Beheshti University of Medical SciencesTehranIran
| | - Chantal Matar
- Cellular and Molecular Medicine Department, Faculty of MedicineUniversity of OttawaOttawaOntarioCanada
- School of Nutrition Sciences, Faculty of Health SciencesUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
2
|
Gaspari S, Akkermans S, Akritidou T, Whelan R, Devine F, Van Impe JFM. Interference of gastrointestinal barriers with antibiotic susceptibility of foodborne pathogens: an in vitro case study of ciprofloxacin and tetracycline against Salmonella enterica and Listeria monocytogenes. Food Res Int 2024; 188:114491. [PMID: 38823842 DOI: 10.1016/j.foodres.2024.114491] [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: 02/29/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
Minimum inhibitory concentrations (MIC) assays are often questioned for their representativeness. Especially when foodborne pathogens are tested, it is of crucial importance to also consider parameters of the human digestive system. Hence, the current study aimed to assess the inhibitory capacity of two antibiotics, ciprofloxacin and tetracycline, against Salmonella enterica and Listeria monocytogenes, under representative environmental conditions. More specifically, aspects of the harsh environment of the human gastrointestinal tract (GIT) were gradually added to the experimental conditions starting from simple aerobic lab conditions into an in vitro simulation of the GIT. In this way, the effects of parameters including the anoxic environment, physicochemical conditions of the GIT (low gastric pH, digestive enzymes, bile acids) and the gut microbiota were evaluated. The latter was simulated by including a representative consortium of selected gut bacteria species. In this study, the MIC of the two antibiotics against the relevant foodborne pathogens were established, under the previously mentioned environmental conditions. The results of S. enterica highlighted the importance of the anaerobic environment when conducting such studies, since the pathogen thrived under such conditions. Inclusion of physicochemical barriers led to exactly opposite results for S. enterica and L. monocytogenes since the former became more susceptible to ciprofloxacin while the latter showed lower susceptibility towards tetracycline. Finally, the inclusion of gut bacteria had a bactericidal effect against L. monocytogenes even in the absence of antibiotics, while gut bacteria protected S. enterica from the effect of ciprofloxacin.
Collapse
Affiliation(s)
- Sotiria Gaspari
- BioTeC(+), Chemical and Biochemical Process Technology and Control, KU Leuven, Gent, Belgium
| | - Simen Akkermans
- BioTeC(+), Chemical and Biochemical Process Technology and Control, KU Leuven, Gent, Belgium
| | - Theodora Akritidou
- BioTeC(+), Chemical and Biochemical Process Technology and Control, KU Leuven, Gent, Belgium
| | - Rory Whelan
- BioTeC(+), Chemical and Biochemical Process Technology and Control, KU Leuven, Gent, Belgium; School of Biological, Health and Sport Sciences, Technological University Dublin, Ireland
| | - Faye Devine
- BioTeC(+), Chemical and Biochemical Process Technology and Control, KU Leuven, Gent, Belgium; School of Biological, Health and Sport Sciences, Technological University Dublin, Ireland
| | - Jan F M Van Impe
- BioTeC(+), Chemical and Biochemical Process Technology and Control, KU Leuven, Gent, Belgium.
| |
Collapse
|
3
|
Cossart P. Raising a Bacterium to the Rank of a Model System: The Listeria Paradigm. Annu Rev Microbiol 2023; 77:1-22. [PMID: 37713460 DOI: 10.1146/annurev-micro-110422-112841] [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] [Indexed: 09/17/2023]
Abstract
My scientific career has resulted from key decisions and reorientations, sometimes taken rapidly but not always, guided by discussions or collaborations with amazing individuals from whom I learnt a lot scientifically and humanly. I had never anticipated that I would accomplish so much in what appeared as terra incognita when I started to interrogate the mechanisms underlying the virulence of the bacterium Listeria monocytogenes. All this has been possible thanks to a number of talented team members who ultimately became friends.
Collapse
Affiliation(s)
- Pascale Cossart
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France;
| |
Collapse
|
4
|
Allahverdy J, Rashidi N. MicroRNAs induced by Listeria monocytogenes and their role in cells. Microb Pathog 2023; 175:105997. [PMID: 36669673 DOI: 10.1016/j.micpath.2023.105997] [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: 07/26/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 01/19/2023]
Abstract
Listeria monocytogenes (Lm) causes abortions at high rates and threatens newborns' lives. Also, the elderly and immunocompromised individuals are particularly vulnerable neurologically. The bacterium exerts its pathogenesis intracellularly by manipulating cell organs. It manipulates nucleus elements, microRNAs (miRNAs), in order to increase survival and evade immunity. miRNAs are small non-coding RNAs that degrade gene expression post-transcriptionally. Any alteration to the expression of miRNAs affects various cascades in cells, especially immunity-related responses. Thus, utilizing miRNAs as a novel therapeutic agent not only restricts infection but enhances immunity reactions. This review provides an overview of miRNAs in listeriosis, their role in cells, and their prospects as therapy.
Collapse
Affiliation(s)
- Javad Allahverdy
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Niloufar Rashidi
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Duodu P, Sosa G, Canar J, Chhugani O, Gamero AM. Exposing the Two Contrasting Faces of STAT2 in Inflammation. J Interferon Cytokine Res 2022; 42:467-481. [PMID: 35877097 PMCID: PMC9527059 DOI: 10.1089/jir.2022.0117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 06/27/2022] [Indexed: 11/12/2022] Open
Abstract
Inflammation is a natural immune defense mechanism of the body's response to injury, infection, and other damaging triggers. Uncontrolled inflammation may become chronic and contribute to a range of chronic inflammatory diseases. Signal transducer and activator of transcription 2 (STAT2) is an essential transcription factor exclusive to type I and type III interferon (IFN) signaling pathways. Both pathways are involved in multiple biological processes, including powering the immune system as a means of controlling infection that must be tightly regulated to offset the development of persistent inflammation. While studies depict STAT2 as protective in promoting host defense, new evidence is accumulating that exposes the deleterious side of STAT2 when inappropriately regulated, thus prompting its reevaluation as a signaling molecule with detrimental effects in human disease. This review aims to provide a comprehensive summary of the findings based on literature regarding the inflammatory behavior of STAT2 in microbial infections, cancer, autoimmune, and inflammatory diseases. In conveying the extent of our knowledge of STAT2 as a proinflammatory mediator, the aim of this review is to stimulate further investigations into the role of STAT2 in diseases characterized by deregulated inflammation and the mechanisms responsible for triggering severe responses.
Collapse
Affiliation(s)
- Philip Duodu
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Geohaira Sosa
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Jorge Canar
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Olivia Chhugani
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Ana M. Gamero
- Department of Medical Genetics and Molecular Biochemistry, Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Xing J, Liao Y, Zhang H, Zhang W, Zhang Z, Zhang J, Wang D, Tang D. Impacts of MicroRNAs Induced by the Gut Microbiome on Regulating the Development of Colorectal Cancer. Front Cell Infect Microbiol 2022; 12:804689. [PMID: 35493741 PMCID: PMC9047021 DOI: 10.3389/fcimb.2022.804689] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 03/18/2022] [Indexed: 02/05/2023] Open
Abstract
Although a dysfunctional gut microbiome is strongly linked to colorectal cancer (CRC), our knowledge of the mediators between CRC and the microbiome is limited. MicroRNAs (miRNAs) affect critical cellular processes, such as apoptosis, proliferation, and differentiation, and contribute to the regulation of CRC progression. Increasingly, studies found that miRNAs can significantly mediate bidirectional interactions between the host and the microbiome. Notably, miRNA expression is regulated by the gut microbiome, which subsequently affects the host transcriptome, thereby influencing the development of CRC. This study typically focuses on the specific functions of the microbiome in CRC and their effect on CRC-related miRNA production and reviews the role of several bacteria on miRNA, including Fusobacterium nucleatum, Escherichia coli, enterotoxigenic Bacteroides fragilis, and Faecalibacterium prausnitzii. Based on the important roles of miRNAs and the gut microbiome in CRC, strategies for modulating miRNA expression and regulating the gut microbiome composition need to be applied, such as bioactive dietary components and fecal microorganism transplantation.
Collapse
Affiliation(s)
- Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Yiqun Liao
- Department of Clinical Medical College, Dalian Medical University, Dalian, China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Zhilin Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Yangzhou University, Yangzhou, China
- *Correspondence: Dong Tang,
| |
Collapse
|
7
|
Casado-Bedmar M, Viennois E. MicroRNA and Gut Microbiota: Tiny but Mighty-Novel Insights into Their Cross-talk in Inflammatory Bowel Disease Pathogenesis and Therapeutics. J Crohns Colitis 2021; 16:992-1005. [PMID: 34918052 PMCID: PMC9282881 DOI: 10.1093/ecco-jcc/jjab223] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022]
Abstract
MicroRNAs [miRNAs], small non-coding RNAs, have recently been described as crucial contributors to intestinal homeostasis. They can interact with the gut microbiota in a reciprocal manner and deeply affect host health status, leading to several disorders when unbalanced. Inflammatory bowel disease [IBD] is a chronic inflammation of the gastrointestinal tract that co-occurs with alterations of the gut microbiota, and whose aetiology remains largely unclear. On one hand, host miRNA could be playing a relevant role in IBD pathophysiology by shaping the gut microbiota. The gut microbiome, on the other hand, may regulate the expression of host miRNAs, resulting in intestinal epithelial dysfunction, altered autophagy, and immune hyperactivation. Interestingly, it has been hypothesised that their reciprocal impact may be used for therapeutic goals. This review describes the latest research and suggests mechanisms through which miRNA and intestinal microbiota, as joint actors, may participate specifically in IBD pathophysiology. Furthermore, we discuss the diagnostic power and therapeutic potential resulting from their bidirectional communication after faecal transplantation, probiotics intake, or anti-miRNAs or miRNA mimics administration. The current literature is summarised in the present work in a comprehensive manner, hoping to provide a better understanding of the miRNA-microbiota cross-talk and to facilitate their application in IBD.
Collapse
Affiliation(s)
- Maite Casado-Bedmar
- INSERM, U1149, Center for Research on Inflammation, Université de Paris, Paris, France
| | - Emilie Viennois
- Corresponding author: Emilie Viennois, INSERM, U1149, Center for Research on Inflammation, Université de Paris, 75018 Paris, France.
| |
Collapse
|
8
|
Williams MR, Stedtfeld RD, Stedtfeld TM, Crawford RB, Kuwahara T, Kaminski NE, Tiedje JM, Hashsham SA. MicroRNA-based host response to toxicant exposure is influenced by the presence of gut microbial populations. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 797:149130. [PMID: 34311349 PMCID: PMC8464502 DOI: 10.1016/j.scitotenv.2021.149130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/27/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Segmented filamentous bacteria (SFB) and Bacteroides fragilis are known to interact with the host immune response through the aryl hydrocarbon receptor (Ahr). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD), an environmental toxicant and a high-affinity Ahr ligand has the potential to modify the effect of SFB and B. fragilis. MicroRNAs (miRNA) with their role in regulating gene expression post-transcriptionally, may potentially be used to observe such interactions between SFB, B. fragilis, and TCDD. However, little is known regarding the impact of gut microbial members on miRNA expression or its modulation in the presence of an environmental toxicant. This information is important in understanding toxicant-mediated dysbiosis in gut microbiome and the resulting human health impacts. In this study, C57BL/6 germ-free (GF) mice were colonized with SFB and B. fragilis and administered 30 μg/kg TCDD every 4 d for 28 d and miRNA were measured. Compared to GF mice, colonization with SFB resulted in an increase in up- and down-regulated Ileal miRNAs. TCDD treatment of this group decreased the number of upregulated miRNA and increased the number of down-regulated miRNAs. Association with SFB and B. fragilis together had a similar but less pronounced effect in response to TCDD treatment. TCDD treatment of GF mice had no miRNA expression response. Immune and inflammatory responses and T-cell differentiation were the key functions impacted by these miRNAs. Overall, these results reveal that the host response to toxicants may also depend on the presence of specific gut microbial populations.
Collapse
Affiliation(s)
- Maggie R Williams
- School of Engineering & Technology, Institute for Great Lakes Research, Central Michigan University, Mt Pleasant, MI, USA
| | | | | | - Robert B Crawford
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Tomomi Kuwahara
- Department of Microbiology, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Norbert E Kaminski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA; Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI 48824, USA
| | - James M Tiedje
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA; Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI 48824, USA
| | - Syed A Hashsham
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA; Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI 48824, USA; Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA.
| |
Collapse
|
9
|
Abstract
The vaginal microbiota plays an essential role in vaginal health. The vaginas of many reproductive-age women are dominated by one of the Lactobacillus species. However, the vaginas of a large number of women are characterized by the colonization of several other anaerobes. Notably, some women with the non-Lactobacillus-dominated vaginal microbiota develop bacterial vaginosis, which has been correlated with sexually transmitted infections and other adverse outcomes. However, interactions and mechanisms linking the vaginal microbiota to host response are still under investigation. There are studies suggesting a link between human microRNAs and gut microbiota, but limited analysis has been carried out on the interplay of microRNAs and vaginal microbiota. In this study, we performed a microRNA expression array profiling on 67 vaginal samples from young Swedish women. MicroRNAs were clustered into distinct groups according to vaginal microbiota composition. Interestingly, 182 microRNAs were significantly elevated in their expression in the non-Lactobacillus-dominated community, suggesting an antagonistic relationship between Lactobacillus and microRNAs. Of the elevated microRNAs, 10 microRNAs displayed excellent diagnostic potential, visualized by receiver operating characteristics analysis. We further validated our findings in 34 independent samples where expression of top microRNA candidates strongly separated the Lactobacillus-dominated community from the non-Lactobacillus-dominated community in the vaginal microbiota. Notably, the Lactobacillus crispatus-dominated community showed the most profound differential microRNA expression compared with the non-Lactobacillus-dominated community. In conclusion, we demonstrate a strong relationship between the vaginal microbiota and numerous genital microRNAs, which may facilitate a deeper mechanistic interplay in this biological niche. IMPORTANCE Vaginal microbiota is correlated with women’s health, where a non-Lactobacillus-dominated community predisposes women to a higher risk of disease, including human papillomavirus (HPV). However, the molecular relationship between the vaginal microbiota and host is largely unexplored. In this study, we investigated a link between the vaginal microbiota and host microRNAs in a group of young women. We uncovered an inverse correlation of the expression of microRNAs with the abundance of Lactobacillus species in the vaginal microbiota. Particularly, the expression of microRNA miR-23a-3p and miR-130a-3p, displaying significantly elevated levels in non-Lactobacillus-dominated communities, predicted the bacterial composition of the vaginal microbiota in an independent validation group. Since targeting of microRNAs is explored in the clinical setting, our results warrant investigation of whether microRNA modulation could be used for treating vaginosis recurrence and vaginosis-related diseases. Conversely, commensal bacteria could be used for treating diseases with microRNA aberrations.
Collapse
|
10
|
Cho K, Spasova D, Hong SW, O E, Surh CD, Im SH, Kim KS. Listeria monocytogenes Establishes Commensalism in Germ-Free Mice Through the Reversible Downregulation of Virulence Gene Expression. Front Immunol 2021; 12:666088. [PMID: 34012449 PMCID: PMC8126713 DOI: 10.3389/fimmu.2021.666088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/19/2021] [Indexed: 11/26/2022] Open
Abstract
The intestine harbors a complex community of bacterial species collectively known as commensal microbiota. Specific species of resident bacteria, as known as pathobiont, have pathogenic potential and can induce apparent damage to the host and intestinal inflammation in a certain condition. However, the host immune factors that permit its commensalism under steady state conditions are not clearly understood. Here, we studied the gut fitness of Listeria monocytogenes by using germ-free (GF) mice orally infected with this food-borne pathogen. L. monocytogenes persistently exists in the gut of GF mice without inducing chronic immunopathology. L. monocytogenes at the late phase of infection is not capable of infiltrating through the intestinal barrier. L. monocytogenes established the commensalism through the reversible down regulation of virulence gene expression. CD8+ T cells were found to be sufficient for the commensalism of L. monocytogenes. CD8+ T cells responding to L. monocytogenes contributed to the down-regulation of virulence gene expression. Our data provide important insights into the host-microbe interaction and have implications for developing therapeutics against immune disorders induced by intestinal pathogens or pathobionts.
Collapse
Affiliation(s)
- Kyungjin Cho
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Darina Spasova
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Sung-Wook Hong
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Eunju O
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Charles D Surh
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.,Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Sin-Hyeog Im
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| |
Collapse
|
11
|
Marzano M, Fosso B, Piancone E, Defazio G, Pesole G, De Robertis M. Stem Cell Impairment at the Host-Microbiota Interface in Colorectal Cancer. Cancers (Basel) 2021; 13:996. [PMID: 33673612 PMCID: PMC7957811 DOI: 10.3390/cancers13050996] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) initiation is believed to result from the conversion of normal intestinal stem cells (ISCs) into cancer stem cells (CSCs), also known as tumor-initiating cells (TICs). Hence, CRC evolves through the multiple acquisition of well-established genetic and epigenetic alterations with an adenoma-carcinoma sequence progression. Unlike other stem cells elsewhere in the body, ISCs cohabit with the intestinal microbiota, which consists of a diverse community of microorganisms, including bacteria, fungi, and viruses. The gut microbiota communicates closely with ISCs and mounting evidence suggests that there is significant crosstalk between host and microbiota at the ISC niche level. Metagenomic analyses have demonstrated that the host-microbiota mutually beneficial symbiosis existing under physiologic conditions is lost during a state of pathological microbial imbalance due to the alteration of microbiota composition (dysbiosis) and/or the genetic susceptibility of the host. The complex interaction between CRC and microbiota is at the forefront of the current CRC research, and there is growing attention on a possible role of the gut microbiome in the pathogenesis of CRC through ISC niche impairment. Here we primarily review the most recent findings on the molecular mechanism underlying the complex interplay between gut microbiota and ISCs, revealing a possible key role of microbiota in the aberrant reprogramming of CSCs in the initiation of CRC. We also discuss recent advances in OMICS approaches and single-cell analyses to explore the relationship between gut microbiota and ISC/CSC niche biology leading to a desirable implementation of the current precision medicine approaches.
Collapse
Affiliation(s)
- Marinella Marzano
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy; (M.M.); (B.F.); (G.P.)
| | - Bruno Fosso
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy; (M.M.); (B.F.); (G.P.)
| | - Elisabetta Piancone
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| | - Giuseppe Defazio
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| | - Graziano Pesole
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy; (M.M.); (B.F.); (G.P.)
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| | - Mariangela De Robertis
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari ‘Aldo Moro’, 70126 Bari, Italy; (E.P.); (G.D.)
| |
Collapse
|
12
|
Riahi Rad Z, Riahi Rad Z, Goudarzi H, Goudarzi M, Mahmoudi M, Yasbolaghi Sharahi J, Hashemi A. MicroRNAs in the interaction between host-bacterial pathogens: A new perspective. J Cell Physiol 2021; 236:6249-6270. [PMID: 33599300 DOI: 10.1002/jcp.30333] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 12/17/2022]
Abstract
Gene expression regulation plays a critical role in host-pathogen interactions, and RNAs function is essential in this process. miRNAs are small noncoding, endogenous RNA fragments that affect stability and/or translation of mRNAs, act as major posttranscriptional regulators of gene expression. miRNA is involved in regulating many biological or pathological processes through targeting specific mRNAs, including development, differentiation, apoptosis, cell cycle, cytoskeleton organization, and autophagy. Deregulated microRNA expression is associated with many types of diseases, including cancers, immune disturbances, and infection. miRNAs are a vital section of the host immune response to bacterial-made infection. Bacterial pathogens suppress host miRNA expression for their benefit, promoting survival, replication, and persistence. The role played through miRNAs in interaction with host-bacterial pathogen has been extensively studied in the past 10 years, and knowledge about these staggering molecules' function can clarify the complicated and ambiguous interactions of the host-bacterial pathogen. Here, we review how pathogens prevent the host miRNA expression. We briefly discuss emerging themes in this field, including their role as biomarkers in identifying bacterial infections, as part of the gut microbiota, on host miRNA expression.
Collapse
Affiliation(s)
- Zohreh Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Riahi Rad
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahmoudi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Javad Yasbolaghi Sharahi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Morris NL, Choudhry MA. Maintenance of gut barrier integrity after injury: Trust your gut microRNAs. J Leukoc Biol 2021; 110:979-986. [PMID: 33577717 DOI: 10.1002/jlb.3ru0120-090rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal (GI) tract is a highly dynamic structure essential for digestion, nutrient absorption, and providing an interface to prevent gut bacterial translocation. In order to maintain the barrier function, the gut utilizes many defense mechanisms including proliferation, apoptosis, and apical junctional complexes. Disruption of any of these parameters due to injury or disease could negatively impact the intestinal barrier function and homeostasis resulting in increased intestine inflammation, permeability, bacterial dysbiosis, and tissue damage. MicroRNAs are small noncoding RNA sequences that are master regulators of normal cellular homeostasis. These regulatory molecules affect cellular signaling pathways and potentially serve as candidates for providing a mechanism of impaired gut barrier integrity following GI-related pathologic conditions, ethanol exposure, or trauma such as burn injury. MicroRNAs influence cellular apoptosis, proliferation, apical junction complex expression, inflammation, and the microbiome. Due to their widespread functional affiliations, altered expression of microRNAs are associated with many pathologic conditions. This review explores the role of microRNAs in regulation of intestinal barrier integrity. The studies reviewed demonstrate that microRNAs largely impact intestine barrier function and provide insight behind the observed adverse effects following ethanol and burn injury. Furthermore, these studies suggest that microRNAs are excellent candidates for therapeutic intervention or for biomarkers to manage gut barrier integrity following trauma such as burn injury and other GI-related pathologic conditions.
Collapse
Affiliation(s)
- Niya L Morris
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Current address: Department of Medicine: Pulmonary, Allergy, Critical Care and Sleep, Emory University/Atlanta VA Medical Center, Decatur, Geogia, USA
| | - Mashkoor A Choudhry
- Alcohol Research Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Burn & Shock Trauma Research Institute, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Integrative Cell Biology Program, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA.,Department of Surgery, Loyola University Chicago Health Sciences Division, Maywood, Illinois, USA
| |
Collapse
|
14
|
Li M, Chen WD, Wang YD. The roles of the gut microbiota-miRNA interaction in the host pathophysiology. Mol Med 2020; 26:101. [PMID: 33160314 PMCID: PMC7648389 DOI: 10.1186/s10020-020-00234-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/26/2020] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota regulates the biological processes of organisms acting like ‘another’ genome, affecting the health and disease of the host. MicroRNAs, as important physiological regulators, have been found to be involved in health and disease. Recently, the gut microbiota has been reported to affect host health by regulating host miRNAs. For example, Fusobacterium nucleatum could aggravate chemoresistance of colorectal cancer by decreasing the expression of miR-18a* and miR-4802. What’s more, miRNAs can shape the gut microbiota composition, ultimately affecting the host's physiology and disease. miR-515-5p and miR-1226-5p could promote the growth of Fusobacterium nucleatum (Fn) and Escherichia coli (E.coli), which have been reported to drive colorectal cancer. Here, we will review current findings of the interactions between the gut microbiota and microRNAs and discuss how the gut microbiota–microRNA interactions affect host pathophysiology including intestinal, neurological, cardiovascular, and immune health and diseases.
Collapse
Affiliation(s)
- Meihong Li
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People's Republic of China
| | - Wei-Dong Chen
- Key Laboratory of Molecular Pathology, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, People's Republic of China. .,Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, The People's Hospital of Hebi, School of Medicine, Henan University, Henan, People's Republic of China.
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People's Republic of China.
| |
Collapse
|
15
|
Bi K, Zhang X, Chen W, Diao H. MicroRNAs Regulate Intestinal Immunity and Gut Microbiota for Gastrointestinal Health: A Comprehensive Review. Genes (Basel) 2020; 11:genes11091075. [PMID: 32932716 PMCID: PMC7564790 DOI: 10.3390/genes11091075] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs are small non-coding RNAs regulating gene expression at the post-transcriptional level. The regulation of microRNA expression in the gut intestine is gradually recognized as one of the crucial contributors of intestinal homeostasis and overall health. Recent studies indicated that both the microRNAs endogenous in the gut intestine and exogenous from diets could play influential roles in modulating microbial colonization and intestinal immunity. In this review, we discuss the biological functions of microRNAs in regulating intestinal homeostasis by modulating intestinal immune responses and gut microbiota. We particularly focus on addressing the microRNA-dependent communication and interactions among microRNA, gut microbiota, and intestinal immune system. Besides, we also summarize the roles of diet-derived microRNAs in host-microbiome homeostasis and their benefits on intestinal health. A better understanding of the relationships among intestinal disorders, microRNAs, and other factors influencing intestinal health can facilitate the application of microRNA-based therapeutics for gastrointestinal diseases.
Collapse
|
16
|
Sarshar M, Scribano D, Ambrosi C, Palamara AT, Masotti A. Fecal microRNAs as Innovative Biomarkers of Intestinal Diseases and Effective Players in Host-Microbiome Interactions. Cancers (Basel) 2020; 12:E2174. [PMID: 32764361 PMCID: PMC7463924 DOI: 10.3390/cancers12082174] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/30/2020] [Accepted: 08/02/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, short non-coding microRNAs (miRNAs), including circulating and fecal miRNAs have emerged as important modulators of various cellular processes by regulating the expression of target genes. Recent studies revealed the role of miRNAs as powerful biomarkers in disease diagnosis and for the development of innovative therapeutic applications in several human conditions, including intestinal diseases. In this review, we explored the literature and summarized the role of identified dysregulated fecal miRNAs in intestinal diseases, with particular focus on colorectal cancer (CRC) and celiac disease (CD). The aim of this review is to highlight one fascinating aspect of fecal miRNA function related to gut microbiota shaping and bacterial metabolism influencing. The role of miRNAs as "messenger" molecules for inter kingdom communications will be analyzed to highlight their role in the complex host-bacteria interactions. Moreover, whether fecal miRNAs could open up new perspectives to develop novel suitable biomarkers for disease detection and innovative therapeutic approaches to restore microbiota balance will be discussed.
Collapse
Affiliation(s)
- Meysam Sarshar
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur Italia-Cenci Bolognetti Foundation, 00185 Rome, Italy;
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
- Microbiology Research Center (MRC), Pasteur Institute of Iran, 1316943551 Tehran, Iran
| | - Daniela Scribano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy;
- Dani Di Giò Foundation-Onlus, 00193 Rome, Italy
| | - Cecilia Ambrosi
- IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur Italia-Cenci Bolognetti Foundation, 00185 Rome, Italy;
- IRCCS San Raffaele Pisana, Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, 00146 Rome, Italy;
| |
Collapse
|
17
|
Denzer L, Schroten H, Schwerk C. From Gene to Protein-How Bacterial Virulence Factors Manipulate Host Gene Expression During Infection. Int J Mol Sci 2020; 21:ijms21103730. [PMID: 32466312 PMCID: PMC7279228 DOI: 10.3390/ijms21103730] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023] Open
Abstract
Bacteria evolved many strategies to survive and persist within host cells. Secretion of bacterial effectors enables bacteria not only to enter the host cell but also to manipulate host gene expression to circumvent clearance by the host immune response. Some effectors were also shown to evade the nucleus to manipulate epigenetic processes as well as transcription and mRNA procession and are therefore classified as nucleomodulins. Others were shown to interfere downstream with gene expression at the level of mRNA stability, favoring either mRNA stabilization or mRNA degradation, translation or protein stability, including mechanisms of protein activation and degradation. Finally, manipulation of innate immune signaling and nutrient supply creates a replicative niche that enables bacterial intracellular persistence and survival. In this review, we want to highlight the divergent strategies applied by intracellular bacteria to evade host immune responses through subversion of host gene expression via bacterial effectors. Since these virulence proteins mimic host cell enzymes or own novel enzymatic functions, characterizing their properties could help to understand the complex interactions between host and pathogen during infections. Additionally, these insights could propose potential targets for medical therapy.
Collapse
|
18
|
Rolhion N, Chassaing B, Nahori MA, de Bodt J, Moura A, Lecuit M, Dussurget O, Bérard M, Marzorati M, Fehlner-Peach H, Littman DR, Gewirtz AT, Van de Wiele T, Cossart P. A Listeria monocytogenes Bacteriocin Can Target the Commensal Prevotella copri and Modulate Intestinal Infection. Cell Host Microbe 2020; 26:691-701.e5. [PMID: 31726031 PMCID: PMC6854461 DOI: 10.1016/j.chom.2019.10.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 09/05/2019] [Accepted: 10/23/2019] [Indexed: 12/13/2022]
Abstract
Understanding the role of the microbiota components in either preventing or favoring enteric infections is critical. Here, we report the discovery of a Listeria bacteriocin, Lmo2776, which limits Listeria intestinal colonization. Oral infection of conventional mice with a Δlmo2776 mutant leads to a thinner intestinal mucus layer and higher Listeria loads both in the intestinal content and deeper tissues compared to WT Listeria. This latter difference is microbiota dependent, as it is not observed in germ-free mice. Strikingly, it is phenocopied by pre-colonization of germ-free mice before Listeria infection with Prevotella copri, an abundant gut-commensal bacteria, but not with the other commensals tested. We further show that Lmo2776 targets P. copri and reduces its abundance. Together, these data unveil a role for P.copri in exacerbating intestinal infection, highlighting that pathogens such as Listeria may selectively deplete microbiota bacterial species to avoid excessive inflammation. L. monocytogenes secretes a bacteriocin (Lmo2776) homologous to the lactococcin 972 Lmo2776 controls Listeria intestinal colonization in a microbiota-dependent manner Lmo2776 targets the abundant gut commensal Prevotella copri Presence of P. copri exacerbates infection
Collapse
Affiliation(s)
- Nathalie Rolhion
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France
| | - Benoit Chassaing
- Neurosciences Institute, Georgia State University (GSU), Atlanta, GA 30303, USA; Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, GSU, Atlanta, GA 30303, USA
| | - Marie-Anne Nahori
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France
| | - Jana de Bodt
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Alexandra Moura
- Institut Pasteur, Unité Biologie des Infections, 75015 Paris, France; Inserm, U1117, 75015 Paris, France
| | - Marc Lecuit
- Institut Pasteur, Unité Biologie des Infections, 75015 Paris, France; Inserm, U1117, 75015 Paris, France; Paris Descartes University, Sorbonne Paris Cité, Division of Infectious Diseases and Tropical Medicine, Necker-Enfants Malades University Hospital, Institut Imagine, 75743 Paris, France
| | - Olivier Dussurget
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France; Université de Paris, 75013 Paris, France
| | - Marion Bérard
- Animalerie Centrale, Department of Technology and Scientific Programmes, Institut Pasteur, 75015 Paris, France
| | - Massimo Marzorati
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Hannah Fehlner-Peach
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Dan R Littman
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York, NY 10016, USA
| | - Andrew T Gewirtz
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, GSU, Atlanta, GA 30303, USA
| | - Tom Van de Wiele
- Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, 9000 Ghent, Belgium
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, Unité sous-contrat 2020, 75015 Paris, France.
| |
Collapse
|
19
|
Fouhse JM, Tsoi S, Clark B, Gartner S, Patterson JL, Foxcroft GR, Willing BP, Dyck MK. Outcomes of a low birth weight phenotype on piglet gut microbial composition and intestinal transcriptomic profile. CANADIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1139/cjas-2019-0066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Decades of selection for increased litter size has caused a proportion of sows to consistently produce low birth weight (LBW) litters resulting in economic loss for producers due to reduced piglet survivability and growth. We hypothesized that piglets from LBW litters would have altered gut microbial composition, intestinal architecture, and intestinal transcriptomic profiles compared with piglets from high birth weight (HBW) litters. Sows were designated LBW (n = 45) or HBW (n = 46) based on litter birth weights of three successive parities. LBW piglets were 22% lighter (P < 0.001) at birth; however, no longer differed (P > 0.05) in weight at weaning compared with HBW piglets. LBW piglets had reduced (P < 0.05) fecal microbial diversity with a 114% increase in fecal Enterobacteriaceae (P < 0.05), as well as reduced (P < 0.05) abundance of cecal Roseburia and Faecalibacterium, fiber-degrading butyrate producers. Several genes associated with metabolic (PER2, CES1, KLHL38, and HK2) and immune pathways (IL-1B, IRF8, and TNIP3) were differentially expressed, suggesting altered metabolic and immune function in LBW piglets. In conclusion, LBW piglets had potentially unfavorable shifts in microbial structure in comparison to HBW piglets accompanied with alterations in metabolic and immune gene expression. Results indicate some biological consequences linking LBW phenotype to changes in production efficiency later in life.
Collapse
Affiliation(s)
- Janelle M. Fouhse
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Stephen Tsoi
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Brenna Clark
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Stephanie Gartner
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Jennifer L. Patterson
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - George R. Foxcroft
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Benjamin P. Willing
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Michael K. Dyck
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| |
Collapse
|
20
|
Fu H, Elena RC, Marquez PH. The roles of small RNAs: insights from bacterial quorum sensing. ACTA ACUST UNITED AC 2019. [DOI: 10.1186/s41544-019-0027-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Abstract
Ribonucleic acids (RNAs) mainly played auxiliary roles in regulations of genetic processes while recent explorations into small non-coding RNAs (sRNAs) in bacteria have broadened the scope of RNAs studies in these processes. sRNAs have been demonstrated to be involved in various genetic processes and to regulate a variety of bacterial physiologies. Comparatively, quorum sensing (QS) is a mature bacterial cell signaling system which regulates bacteria physiologies as well. Prokaryotic sRNAs studies in the status quo have revealed an emerging picture of trans-kingdom signaling regulation and increasing investigations have demonstrated the feasibility of inter-kingdom signaling as the consequence of QS. We therefore review such phenomena and their similarities to investigate the potential of prokaryote-sourced interkingdom signaling and regulation.
Collapse
|
21
|
Rosenfeld CS. Effects of Phytoestrogens on the Developing Brain, Gut Microbiota, and Risk for Neurobehavioral Disorders. Front Nutr 2019; 6:142. [PMID: 31555657 PMCID: PMC6727358 DOI: 10.3389/fnut.2019.00142] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/14/2019] [Indexed: 01/11/2023] Open
Abstract
Many pregnant and nursing women consume high amounts of soy and other plant products that contain phytoestrogens, such as genistein (GEN) and daidzein. Infants may also be provided soy based formulas. With their ability to bind and activate estrogen receptors (ESR) in the brain, such compounds can disrupt normal brain programming and lead to later neurobehavioral disruptions. However, other studies suggest that maternal consumption of soy and soy based formulas containing such phytoestrogens might lead to beneficial behavioral effects. Select gut microbes might also convert daidzein and to a lesser extent genistein to even more potent forms, e.g., equol derivatives. Thus, infant exposure to phytoestrogens may result in contrasting effects dependent upon the gut flora. It is also becoming apparent that consumption or exposure to these xenoestrogens may lead to gut dysbiosis. Phytoestrogen-induced changes in gut bacteria might in turn affect the brain through various mechanisms. This review will consider the evidence to date in rodent and other animal models and human epidemiological data as to whether developmental exposure to phytoestrogens, in particular genistein and daidzein, adversely or beneficially impact offspring neurobehavioral programming. Consideration will be given to potential mechanisms by which such compounds might affect neurobehavioral responses. A better understanding of effects perinatal exposure to phytoestrogen can exert on brain programming will permit pregnant women and those seeking to become pregnant to make better-educated choices. If phytoestrogen-induced gut dysbiosis contributes to neurobehavioral disruptions, remediation strategies may be designed to prevent such gut microbiota alterations and thereby improve neurobehavioral outcomes.
Collapse
Affiliation(s)
- Cheryl S. Rosenfeld
- Bond Life Sciences Center, University of Missouri, Columbia, MO, United States
- MU Informatics Institute, University of Missouri, Columbia, MO, United States
- Thompson Center for Autism and Neurobehavioral Disorders, University of Missouri, Columbia, MO, United States
- Biomedical Sciences, University of Missouri, Columbia, MO, United States
- Genetics Area Program, University of Missouri, Columbia, MO, United States
| |
Collapse
|
22
|
Iacob S, Iacob DG. Infectious Threats, the Intestinal Barrier, and Its Trojan Horse: Dysbiosis. Front Microbiol 2019; 10:1676. [PMID: 31447793 PMCID: PMC6692454 DOI: 10.3389/fmicb.2019.01676] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/08/2019] [Indexed: 02/06/2023] Open
Abstract
The ecosystem of the gut microbiota consists of diverse intestinal species with multiple metabolic and immunologic activities and it is closely connected with the intestinal epithelia and mucosal immune response, with which it builds a complex barrier against intestinal pathogenic bacteria. The microbiota ensures the integrity of the gut barrier through multiple mechanisms, either by releasing antibacterial molecules (bacteriocins) and anti-inflammatory short-chain fatty acids or by activating essential cell receptors for the immune response. Experimental studies have confirmed the role of the intestinal microbiota in the epigenetic modulation of the gut barrier through posttranslational histone modifications and regulatory mechanisms induced by epithelial miRNA in the epithelial lumen. Any quantitative or functional changes of the intestinal microbiota, referred to as dysbiosis, alter the immune response, decrease epithelial permeability and destabilize intestinal homeostasis. Consequently, the overgrowth of pathobionts (Staphylococcus, Pseudomonas, and Escherichia coli) favors intestinal translocations with Gram negative bacteria or their endotoxins and could trigger sepsis, septic shock, secondary peritonitis, or various intestinal infections. Intestinal infections also induce epithelial lesions and perpetuate the risk of bacterial translocation and dysbiosis through epithelial ischemia and pro-inflammatory cytokines. Furthermore, the decline of protective anaerobic bacteria (Bifidobacterium and Lactobacillus) and inadequate release of immune modulators (such as butyrate) affects the release of antimicrobial peptides, de-represses microbial virulence factors and alters the innate immune response. As a result, intestinal germs modulate liver pathology and represent a common etiology of infections in HIV immunosuppressed patients. Antibiotic and antiretroviral treatments also promote intestinal dysbiosis, followed by the selection of resistant germs which could later become a source of infections. The current article addresses the strong correlations between the intestinal barrier and the microbiota and discusses the role of dysbiosis in destabilizing the intestinal barrier and promoting infectious diseases.
Collapse
Affiliation(s)
- Simona Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,National Institute of Infectious Diseases "Prof. Dr. Matei Balş", Bucharest, Romania
| | - Diana Gabriela Iacob
- Infectious Diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
23
|
Aguilar C, Mano M, Eulalio A. Multifaceted Roles of microRNAs in Host-Bacterial Pathogen Interaction. Microbiol Spectr 2019; 7:10.1128/microbiolspec.bai-0002-2019. [PMID: 31152522 PMCID: PMC11026079 DOI: 10.1128/microbiolspec.bai-0002-2019] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) are a well-characterized class of small noncoding RNAs that act as major posttranscriptional regulators of gene expression. Accordingly, miRNAs have been associated with a wide range of fundamental biological processes and implicated in human diseases. During the past decade, miRNAs have also been recognized for their role in the complex interplay between the host and bacterial pathogens, either as part of the host response to counteract infection or as a molecular strategy employed by bacteria to subvert host pathways for their own benefit. Importantly, the characterization of downstream miRNA targets and their underlying mechanisms of action has uncovered novel molecular factors and pathways relevant to infection. In this article, we review the current knowledge of the miRNA response to bacterial infection, focusing on different bacterial pathogens, including Salmonella enterica, Listeria monocytogenes, Mycobacterium spp., and Helicobacter pylori, among others.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-Based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
- RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
24
|
Allen J, Sears CL. Impact of the gut microbiome on the genome and epigenome of colon epithelial cells: contributions to colorectal cancer development. Genome Med 2019; 11:11. [PMID: 30803449 PMCID: PMC6388476 DOI: 10.1186/s13073-019-0621-2] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In recent years, the number of studies investigating the impact of the gut microbiome in colorectal cancer (CRC) has risen sharply. As a result, we now know that various microbes (and microbial communities) are found more frequently in the stool and mucosa of individuals with CRC than healthy controls, including in the primary tumors themselves, and even in distant metastases. We also know that these microbes induce tumors in various mouse models, but we know little about how they impact colon epithelial cells (CECs) directly, or about how these interactions might lead to modifications at the genetic and epigenetic levels that trigger and propagate tumor growth. Rates of CRC are increasing in younger individuals, and CRC remains the second most frequent cause of cancer-related deaths globally. Hence, a more in-depth understanding of the role that gut microbes play in CRC is needed. Here, we review recent advances in understanding the impact of gut microbes on the genome and epigenome of CECs, as it relates to CRC. Overall, numerous studies in the past few years have definitively shown that gut microbes exert distinct impacts on DNA damage, DNA methylation, chromatin structure and non-coding RNA expression in CECs. Some of the genes and pathways that are altered by gut microbes relate to CRC development, particularly those involved in cell proliferation and WNT signaling. We need to implement more standardized analysis strategies, collate data from multiple studies, and utilize CRC mouse models to better assess these effects, understand their functional relevance, and leverage this information to improve patient care.
Collapse
Affiliation(s)
- Jawara Allen
- Department of Medicine, Johns Hopkins University School of Medicine, Orleans Street, Baltimore, MD, 21231, USA
| | - Cynthia L Sears
- Department of Medicine, Johns Hopkins University School of Medicine, Orleans Street, Baltimore, MD, 21231, USA. .,Bloomberg-Kimmel Institute for Immunotherapy and Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Medical Institutions, North Broadway, Baltimore, MD, 21231, USA.
| |
Collapse
|
25
|
Abstract
All natural animals and plants are holobionts, consisting of the host and microbiome, which is composed of abundant and diverse microorganisms. Health and disease of holobionts depend as much on interactions between host and microbiome and within the microbiome, as on interactions between organs and body parts of the host. Recent evidence indicates that a significant fraction of the microbiome is transferred by a variety of mechanisms from parent to offspring for many generations. Genetic variation in holobionts can occur in the microbiome as well as in the host genome, and it occurs more rapidly and by more mechanisms in genomes of microbiomes than in host genomes (e.g. via acquisition of novel microbes and horizontal gene transfer of microbial genes into host chromosomes). Evidence discussed in this review supports the concept that holobionts with their hologenomes can be considered levels of selection in evolution. Though changes in the microbiome can lead to evolution of the holobiont, it can also lead to dysbiosis and diseases (e.g. obesity, diarrhea, inflammatory bowel disease, and autism). In practice, the possibility of manipulating microbiomes offers the potential to prevent and cure diseases.
Collapse
|
26
|
Drolia R, Bhunia AK. Crossing the Intestinal Barrier via Listeria Adhesion Protein and Internalin A. Trends Microbiol 2019; 27:408-425. [PMID: 30661918 DOI: 10.1016/j.tim.2018.12.007] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 11/11/2018] [Accepted: 12/14/2018] [Indexed: 12/24/2022]
Abstract
The intestinal epithelial cell lining provides the first line of defense, yet foodborne pathogens such as Listeria monocytogenes can overcome this barrier; however, the underlying mechanism is not well understood. Though the host M cells in Peyer's patch and the bacterial invasion protein internalin A (InlA) are involved, L. monocytogenes can cross the gut barrier in their absence. The interaction of Listeria adhesion protein (LAP) with the host cell receptor (heat shock protein 60) disrupts the epithelial barrier, promoting bacterial translocation. InlA aids L. monocytogenes transcytosis via interaction with the E-cadherin receptor, which is facilitated by epithelial cell extrusion and goblet cell exocytosis; however, LAP-induced cell junction opening may be an alternative bacterial strategy for InlA access to E-cadherin and its translocation. Here, we summarize the strategies that L. monocytogenes employs to circumvent the intestinal epithelial barrier and compare and contrast these strategies with other enteric bacterial pathogens. Additionally, we provide implications of recent findings for food safety regulations.
Collapse
Affiliation(s)
- Rishi Drolia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA
| | - Arun K Bhunia
- Molecular Food Microbiology Laboratory, Department of Food Science, Purdue University, West Lafayette, IN 47907, USA; Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
27
|
Aguilar C, Mano M, Eulalio A. MicroRNAs at the Host-Bacteria Interface: Host Defense or Bacterial Offense. Trends Microbiol 2018; 27:206-218. [PMID: 30477908 DOI: 10.1016/j.tim.2018.10.011] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 10/17/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
MicroRNAs are a class of small noncoding RNAs that act as major post-transcriptional regulators of gene expression. They are currently recognized for their important role in the intricate interaction between host and bacterial pathogens, either as part of the host immune response to neutralize infection, or as a molecular strategy employed by bacteria to hijack host pathways for their own benefit. Here, we summarize recent advances on the function of miRNAs during infection of mammalian hosts by bacterial pathogens, highlighting key cellular pathways. In addition, we discuss emerging themes in this field, including the participation of miRNAs in host-microbiota crosstalk and cell-to-cell communication.
Collapse
Affiliation(s)
- Carmen Aguilar
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
| | - Miguel Mano
- Functional Genomics and RNA-based Therapeutics Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Ana Eulalio
- Host RNA Metabolism Group, Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany; RNA & Infection Group, Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
28
|
miRNAs reshape immunity and inflammatory responses in bacterial infection. Signal Transduct Target Ther 2018; 3:14. [PMID: 29844933 PMCID: PMC5968033 DOI: 10.1038/s41392-018-0006-9] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/20/2017] [Accepted: 12/10/2017] [Indexed: 12/15/2022] Open
Abstract
Pathogenic bacteria cause various infections worldwide, especially in immunocompromised and other susceptible individuals, and are also associated with high infant mortality rates in developing countries. MicroRNAs (miRNAs), small non-coding RNAs with evolutionarily conserved sequences, are expressed in various tissues and cells that play key part in various physiological and pathologic processes. Increasing evidence implies roles for miRNAs in bacterial infectious diseases by modulating inflammatory responses, cell penetration, tissue remodeling, and innate and adaptive immunity. This review highlights some recent intriguing findings, ranging from the correlation between aberrant expression of miRNAs with bacterial infection progression to their profound impact on host immune responses. Harnessing of dysregulated miRNAs in bacterial infection may be an approach to improving the diagnosis, prevention and therapy of infectious diseases. Changes in production of tiny cellular RNAs in response to bacterial infection could guide the development of better diagnostics and therapies. MicroRNAs regulate other genes by binding to messenger RNA strands and controlling their translation into proteins. Xikun Zhou, Min Wu and colleagues of the University of North Dakota have now reviewed current knowledge about how microRNA levels shift during infection with various bacterial pathogens. These microRNAs can modulate the immune response as well as pathways that influence metabolic activity and cell survival. Increasing studies have indicated that shifts in microRNA levels in response to different infections could provide a potential bacterial ‘fingerprint’ for achieving accurate diagnosis. With deeper insight into how different microRNAs influence infection, it might one day day become possible to target these molecules with ‘antisense’ or ‘agonist’ drugs that modulate their activity.
Collapse
|
29
|
Citrobacter rodentium alters the mouse colonic miRNome. Genes Immun 2018; 20:207-213. [PMID: 29728609 DOI: 10.1038/s41435-018-0026-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 02/11/2018] [Accepted: 02/22/2018] [Indexed: 12/12/2022]
Abstract
Citrobacter rodentium is a murine pathogen causing transmissible colonic hyperplasia and colitis with a pathogenic mechanism similar to foodborne enterohaemorrhagic Escherichia coli in humans. Mechanisms underlying intestinal responses to C. rodentium infection are incompletely understood. We identified 24 colonic microRNAs (miRNAs) as significantly deregulated in response to C. rodentium, including miR-7a, -17, -19a, -20a, -20b, -92a, -106a, -132, -200a, and -2137; most of these miRNAs belong to the oncogenic miR-17-92 clusters. Pathways involved in cell cycle, cancers, and immune responses were enriched among the predicted targets of these miRNAs. We further demonstrated that an apoptosis facilitator, Bim, is a candidate gene target of miRNA-mediated host response to the infection. These findings suggest that host miRNAs participate in C. rodentium pathogenesis and may represent novel treatment targets.
Collapse
|
30
|
Abstract
Noncoding RNAs (ncRNAs) regulating virulence have been identified in most pathogens. This review discusses RNA-mediated mechanisms exploited by bacterial pathogens to successfully infect and colonize their hosts. It discusses the most representative RNA-mediated regulatory mechanisms employed by two intracellular [Listeria monocytogenes and Salmonella enterica serovar Typhimurium (S. Typhimurium)] and two extracellular (Vibrio cholerae and Staphylococcus aureus) bacterial pathogens. We review the RNA-mediated regulators (e.g., thermosensors, riboswitches, cis- and trans-encoded RNAs) used for adaptation to the specific niches colonized by these bacteria (intestine, blood, or the intracellular environment, for example) in the framework of the specific pathophysiological aspects of the diseases caused by these microorganisms. A critical discussion of the newest findings in the field of bacterial ncRNAs shows how examples in model pathogens could pave the way for the discovery of new mechanisms in other medically important bacterial pathogens.
Collapse
Affiliation(s)
- Juan J Quereda
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris F-75015, France; , .,Institut National de la Santé et de la Recherche Médicale, U604, Paris F-75015, France.,Institut National de la Recherche Agronomique, USC2020, Paris F-75015, France
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Paris F-75015, France; , .,Institut National de la Santé et de la Recherche Médicale, U604, Paris F-75015, France.,Institut National de la Recherche Agronomique, USC2020, Paris F-75015, France
| |
Collapse
|
31
|
Rosenberg E, Zilber-Rosenberg I. The hologenome concept of evolution after 10 years. MICROBIOME 2018; 6:78. [PMID: 29695294 PMCID: PMC5922317 DOI: 10.1186/s40168-018-0457-9] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 04/05/2018] [Indexed: 05/11/2023]
Abstract
The holobiont (host with its endocellular and extracellular microbiome) can function as a distinct biological entity, an additional organismal level to the ones previously considered, on which natural selection operates. The holobiont can function as a whole: anatomically, metabolically, immunologically, developmentally, and during evolution. Consideration of the holobiont with its hologenome as an independent level of selection in evolution has led to a better understanding of underappreciated modes of genetic variation and evolution. The hologenome is comprised of two complimentary parts: host and microbiome genomes. Changes in either genome can result in variations that can be selected for or against. The host genome is highly conserved, and genetic changes within it occur slowly, whereas the microbiome genome is dynamic and can change rapidly in response to the environment by increasing or reducing particular microbes, by acquisition of novel microbes, by horizontal gene transfer, and by mutation. Recent experiments showing that microbiota can play an initial role in speciation have been suggested as an additional mode of enhancing evolution. Some of the genetic variations can be transferred to offspring by a variety of mechanisms. Strain-specific DNA analysis has shown that at least some of the microbiota can be maintained across hundreds of thousands of host generations, implying the existence of a microbial core. We argue that rapid changes in the microbiome genome could allow holobionts to adapt and survive under changing environmental conditions thus providing the time necessary for the host genome to adapt and evolve. As Darwin wrote, "It is not the strongest of the species that survives but the most adaptable".
Collapse
Affiliation(s)
- Eugene Rosenberg
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Ramat Aviv, Israel
| | - Ilana Zilber-Rosenberg
- Department of Molecular Microbiology and Biotechnology, Tel Aviv University, Ramat Aviv, Israel
| |
Collapse
|
32
|
Impact of dietary compounds on cancer-related gut microbiota and microRNA. Appl Microbiol Biotechnol 2018; 102:4291-4303. [PMID: 29589094 DOI: 10.1007/s00253-018-8935-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/12/2018] [Accepted: 03/13/2018] [Indexed: 02/07/2023]
Abstract
Cancer is one of the most common causes of death worldwide. Extensive research has been conducted on cancer; regardless, the link between cancer and diet remains undetermined. Recent studies have emphasized the importance of miRNAs in cancer-associated pathways from the perspective of dietary modulation. We highlighted the recent data on dietary modulation of gut microbiota and miRNAs related to cancer on the basis of recently published results. The targets of miRNAs are oncogenes or tumor suppressors that mediate the progression and initiation of carcinogenesis. Different miRNAs display complex expression profiles in response to dietary manipulation. Various dietary components, such as fatty acids, resveratrol, isothiocyanate, and curcumin, have been effectively used in cancer prevention and treatment. This potency is attributed to the capability of these components to alter miRNA expression, thereby modulating the vital pathways involved in metastasis, invasion, apoptosis, tumor growth, and cell proliferation.
Collapse
|
33
|
MicroRNA-146a Deficiency Protects against Listeria monocytogenes Infection by Modulating the Gut Microbiota. Int J Mol Sci 2018; 19:ijms19040993. [PMID: 29587465 PMCID: PMC5979314 DOI: 10.3390/ijms19040993] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/15/2018] [Accepted: 03/24/2018] [Indexed: 12/21/2022] Open
Abstract
The gut microbiota and microRNAs play important roles in the defense against infection. However, the role of miR-146a in L. monocytogenes infection and gut microbiota remains unclear. We tried to determine whether miR-146a controlled L. monocytogenes infection by regulating the gut microbiota. Wild-type and miR-146a-deficient mice or macrophages were used to characterize the impact of miR-146a on animal survival, cell death, bacterial clearance, and gut microbiota following L. monocytogenes challenge. We found that L. monocytogenes infection induced miR-146a expression both in vitro and in vivo. When compared to wild-type mice, miR-146a-deficient mice were more resistant to L. monocytogenes infection. MiR-146a deficiency in macrophages resulted in reduced invasion and intracellular survival of L. monocytogenes. High-throughput sequencing of 16S rRNA revealed that the gut microbiota composition differed between miR-146a-deficient and wild-type mice. Relative to wild-type mice, miR-146a-deficient mice had decreased levels of the Proteobacteria phylum, Prevotellaceae family, and Parasutterella genus, and significantly increased short-chain fatty acid producing bacteria, including the genera Alistipes, Blautia, Coprococcus_1, and Ruminococcus_1. Wild-type mice co-housed with miR-146a-deficient mice had increased resistance to L. monocytogenes, indicating that miR-146a deficiency guides the gut microbiota to alleviate infection. Together, these results suggest that miR-146a deficiency protects against L. monocytogenes infection by regulating the gut microbiota.
Collapse
|
34
|
Radoshevich L, Cossart P. Listeria monocytogenes: towards a complete picture of its physiology and pathogenesis. Nat Rev Microbiol 2018; 16:32-46. [PMID: 29176582 DOI: 10.1038/nrmicro.2017.126] [Citation(s) in RCA: 469] [Impact Index Per Article: 78.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Listeria monocytogenes is a food-borne pathogen responsible for a disease called listeriosis, which is potentially lethal in immunocompromised individuals. This bacterium, first used as a model to study cell-mediated immunity, has emerged over the past 20 years as a paradigm in infection biology, cell biology and fundamental microbiology. In this Review, we highlight recent advances in the understanding of human listeriosis and L. monocytogenes biology. We describe unsuspected modes of hijacking host cell biology, ranging from changes in organelle morphology to direct effects on host transcription via a new class of bacterial effectors called nucleomodulins. We then discuss advances in understanding infection in vivo, including the discovery of tissue-specific virulence factors and the 'arms race' among bacteria competing for a niche in the microbiota. Finally, we describe the complexity of bacterial regulation and physiology, incorporating new insights into the mechanisms of action of a series of riboregulators that are critical for efficient metabolic regulation, antibiotic resistance and interspecies competition.
Collapse
Affiliation(s)
- Lilliana Radoshevich
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, F-75015 Paris, France
- Inserm, U604, F-75015 Paris, France
- French National Institute for Agricultural Research (INRA), Unité sous-contrat 2020, F-75015 Paris, France
- Department of Microbiology and Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, Département de Biologie Cellulaire et Infection, F-75015 Paris, France
- Inserm, U604, F-75015 Paris, France
- French National Institute for Agricultural Research (INRA), Unité sous-contrat 2020, F-75015 Paris, France
| |
Collapse
|
35
|
Mannala GK, Izar B, Rupp O, Schultze T, Goesmann A, Chakraborty T, Hain T. Listeria monocytogenes Induces a Virulence-Dependent microRNA Signature That Regulates the Immune Response in Galleria mellonella. Front Microbiol 2017; 8:2463. [PMID: 29312175 PMCID: PMC5733040 DOI: 10.3389/fmicb.2017.02463] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/27/2017] [Indexed: 12/11/2022] Open
Abstract
microRNAs (miRNAs) coordinate several physiological and pathological processes by regulating the fate of mRNAs. Studies conducted in vitro indicate a role of microRNAs in the control of host-microbe interactions. However, there is limited understanding of miRNA functions in in vivo models of bacterial infections. In this study, we systematically explored changes in miRNA expression levels of Galleria mellonella larvae (greater-wax moth), a model system that recapitulates the vertebrate innate immunity, following infection with L. monocytogenes. Using an insect-specific miRNA microarray with more than 2000 probes, we found differential expression of 90 miRNAs (39 upregulated and 51 downregulated) in response to infection with L. monocytogenes. We validated the expression of a subset of miRNAs which have mammalian homologs of known or predicted function. In contrast, non-pathogenic L. innocua failed to induce these miRNAs, indicating a virulence-dependent miRNA deregulation. To predict miRNA targets using established algorithms, we generated a publically available G. mellonella transcriptome database. We identified miRNA targets involved in innate immunity, signal transduction and autophagy, including spätzle, MAP kinase, and optineurin, respectively, which exhibited a virulence-specific differential expression. Finally, in silico estimation of minimum free energy of miRNA-mRNA duplexes of validated microRNAs and target transcripts revealed a regulatory network of the host immune response to L. monocytogenes. In conclusion, this study provides evidence for a role of miRNAs in the regulation of the innate immune response following bacterial infection in a simple, rapid and scalable in vivo model that may predict host-microbe interactions in higher vertebrates.
Collapse
Affiliation(s)
- Gopala K Mannala
- Institute of Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Benjamin Izar
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States.,Broad Institute of MIT and Harvard University, Cambridge, MA, United States
| | - Oliver Rupp
- Department of Bioinformatics and Systems Biology, Justus-Liebig University, Giessen, Germany
| | - Tilman Schultze
- Institute of Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Alexander Goesmann
- Department of Bioinformatics and Systems Biology, Justus-Liebig University, Giessen, Germany
| | - Trinad Chakraborty
- Institute of Medical Microbiology, Justus-Liebig University, Giessen, Germany
| | - Torsten Hain
- Institute of Medical Microbiology, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
36
|
Victoria B, Nunez Lopez YO, Masternak MM. MicroRNAs and the metabolic hallmarks of aging. Mol Cell Endocrinol 2017; 455:131-147. [PMID: 28062199 PMCID: PMC5724961 DOI: 10.1016/j.mce.2016.12.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 10/10/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022]
Abstract
Aging, the natural process of growing older, is characterized by a progressive deterioration of physiological homeostasis at the cellular, tissue, and organismal level. Metabolically, the aging process is characterized by extensive changes in body composition, multi-tissue/multi-organ insulin resistance, and physiological declines in multiple signaling pathways including growth hormone, insulin/insulin-like growth factor 1, and sex steroids regulation. With this review, we intend to consolidate published information about microRNAs that regulate critical metabolic processes relevant to aging. In certain occasions we uncover relationships likely relevant to aging, which has not been directly described before, such as the miR-451/AMPK axis. We have also included a provocative section highlighting the potential role in aging of a new designation of miRNAs, namely fecal miRNAs, recently discovered to regulate intestinal microbiota in mammals.
Collapse
Affiliation(s)
- Berta Victoria
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, USA.
| | - Yury O Nunez Lopez
- Translational Research Institute for Metabolism & Diabetes. Florida Hospital, 301 East Princeton St, Orlando, FL 32804, USA.
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, USA; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, 15 Garbary St., 61-866, Poznan, Poland.
| |
Collapse
|
37
|
Williams MR, Stedtfeld RD, Tiedje JM, Hashsham SA. MicroRNAs-Based Inter-Domain Communication between the Host and Members of the Gut Microbiome. Front Microbiol 2017; 8:1896. [PMID: 29021788 PMCID: PMC5624305 DOI: 10.3389/fmicb.2017.01896] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Accepted: 09/15/2017] [Indexed: 12/31/2022] Open
Abstract
The gut microbiome is an important modulator of host gene expression, impacting important functions such as the innate immune response. Recent evidence suggests that the inter-domain communication between the gut microbiome and host may in part occur via microRNAs (small, non-coding RNA molecules) which are often differentially expressed in the presence of bacteria and can even be released and taken up by bacteria. The role of microRNAs in microbiome–host communication in intestinal diseases is not fully understood, particularly in diseases impacted by exposure to environmental toxicants. Here, we review the present knowledge in the areas of microbiome and microRNA expression-based communication, microbiome and intestinal disease relationships, and microRNA expression responses to intestinal diseases. We also examine potential links between host microRNA–microbiota communication and exposure to environmental toxicants by reviewing connections between (i) toxicants and microRNA expression, (ii) toxicants and gut diseases, and (iii) toxicants and the gut microbiome. Future multidisciplinary research in this area is needed to uncover these interactions with the potential to impact how gut-microbiome associated diseases [e.g., inflammatory bowel disease (IBD) and many others] are managed.
Collapse
Affiliation(s)
- Maggie R Williams
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI, United States
| | - Robert D Stedtfeld
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI, United States
| | - James M Tiedje
- Center for Microbial Ecology, Michigan State University, East Lansing, MI, United States.,Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI, United States
| | - Syed A Hashsham
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI, United States.,Center for Microbial Ecology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
38
|
David DJV, Cossart P. Recent advances in understanding Listeria monocytogenes infection: the importance of subcellular and physiological context. F1000Res 2017; 6. [PMID: 28781746 PMCID: PMC5516218 DOI: 10.12688/f1000research.11363.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2017] [Indexed: 01/04/2023] Open
Abstract
The bacterial pathogen
Listeria monocytogenes (
Lm) is the causative agent of listeriosis, a rare but fatal foodborne disease. During infection,
Lm can traverse several host barriers and enter the cytosol of a variety of cell types. Thus, consideration of the extracellular and intracellular niches of
Lm is critical for understanding the infection process. Here, we review advances in our understanding of
Lm infection and highlight how the interactions between the host and the pathogen are context dependent. We discuss discoveries of how
Lm senses entry into the host cell cytosol. We present findings concerning how the nature of the various cytoskeleton components subverted by
Lm changes depending on both the stage of infection and the subcellular context. We present discoveries of critical components required for
Lm traversal of physiological barriers. Interactions between the host gut microbiota and
Lm will be briefly discussed. Finally, the importance of
Lm biodiversity and post-genomics approaches as a promising way to discover novel virulence factors will be highlighted.
Collapse
Affiliation(s)
- Daryl J V David
- Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Pascale Cossart
- Unité des Interactions Bactéries-Cellules, Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| |
Collapse
|
39
|
Chen Q, Tong C, Ma S, Zhou L, Zhao L, Zhao X. Involvement of MicroRNAs in Probiotics-Induced Reduction of the Cecal Inflammation by Salmonella Typhimurium. Front Immunol 2017; 8:704. [PMID: 28659929 PMCID: PMC5468434 DOI: 10.3389/fimmu.2017.00704] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 05/31/2017] [Indexed: 12/20/2022] Open
Abstract
The microRNAs (miRNAs) have been shown to play important roles in the development of the immune system and in regulation of host inflammation responses. Probiotics can effectively alleviate the inflammation caused by Salmonella in chickens. However, whether and how miRNAs are involved in modulation of the inflammation response in the gut of chickens have not been reported. In this study, the impact of a probiotics, Lactobacillus plantarum Z01 (LPZ01), was investigated on the cecal miRNAs and cytokine secretions in Salmonella Typhimurium (S. Typhimurium)-infected chickens at the age of 3 days. Newly hatched chicks were assigned to four groups (1): NC (basal diet) (2): S (basal diet + S. Typhimurium challenged) (3): SP (basal diet + S. Typhimurium challenged + LPZ01) (4): P (basal diet + LPZ01). In comparison with the S group, chicks in the SP group reduced the number of S. Typhimurium and had lower levels of interferon-γ and lipopolysaccharide-induced tumor necrosis factor alpha factor (LITAF) in ceca post challenge. Expression of 14 miRNAs was significantly affected by the presence of S. Typhimurium and/or lactobacillus. Five differential expression miRNAs (gga-miR-215-5p, gga-miR-3525, gga-miR-193a-5p, gga-miR-122-5p, and gga-miR-375) were randomly selected for confirmation by the RT-PCR. Predicted target genes of differentially expressed miRNAs were enriched in regulation of cAMP-dependent protein kinase activity, stress-activated MAPK cascade, immune system development and regulation of immune system process as well as in immune related pathways such as MAPK and Wnt signaling pathways. The relationship between changes of miRNAs and changes of cytokines was explored. Finally, 119 novel miRNAs were identified in 36 libraries totally. Identification of novel miRNAs significantly expanded the repertoire of chicken miRNAs and provided the basis for understanding the function of miRNAs in the host. Our results suggest that the probiotics reduce the inflammation of the S. Typhimurium infection in neonatal broiler chicks, at least partially, through regulation of miRNAs expression.
Collapse
Affiliation(s)
- Qiaoling Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chao Tong
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Shaoyang Ma
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Luoxiong Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Lili Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.,Department of Animal Science, McGill University, Montreal, QC, Canada
| |
Collapse
|
40
|
Rolhion N, Cossart P. How the study of Listeria monocytogenes has led to new concepts in biology. Future Microbiol 2017; 12:621-638. [PMID: 28604108 DOI: 10.2217/fmb-2016-0221] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The opportunistic intracellular bacterial pathogen Listeria monocytogenes has in 30 years emerged as an exceptional bacterial model system in infection biology. Research on this bacterium has provided considerable insight into how pathogenic bacteria adapt to mammalian hosts, invade eukaryotic cells, move intracellularly, interfere with host cell functions and disseminate within tissues. It also contributed to unveil features of normal host cell pathways and unsuspected functions of previously known cellular proteins. This review provides an updated overview of our knowledge on this pathogen. In many examples, findings on L. monocytogenes provided the basis for new concepts in bacterial regulation, cell biology and infection processes.
Collapse
Affiliation(s)
- Nathalie Rolhion
- Département de Biologie Cellulaire et Infection, Unité des Interactions Bactéries-Cellules, Institut Pasteur, F-75015 Paris, France.,INSERM, U604, F-75015 Paris, France.,INRA, Unité sous-contrat 2020, F-75015 Paris, France
| | - Pascale Cossart
- Département de Biologie Cellulaire et Infection, Unité des Interactions Bactéries-Cellules, Institut Pasteur, F-75015 Paris, France.,INSERM, U604, F-75015 Paris, France.,INRA, Unité sous-contrat 2020, F-75015 Paris, France
| |
Collapse
|
41
|
Duval M, Cossart P, Lebreton A. Mammalian microRNAs and long noncoding RNAs in the host-bacterial pathogen crosstalk. Semin Cell Dev Biol 2017; 65:11-19. [PMID: 27381344 PMCID: PMC7089780 DOI: 10.1016/j.semcdb.2016.06.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 05/30/2016] [Accepted: 06/01/2016] [Indexed: 12/20/2022]
Abstract
Gene expression regulation is a critical question in host-pathogen interactions, and RNAs act as key players in this process. In this review, we focus on the mammalian RNA response to bacterial infection, with a special interest on microRNAs and long non-coding RNAs. We discuss the role of cellular miRNAs in immunity, the implication of circulating miRNAs as well as the influence of the microbiome on the miRNA response. We also review how pathogens counteract the host miRNA expression. Interestingly, bacterial non-coding RNAs regulate host gene expression and conversely eukaryotic miRNAs may regulate bacterial gene expression. Overall, the characterization of RNA regulatory networks represents an emerging theme in the field of host pathogen interactions.
Collapse
Affiliation(s)
- Mélodie Duval
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, USC2020, 75015 Paris, France
| | - Pascale Cossart
- Institut Pasteur, Unité des Interactions Bactéries-Cellules, 75015 Paris, France; Inserm, U604, 75015 Paris, France; INRA, USC2020, 75015 Paris, France.
| | - Alice Lebreton
- École Normale Supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'École Normale Supérieure (IBENS), Équipe Infection et Devenir de l'ARN, 75005 Paris, France; INRA, IBENS, 75005 Paris, France.
| |
Collapse
|
42
|
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: 1.6] [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
|
43
|
Peck BCE, Mah AT, Pitman WA, Ding S, Lund PK, Sethupathy P. Functional Transcriptomics in Diverse Intestinal Epithelial Cell Types Reveals Robust MicroRNA Sensitivity in Intestinal Stem Cells to Microbial Status. J Biol Chem 2017; 292:2586-2600. [PMID: 28053090 DOI: 10.1074/jbc.m116.770099] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 12/23/2016] [Indexed: 01/01/2023] Open
Abstract
Gut microbiota play an important role in regulating the development of the host immune system, metabolic rate, and at times, disease pathogenesis. The factors and mechanisms that mediate interactions between microbiota and the intestinal epithelium are not fully understood. We provide novel evidence that microbiota may control intestinal epithelial stem cell (IESC) proliferation in part through microRNAs (miRNAs). We demonstrate that miRNA profiles differ dramatically across functionally distinct cell types of the mouse jejunal intestinal epithelium and that miRNAs respond to microbiota in a highly cell type-specific manner. Importantly, we also show that miRNAs in IESCs are more prominently regulated by microbiota compared with miRNAs in any other intestinal epithelial cell subtype. We identify miR-375 as one miRNA that is significantly suppressed by the presence of microbiota in IESCs. Using a novel method to knockdown gene and miRNA expression ex vivo enteroids, we demonstrate that we can knock down gene expression in Lgr5+ IESCs. Furthermore, when we knock down miR-375 in IESCs, we observe significantly increased proliferative capacity. Understanding the mechanisms by which microbiota regulate miRNA expression in IESCs and other intestinal epithelial cell subtypes will elucidate a critical molecular network that controls intestinal homeostasis and, given the heightened interest in miRNA-based therapies, may offer novel therapeutic strategies in the treatment of gastrointestinal diseases associated with altered IESC function.
Collapse
Affiliation(s)
- Bailey C E Peck
- From the Curriculum in Genetics and Molecular Biology, .,Department of Genetics
| | | | | | - Shengli Ding
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - P Kay Lund
- From the Curriculum in Genetics and Molecular Biology.,Department of Nutrition, and.,Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Praveen Sethupathy
- From the Curriculum in Genetics and Molecular Biology, .,Department of Genetics
| |
Collapse
|
44
|
Yau TO, Wu CW, Tang CM, Chen Y, Fang J, Dong Y, Liang Q, Ng SSM, Chan FKL, Sung JJY, Yu J. MicroRNA-20a in human faeces as a non-invasive biomarker for colorectal cancer. Oncotarget 2016; 7:1559-68. [PMID: 26621842 PMCID: PMC4811480 DOI: 10.18632/oncotarget.6403] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 11/15/2015] [Indexed: 12/18/2022] Open
Abstract
Objective Detection of microRNA (miRNA) aberrations in human faeces is a new approach for colorectal cancer (CRC) screening. The aim of this study was to characterise miR-20a in faeces as a non-invasive biomarker for diagnosis of CRC. Results miR-20a expression was significantly higher in the 40 CRC tumours compared to their respective adjacent normal tissues (P = 0.0065). Levels of miR-20a were also significantly higher in faecal samples from CRC patients (P < 0.0001). The area under receiver operating characteristic (AUROC) curve for miR-20a was 0.73, with a sensitivity of 55% and specificity of 82% for CRC patients compared with controls. No significant difference in the level of miR-20a was found between patients with proximal, distal, and rectal cancer. The use of antibiotics did not influence faecal miR-20a levels. Patients and Methods miR-20a was selected from an expression microarray containing 667 miRNAs. Further verification of miR-20a was performed in 40 pairs of primary CRC tissues, as well as 595 faecal samples (198 CRCs, 199 adenomas, and 198 healthy controls) using TaqMan probe based quantitative Real-Time PCR (qRT-PCR). Conclusions Faecal-based miR-20a can be utilised as a potential non-invasive biomarker for CRC screening.
Collapse
Affiliation(s)
- Tung On Yau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Chung Wah Wu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Ceen-Ming Tang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong.,Oxford University Clinical Academic Graduate School, John Radcliffe Hospital, Oxford, UK
| | - Yingxuan Chen
- Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Jingyuan Fang
- Renji Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yujuan Dong
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong.,Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Qiaoyi Liang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Simon Siu Man Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong
| | - Francis Ka Leung Chan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Joseph Jao Yiu Sung
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong
| |
Collapse
|
45
|
Das K, Garnica O, Dhandayuthapani S. Modulation of Host miRNAs by Intracellular Bacterial Pathogens. Front Cell Infect Microbiol 2016; 6:79. [PMID: 27536558 PMCID: PMC4971075 DOI: 10.3389/fcimb.2016.00079] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/14/2016] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs that regulate the expression of protein coding genes of viruses and eukaryotes at the post-transcriptional level. The eukaryotic genes regulated by miRNAs include those whose products are critical for biological processes such as cell proliferation, metabolic pathways, immune response, and development. It is now increasingly recognized that modulation of miRNAs associated with biological processes is one of the strategies adopted by bacterial pathogens to survive inside host cells. In this review, we present an overview of the recent findings on alterations of miRNAs in the host cells by facultative intracellular bacterial pathogens. In addition, we discuss how the altered miRNAs help in the survival of these pathogens in the intracellular environment.
Collapse
Affiliation(s)
| | | | - Subramanian Dhandayuthapani
- Center of Emphasis in Infectious Diseases and Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El PasoEl Paso, TX, USA
| |
Collapse
|
46
|
Fritz JV, Heintz-Buschart A, Ghosal A, Wampach L, Etheridge A, Galas D, Wilmes P. Sources and Functions of Extracellular Small RNAs in Human Circulation. Annu Rev Nutr 2016; 36:301-36. [PMID: 27215587 PMCID: PMC5479634 DOI: 10.1146/annurev-nutr-071715-050711] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Various biotypes of endogenous small RNAs (sRNAs) have been detected in human circulation, including microRNAs, transfer RNAs, ribosomal RNA, and yRNA fragments. These extracellular sRNAs (ex-sRNAs) are packaged and secreted by many different cell types. Ex-sRNAs exhibit differences in abundance in several disease states and have, therefore, been proposed for use as effective biomarkers. Furthermore, exosome-borne ex-sRNAs have been reported to elicit physiological responses in acceptor cells. Exogenous ex-sRNAs derived from diet (most prominently from plants) and microorganisms have also been reported in human blood. Essential issues that remain to be conclusively addressed concern the (a) presence and sources of exogenous ex-sRNAs in human bodily fluids, (b) detection and measurement of ex-sRNAs in human circulation, (c) selectivity of ex-sRNA export and import, (d) sensitivity and specificity of ex-sRNA delivery to cellular targets, and (e) cell-, tissue-, organ-, and organism-wide impacts of ex-sRNA-mediated cell-to-cell communication. We survey the present state of knowledge of most of these issues in this review.
Collapse
MESH Headings
- Animals
- Biological Transport
- Biomarkers/blood
- Cell Communication
- Diet
- Gastrointestinal Microbiome/immunology
- Gene Expression Regulation
- Host-Parasite Interactions
- Host-Pathogen Interactions
- Humans
- Immunity, Innate
- MicroRNAs/blood
- MicroRNAs/metabolism
- Models, Biological
- RNA, Bacterial/blood
- RNA, Bacterial/metabolism
- RNA, Plant/blood
- RNA, Plant/metabolism
- RNA, Ribosomal/blood
- RNA, Ribosomal/metabolism
- RNA, Small Interfering/blood
- RNA, Small Interfering/metabolism
- RNA, Small Untranslated/blood
- RNA, Small Untranslated/metabolism
- RNA, Transfer/blood
- RNA, Transfer/metabolism
- RNA, Viral/blood
- RNA, Viral/metabolism
Collapse
Affiliation(s)
- Joëlle V Fritz
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| | - Anna Heintz-Buschart
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| | - Anubrata Ghosal
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Linda Wampach
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| | - Alton Etheridge
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122
| | - David Galas
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
- Pacific Northwest Diabetes Research Institute, Seattle, Washington 98122
| | - Paul Wilmes
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Campus Belval, L-4367 Belvaux, Luxembourg; ,
| |
Collapse
|
47
|
Kalabat DY, Vitsky A, Scott W, Kindt E, Hayes K, John-Baptiste A, Huang W, Yang AH. Identification and Evaluation of Novel MicroRNA Biomarkers in Plasma and Feces Associated with Drug-induced Intestinal Toxicity. Toxicol Pathol 2016; 45:302-320. [PMID: 27189632 DOI: 10.1177/0192623316644992] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Gastrointestinal toxicity is dose limiting with many therapeutic and anticancer agents. Real-time, noninvasive detection of markers of toxicity in biofluids is advantageous. Ongoing research has revealed microRNAs as potential diagnostic and predictive biomarkers for the detection of select organ toxicities. To study the potential utility of microRNA biomarkers of intestinal injury in a preclinical toxicology species, we evaluated 3 rodent models of drug-induced intestinal toxicity, each with a distinct mechanism of toxicity. MiR-215 and miR-194 were identified as putative intestinal toxicity biomarkers. Both were evaluated in plasma and feces and compared to plasma citrulline, an established intestinal injury biomarker. Following intestinal toxicant dosing, microRNA changes in feces and plasma were detected noninvasively and correlated with histologic evidence of intestinal injury. Fecal miR-215 and miR-194 levels increased, and plasma miR-215 decreased in a dose- and time-dependent manner. Dose-dependent decreases in plasma miR-215 levels also preceded and correlated positively with plasma citrulline modulation, suggesting miR-215 is a more sensitive biomarker. Moreover, during the drug-free recovery phase, plasma miR-215 returned to predose levels, supporting a corresponding recovery of histologic lesions. Despite limitations, this study provides preliminary evidence that select microRNAs have the potential to act as noninvasive, sensitive, and quantitative biomarkers of intestinal injury.
Collapse
Affiliation(s)
- Dalia Y Kalabat
- 1 Drug Safety R&D, Pfizer Global R&D, San Diego, California, USA
| | - Allison Vitsky
- 1 Drug Safety R&D, Pfizer Global R&D, San Diego, California, USA
| | - Wesley Scott
- 1 Drug Safety R&D, Pfizer Global R&D, San Diego, California, USA
| | - Erick Kindt
- 2 Pharmacokinetics, Dynamics and Metabolism, Pfizer Global R&D, San Diego, California, USA
| | - Kyle Hayes
- 1 Drug Safety R&D, Pfizer Global R&D, San Diego, California, USA
| | | | - Wenhu Huang
- 1 Drug Safety R&D, Pfizer Global R&D, San Diego, California, USA
| | - Amy H Yang
- 1 Drug Safety R&D, Pfizer Global R&D, San Diego, California, USA
| |
Collapse
|
48
|
Circulating cell-free mature microRNAs and their target gene prediction in bovine metritis. Sci Rep 2016; 6:29509. [PMID: 27404038 PMCID: PMC4941693 DOI: 10.1038/srep29509] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/17/2016] [Indexed: 12/23/2022] Open
Abstract
Uterine infections in dairy cows are common after calving, reduce fertility and cause substantial economic losses. Conventional diagnosis (based on clinical signs) and treatment can be challenging. Serum microRNA (miRNA) profiles serve as non-invasive biomarkers in several pathological conditions including inflammatory diseases. The objective was to identify differentially expressed serum miRNAs in cows with metritis and normal uterus (four cows per group), integrate miRNAs to their target genes, and categorize target genes for biological processes involved in bacterial infection and inflammatory responses. Out of 84 bovine-specific, prioritized miRNAs analyzed, 30 were differentially expressed between metritis and normal cows (p ≤ 0.05, fold regulation ≥2 magnitudes). Bta-miR-15b, bta-miR-17-3p, bta-miR-16b, bta-miR-148a, bta-miR-26b, bta-miR-101 and bta-miR-29b were highly up-regulated whereas bta-miR-148b, bta-miR-199a-3p, bta-miR-122, bta-miR-200b and bta-miR-10a were highly down-regulated in cows with metritis compared to cows with normal uterus. Highly scored target genes of up-regulated and down-regulated miRNAs were categorized for various biological processes, including biological regulation, cellular process, developmental process, metabolic process, localization, multicellular organismal process, response to stimulus, immune system process, cellular components organization, apoptotic process, biological adhesion, developmental process, and locomotion that are critical to combat bacterial infections and provoke inflammatory responses.
Collapse
|
49
|
Abstract
MicroRNAs (miRNAs) are small, non-coding, RNA molecules that regulate gene expression. They have a long evolutionary history and are found in plants, viruses, and animals. Although initially discovered in 1993 in Caenorhabditis elegans, they were not appreciated as widespread and abundant gene regulators until the early 2000s. Studies in the last decade have found that miRNAs confer phenotypic robustness in the face of environmental perturbation, may serve as diagnostic and prognostic indicators of disease, underlie the pathobiology of a wide array of complex disorders, and represent compelling therapeutic targets. Pre-clinical studies in animal models have demonstrated that pharmacologic manipulation of miRNAs, mostly in the liver, can modulate metabolic phenotypes and even reverse the course of insulin resistance and diabetes. There is cautious optimism in the field about miRNA-based therapies for diabetes, several of which are already in various stages of clinical trials. This review will highlight both the promise and the most pressing challenges of therapeutic miRNA silencing in diabetes and related conditions.
Collapse
Affiliation(s)
- Praveen Sethupathy
- Department of Genetics, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
50
|
Mao MY, Yang YM, Li KZ, Lei L, Li M, Yang Y, Tao X, Yin JX, Zhang R, Ma XR, Hu T. The rnc Gene Promotes Exopolysaccharide Synthesis and Represses the vicRKX Gene Expressions via MicroRNA-Size Small RNAs in Streptococcus mutans. Front Microbiol 2016; 7:687. [PMID: 27242713 PMCID: PMC4861726 DOI: 10.3389/fmicb.2016.00687] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 04/26/2016] [Indexed: 02/05/2023] Open
Abstract
Dental caries is a biofilm-dependent disease that largely relies on the ability of Streptococcus mutans to synthesize exopolysaccharides. Although the rnc gene is suggested to be involved in virulence mechanisms in many other bacteria, the information regarding it in S. mutans is very limited. Here, using deletion or overexpression mutant assay, we demonstrated that rnc in S. mutans significantly positively regulated exopolysaccharide synthesis and further altered biofilm formation. Meanwhile, the cariogenecity of S. mutans was decreased by deletion of rnc in a specific pathogen-free (SPF) rat model. Interestingly, analyzing the expression at mRNA level, we found the downstream vic locus was repressed by rnc in S. mutans. Using deep sequencing and bioinformatics analysis, for the first time, three putative microRNA-size small RNAs (msRNAs) targeting vicRKX were predicted in S. mutans. The expression levels of these msRNAs were negatively correlated with vicRKX but positively correlated with rnc, indicating rnc probably repressed vicRKX expression through msRNAs at the post-transcriptional level. In all, the results present that rnc has a potential role in the regulation of exopolysaccharide synthesis and can affect vicRKX expressions via post-transcriptional repression in S. mutans. This study provides an alternative avenue for further research aimed at preventing caries.
Collapse
Affiliation(s)
- Meng-Ying Mao
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Ying-Ming Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Ke-Zeng Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengdu, China; Department of Dentistry, Yan'an Hospital Affiliated to Kunming Medical UniversityKunming, China
| | - Lei Lei
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Meng Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Yan Yang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Xiang Tao
- Chengdu Institute of Biology, Chinese Academy of Sciences Chengdu, China
| | - Jia-Xin Yin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| | - Ru Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan UniversityChengdu, China; Department of Endodontics and Operative Dentistry School of Stomatology, Capital Medical UniversityBeijing, China
| | - Xin-Rong Ma
- Chengdu Institute of Biology, Chinese Academy of Sciences Chengdu, China
| | - Tao Hu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu, China
| |
Collapse
|