1
|
Angulo M, Angulo C. Trained immunity-based vaccines: A vision from the one health initiative. Vaccine 2025; 43:126505. [PMID: 39520776 DOI: 10.1016/j.vaccine.2024.126505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Trained immunity-based vaccines (TIbV or TRIMbV) represent a novel approach to combating infectious diseases. The innate immune system in animals, including humans, exhibits "memory-like" functions. Remarkably, the immunological mechanisms -both epigenetic and metabolic-) underlying this memory enables immune cells to develop defensive and protective outcomes against unspecific pathogenic infections. Under this context, the One Health initiative promotes integrative efforts to combat zoonotic (and anthropozoonotic) diseases, which is critical because 3 of 4 animal infections are transmitted to humans. Therefore, TIbV constitutes a potential affordable approach to control zoonotic pathologies, especially under pandemic scenarios. This review describes the state-of-the-art TIbV and their hurdles, opportunities, and prospects for the One Health initiative to prevent, control, and treat infectious diseases.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C. (CIBNOR), Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C.S. 23096, Mexico.; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD), Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S., C.P. 23096, Mexico.
| |
Collapse
|
2
|
Angulo M, Angulo C. Analysis of the potential long-lasting effects of probiotic Debaryomyces hansenii CBS 8339 on trained immunity in newborn goats. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 162:105292. [PMID: 39577680 DOI: 10.1016/j.dci.2024.105292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Trained immunity has been described as the memory capacity of the innate immune system. Several microbial components have been shown to induce trained immunity. Research on the potential of probiotics to trigger these effects has been limited to a few in vitro studies but remains completely unknown in vivo. Components from the probiotic Debaryomyces hansenii CBS 8339 (Dh) have been shown to induce innate immune memory in goat kids and calves. In the present study, stimulating innate immune cells from newborn goats with probiotic Dh increased respiratory burst activity and nitric oxide production, while cell phagocytosis was unaffected. Glucose uptake was enhanced in goat's cells stimulated with Dh, but lactate production was decreased. In newborn goats, after the training scheme (via oral probiotic administration), cell phagocytosis, nitric oxide production and glycolysis - through the upregulation of AKT and HIF1A gene expression, glucose consumption and lactate production- were enhanced. The expression of IL1B gene was similar between the D. hansenii and control groups. Moreover, the potential long-lasting effects were assessed 30 days after initiation of the training scheme. Cell phagocytosis, respiratory burst and myeloperoxidase activity were enhanced, while glycolytic parameters remained unaffected. Altogether, the results of the present study suggest that the immune training scheme may induce trained immunity by the probiotic D. hansenii in newborn goats. However, our findings were not conclusive regarding the long-lasting (one-month) effects of trained immunity by probiotics.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico.
| |
Collapse
|
3
|
Ayyanar MP, Vijayan M. A review on gut microbiota and miRNA crosstalk: implications for Alzheimer's disease. GeroScience 2024:10.1007/s11357-024-01432-5. [PMID: 39562408 DOI: 10.1007/s11357-024-01432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and progressive neuronal damage. Recent research has highlighted the significant roles of the gut microbiota and microRNAs (miRNAs) in the pathogenesis of AD. This review explores the intricate interaction between gut microbiota and miRNAs, emphasizing their combined impact on Alzheimer's progression. First, we discuss the bidirectional communication within the gut-brain axis and how gut dysbiosis contributes to neuroinflammation and neurodegeneration in AD. Changes in gut microbiota composition in Alzheimer's patients have been linked to inflammation, which exacerbates disease progression. Next, we delve into the biology of miRNAs, focusing on their roles in gene regulation, neurodevelopment, and neurodegeneration. Dysregulated miRNAs are implicated in AD pathogenesis, influencing key processes like inflammation, tau pathology, and amyloid deposition. We then examine how the gut microbiota modulates miRNA expression, particularly in the brain, potentially altering neuroinflammatory responses and synaptic plasticity. The interplay between gut microbiota and miRNAs also affects blood-brain barrier integrity, further contributing to Alzheimer's pathology. Lastly, we explore therapeutic strategies targeting this gut microbiota-miRNA axis, including probiotics, prebiotics, and dietary interventions, aiming to modulate miRNA expression and improve AD outcomes. While promising, challenges remain in fully elucidating these interactions and translating them into effective therapies. This review highlights the importance of understanding the gut microbiota-miRNA relationship in AD, offering potential pathways for novel therapeutic approaches aimed at mitigating the disease's progression.
Collapse
Affiliation(s)
- Maruthu Pandian Ayyanar
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram, 624302, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
4
|
Velikova T, Kaouri IE, Bakopoulou K, Gulinac M, Naydenova K, Dimitrov M, Peruhova M, Lazova S. Mucosal Immunity and Trained Innate Immunity of the Gut. GASTROENTEROLOGY INSIGHTS 2024; 15:661-675. [DOI: 10.3390/gastroent15030048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2024] Open
Abstract
Mucosal immunity and trained innate immunity of the gut play a pivotal role in maintaining intestinal homeostasis and defending against microbial pathogens. This review provides an overview of the mechanisms underlying mucosal immunity and the concept of trained innate immunity in the gut. We discuss the interaction between gut microbiota and the host immune system, highlighting the role of epithelial cells, dendritic cells, and innate lymphoid cells, as well as the novel concept of trained innate immunity and its role in perpetuating or attenuating gut inflammation. We also comment on the current models for investigating mucosal immunity, their limitations, and how they can be overcome. Additionally, we explore the potential therapeutic implications of modulating mucosal immunity and trained innate immunity in gastrointestinal diseases. Only by elucidating the mechanisms underlying mucosal immunity and the concept of trained innate immunity, innovative approaches to modulate immune responses and restore intestinal homeostasis in the context of gastrointestinal disorders could be implemented.
Collapse
Affiliation(s)
- Tsvetelina Velikova
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
| | - Issa El Kaouri
- Faculty of Medicine, Medical University of Sofia, Boulevard “Akademik Ivan Evstratiev Geshov” 15, 1431 Sofia, Bulgaria
| | - Konstantina Bakopoulou
- Faculty of Medicine, Medical University of Sofia, Boulevard “Akademik Ivan Evstratiev Geshov” 15, 1431 Sofia, Bulgaria
| | - Milena Gulinac
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Kremena Naydenova
- Clinical Center of Allergology, University Hospital Alexandrovska, Medical University of Sofia, Georgi Sofyiski 1 Str., 1431 Sofia, Bulgaria
| | - Martin Dimitrov
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov Str., 1164 Sofia, Bulgaria
| | - Milena Peruhova
- Division of Gastroenterology, University Hospital “Heart and Brain”, Zdrave St. 1, 8000 Burgas, Bulgaria
| | - Snezhina Lazova
- Medical Faculty, Sofia University “St. Kliment Ohridski”, 1 Kozyak Str., 1407 Sofia, Bulgaria
- Pediatric Clinic, University Hospital “N. I. Pirogov”, 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria
- Department of Healthcare, Faculty of Public Health “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Bialo More 8 Str., 1527 Sofia, Bulgaria
| |
Collapse
|
5
|
Lin X, Han H, Wang N, Wang C, Qi M, Wang J, Liu G. The Gut Microbial Regulation of Epigenetic Modification from a Metabolic Perspective. Int J Mol Sci 2024; 25:7175. [PMID: 39000282 PMCID: PMC11241073 DOI: 10.3390/ijms25137175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Obesity is a global health challenge that has received increasing attention in contemporary research. The gut microbiota has been implicated in the development of obesity, primarily through its involvement in regulating various host metabolic processes. Recent research suggests that epigenetic modifications may serve as crucial pathways through which the gut microbiota and its metabolites contribute to the pathogenesis of obesity and other metabolic disorders. Hence, understanding the interplay between gut microbiota and epigenetic mechanisms is crucial for elucidating the impact of obesity on the host. This review primarily focuses on the understanding of the relationship between the gut microbiota and its metabolites with epigenetic mechanisms in several obesity-related pathogenic mechanisms, including energy dysregulation, metabolic inflammation, and maternal inheritance. These findings could serve as novel therapeutic targets for probiotics, prebiotics, and fecal microbiota transplantation tools in treating metabolic disruptions. It may also aid in developing therapeutic strategies that modulate the gut microbiota, thereby regulating the metabolic characteristics of obesity.
Collapse
Affiliation(s)
- Xingtong Lin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Hui Han
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Nan Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Chengming Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Ming Qi
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Jing Wang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- Yuelushan Laboratory, Changsha 410128, China
| | - Gang Liu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (X.L.); (H.H.); (N.W.); (C.W.); (M.Q.)
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
6
|
Carucci L, Coppola S, Carandente R, Canani RB. Targeting Food Allergy with Probiotics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:79-93. [PMID: 39060732 DOI: 10.1007/978-3-031-58572-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
The globally dramatic increase in food allergy prevalence and severity is demanding effective preventive and therapeutic strategies. Food allergy derives from a defect of immune tolerance mechanisms. Immune tolerance is modulated by gut microbiome composition and function, and gut microbiome dysbiosis has been associated with the development of food allergy. Selected probiotic strains could regulate immune tolerance mechanisms. The mechanisms are multiple and are still not completely defined. Increasing evidence is providing useful information on the choice of optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge on the crucial role played by postbiotic gut microbiome-derived metabolites, such as butyrate, is also opening the way to a post- biotic approach in the stimulation of immune tolerance.
Collapse
Affiliation(s)
- Laura Carucci
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy
| | - Serena Coppola
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy
| | - Rosilenia Carandente
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science at the University of Naples Federico II, Naples, Italy.
| |
Collapse
|
7
|
Gurunathan S, Thangaraj P, Kim JH. Postbiotics: Functional Food Materials and Therapeutic Agents for Cancer, Diabetes, and Inflammatory Diseases. Foods 2023; 13:89. [PMID: 38201117 PMCID: PMC10778838 DOI: 10.3390/foods13010089] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Postbiotics are (i) "soluble factors secreted by live bacteria, or released after bacterial lysis, such as enzymes, peptides, teichoic acids, peptidoglycan-derived muropeptides, polysaccharides, cell-surface proteins and organic acids"; (ii) "non-viable metabolites produced by microorganisms that exert biological effects on the hosts"; and (iii) "compounds produced by microorganisms, released from food components or microbial constituents, including non-viable cells that, when administered in adequate amounts, promote health and wellbeing". A probiotic- and prebiotic-rich diet ensures an adequate supply of these vital nutrients. During the anaerobic fermentation of organic nutrients, such as prebiotics, postbiotics act as a benevolent bioactive molecule matrix. Postbiotics can be used as functional components in the food industry by offering a number of advantages, such as being added to foods that are harmful to probiotic survival. Postbiotic supplements have grown in popularity in the food, cosmetic, and healthcare industries because of their numerous health advantages. Their classification depends on various factors, including the type of microorganism, structural composition, and physiological functions. This review offers a succinct introduction to postbiotics while discussing their salient features and classification, production, purification, characterization, biological functions, and applications in the food industry. Furthermore, their therapeutic mechanisms as antibacterial, antiviral, antioxidant, anticancer, anti-diabetic, and anti-inflammatory agents are elucidated.
Collapse
Affiliation(s)
- Sangiliyandi Gurunathan
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641021, Tamil Nadu, India;
| | - Pratheep Thangaraj
- Department of Biotechnology, Rathinam College of Arts and Science, Eachanari, Coimbatore 641021, Tamil Nadu, India;
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Republic of Korea
| |
Collapse
|
8
|
Guo N, Lv L. Mechanistic insights into the role of probiotics in modulating immune cells in ulcerative colitis. Immun Inflamm Dis 2023; 11:e1045. [PMID: 37904683 PMCID: PMC10571014 DOI: 10.1002/iid3.1045] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/15/2023] [Accepted: 09/29/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a persistent inflammatory disorder that affects the gastrointestinal tract, mainly the colon, which is defined by inflammatory responses and the formation of ulcers. Probiotics have been shown to directly impact various immune cells, including dendritic cells (DCs), macrophages, natural killer (NK) cells, and T and B cells. By interacting with cell surface receptors, they regulate immune cell activity, produce metabolites that influence immune responses, and control the release of cytokines and chemokines. METHODS This article is a comprehensive review wherein we conducted an exhaustive search across published literature, utilizing reputable databases like PubMed and Web of Science. Our focus centered on pertinent keywords, such as "UC," 'DSS," "TNBS," "immune cells," and "inflammatory cytokines," to compile the most current insights regarding the therapeutic potential of probiotics in managing UC. RESULTS This overview aims to provide readers with a comprehensive understanding of the effects of probiotics on immune cells in relation to UC. Probiotics have a crucial role in promoting the proliferation of regulatory T cells (Tregs), which are necessary for preserving immunological homeostasis and regulating inflammatory responses. They also decrease the activation of pro-inflammatory cells like T helper 1 (Th1) and Th17 cells, contributing to UC development. Thus, probiotics significantly impact both direct and indirect pathways of immune cell regulation in UC, promoting Treg differentiation, inhibiting pro-inflammatory cell activation, and regulating cytokine and chemokine release. CONCLUSION Probiotics demonstrate significant potential in modulating the immune reactions in UC. Their capacity to modulate different immune cells and inflammation-related processes makes them a promising therapeutic approach for managing UC. However, further studies are warranted to optimize their use and fully elucidate the molecular mechanisms underlying their beneficial effects in UC treatment.
Collapse
Affiliation(s)
- Ni Guo
- Department of GastroenterologyShengzhou People's Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)ShengzhouZhejiang ProvinceChina
| | - Lu‐lu Lv
- Department of GastroenterologyShengzhou People's Hospital (The First Affiliated Hospital of Zhejiang University Shengzhou Branch)ShengzhouZhejiang ProvinceChina
| |
Collapse
|
9
|
Mirfeizi Z, Mahmoudi M, Faridzadeh A. Probiotics as a complementary treatment in systemic lupus erythematosus: A systematic review. Health Sci Rep 2023; 6:e1640. [PMID: 37877130 PMCID: PMC10591540 DOI: 10.1002/hsr2.1640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/14/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
Introduction Systemic lupus erythematosus (SLE) is a chronic autoimmune disease that primarily affects young women. SLE has no recognized etiology but it is believed to be triggered by a number of factors, including genetic predisposition, hormonal influences, and environmental conditions. Dysbiosis in the gut microbiota has emerged as a potential mechanism connecting the intestinal microbiome to the breakdown of self-tolerance and chronic inflammation. This review aims to investigate the role of probiotics in modulating the gut microbiome and their potential therapeutic benefits in managing SLE, providing insights for future research and clinical practice. Methods We conducted a thorough search for papers published up to June 2023 in databases such as PubMed/MEDLINE, Web of Science, Scopus, and Cochrane Library. Results The systematic review identified 22 articles examining the effects of probiotics on SLE. These studies-which include in vivo tests, in vitro research, and clinical trials-indicate that probiotics may be effective against inflammation, and improve immunological responses and metabolic profiles in SLE patients. Most in vivo studies were assessed as medium to high quality, while the randomized controlled trial was deemed of high quality. Conclusion According to the findings of our systematic review, probiotics may be used in conjunction with other treatments to manage SLE. Nonetheless, current data is limited, and more randomized controlled trials would be required to fully examine their effectiveness.
Collapse
Affiliation(s)
- Zahra Mirfeizi
- Rheumatology Department, Rheumatic Diseases Research CenterMashhad University of Medical SciencesMashhadIran
| | - Mahmoud Mahmoudi
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
| | - Arezoo Faridzadeh
- Immunology Research CenterMashhad University of Medical SciencesMashhadIran
- Department of Immunology and Allergy, School of MedicineMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
10
|
Ahangari Maleki M, Malek Mahdavi A, Soltani-Zangbar MS, Yousefi M, Khabbazi A. Randomized double-blinded controlled trial on the effect of synbiotic supplementation on IL-17/IL-23 pathway and disease activity in patients with axial spondyloarthritis. Immunopharmacol Immunotoxicol 2023; 45:43-51. [PMID: 35947039 DOI: 10.1080/08923973.2022.2112220] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Interleukin 17 (IL17)-expressing CD4+ T cells and IL-17/IL-23 pathway play a key role in the pathogenesis of axial spondyloarthritis (axSpA). Synbiotics have been suggested due to their immunomodulatory effects in the treatment of autoimmune diseases. This randomized double-blind, placebo-controlled trial was designed to assess the effects of synbiotic supplement on IL-17/IL-23 pathway and disease activity in patients with axSpA. METHODS Forty-eight axSpA patients were randomly allocated to use one synbiotic capsule or placebo daily for 12 weeks. Disease activity was assessed using the Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) and ASAS-endorsed disease activity score-C-reactive protein (ASDAS-CRP). The secondary outcome was proportion of IL17-expressing CD4+ T cells, IL-17 and IL-23 gene expression, and supernatant levels of IL-17 and IL-23, which were measured at the baseline and end of the trial. RESULTS A total of 48 patients were randomized into the synbiotic and placebo groups. Thirty-eight patients completed the study. Synbiotic supplementation significantly reduced the proportion of IL17-expressing CD4+ T cells (4.88 ± 2.47 vs. 2.16 ± 1.25), gene expression of IL-17 (1.03 ± 0.24 vs. 0.65 ± 0.26) and IL-23 (1.01 ± 0.13 vs. 0.68 ± 0.24) and serum IL-17 (38.22 ± 14.40 vs. 24.38 ± 11.68) and IL-23 (51.77 ± 17.40 vs. 32.16 ± 12.46) compared with baseline. Significant differences between groups were noticed only in the proportion of IL17-expressing CD4+ T cells, and IL-17 and IL-23 gene expression. Synbiotic supplementation did not significantly alter BASDAI and ASDAS-CRP compared with baseline and placebo group at the end of trial. CONCLUSION Present study indicated beneficial effect of synbiotic supplement on IL-17/IL-23 pathway without improving disease activity in axSpApatients.HighlightsSynbiotic supplementation reduced IL17-expressing CD4+ T cells proportion in axSpA.Synbiotic supplementation decreased IL-17 and IL-23 gene expression in axSpA.Synbiotic supplementation did not change disease activity score in axSpA.
Collapse
Affiliation(s)
- Masoud Ahangari Maleki
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aida Malek Mahdavi
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Rahat Breath and Sleep Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Khabbazi
- Connective Tissue Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
11
|
Pandey H, Tang DWT, Wong SH, Lal D. Gut Microbiota in Colorectal Cancer: Biological Role and Therapeutic Opportunities. Cancers (Basel) 2023; 15:cancers15030866. [PMID: 36765824 PMCID: PMC9913759 DOI: 10.3390/cancers15030866] [Citation(s) in RCA: 40] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths worldwide. While CRC is thought to be an interplay between genetic and environmental factors, several lines of evidence suggest the involvement of gut microbiota in promoting inflammation and tumor progression. Gut microbiota refer to the ~40 trillion microorganisms that inhabit the human gut. Advances in next-generation sequencing technologies and metagenomics have provided new insights into the gut microbial ecology and have helped in linking gut microbiota to CRC. Many studies carried out in humans and animal models have emphasized the role of certain gut bacteria, such as Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, and colibactin-producing Escherichia coli, in the onset and progression of CRC. Metagenomic studies have opened up new avenues for the application of gut microbiota in the diagnosis, prevention, and treatment of CRC. This review article summarizes the role of gut microbiota in CRC development and its use as a biomarker to predict the disease and its potential therapeutic applications.
Collapse
Affiliation(s)
- Himani Pandey
- Redcliffe Labs, Electronic City, Noida 201301, India
| | - Daryl W. T. Tang
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Sunny H. Wong
- Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Correspondence: (S.H.W.); (D.L.)
| | - Devi Lal
- Department of Zoology, Ramjas College, University of Delhi, Delhi 110007, India
- Correspondence: (S.H.W.); (D.L.)
| |
Collapse
|
12
|
Topol IA, Polyakova IS, Elykova AV. Role of intestinal microbiota in regulation of immune reactions of gut-associated lymphoid tissue under stress and following the modulation of its composition by antibiotics and probiotics administration. JOURNAL OF MICROBIOLOGY, EPIDEMIOLOGY AND IMMUNOBIOLOGY 2023. [DOI: 10.36233/0372-9311-270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Over the past two decades, active study of the microbial ecosystem of the host organism gastrointestinal tract has led to the recognition of gut microbiome as a "key player" that carries a significant immune pressure and is responsible both for the course of physiological processes and for the development of pathological conditions in humans and animals. A vast number of bacteria living in the human gastrointestinal tract are considered as an organ functioning in dialogue in formation of immunological tolerance, the regulation of normal functional activity of the immune system and maintaining the intestinal mucosa homeostasis. However, disturbances in interaction between these physiological systems is closely related to the pathogenesis of different immune-mediated diseases. In turn, in a large number of works chronic social stress, along with the use of antibiotics, pre- and probiotics, is recognized as one of the leading factors modulating in the microbiota of the gastrointestinal tract. This review focuses on the role of the gut microbiome in the regulation of immune responses of GALT under stress and modulation of its composition by antibiotics and probiotics administration.
Collapse
|
13
|
Li B, Yang B, Liu X, Zhao J, Ross RP, Stanton C, Zhang H, Chen W. Microbiota-assisted therapy for systemic inflammatory arthritis: advances and mechanistic insights. Cell Mol Life Sci 2022; 79:470. [PMID: 35932328 PMCID: PMC11072763 DOI: 10.1007/s00018-022-04498-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/22/2022]
Abstract
Research on the influence of gut microbiota on systemic inflammatory arthritis has exploded in the past decade. Gut microbiota changes may be a crucial regulatory component in systemic inflammatory arthritis. As a result of advancements in the field, microbiota-assisted therapy has evolved, but this discipline is still in its infancy. Consequently, we review the limitations of current systemic inflammatory arthritis treatment, analyze the connection between the microbiota and arthritis, and summarize the research progress of microbiota regulating systemic inflammatory arthritis and the further development aspects of microbiota-assisted therapy. Finally, the partial mechanisms of microbiota-assisted therapy of systemic inflammatory arthritis are being discussed. In general, this review summarizes the current progress, challenges, and prospects of microbiota-assisted therapy for systemic inflammatory arthritis and points out the direction for the development of microbiota-assisted therapy in the future.
Collapse
Affiliation(s)
- Bowen Li
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
| | - Xiaoming Liu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
| | - R Paul Ross
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China.
- APC Microbiome Ireland, University College Cork, Cork, Ireland.
| | - Catherine Stanton
- International Joint Research Center for Probiotics and Gut Health, Jiangnan University, Wuxi, Jiangsu, China
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China.
- School of Food Science and Technology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, China.
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China.
- Beijing Innovation Center of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China.
| |
Collapse
|
14
|
Kumari A, Bhawal S, Kapila S, Kapila R. Strain-specific effects of probiotic Lactobacilli on mRNA expression of epigenetic modifiers in intestinal epithelial cells. Arch Microbiol 2022; 204:411. [PMID: 35729284 DOI: 10.1007/s00203-022-03027-0] [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: 02/21/2022] [Revised: 05/18/2022] [Accepted: 05/30/2022] [Indexed: 11/27/2022]
Abstract
The epigenome of an organism is as important as the genome for the normal development and functioning of an individual. The human epigenome can be affected by various environmental factors including nutrients, microbiota and probiotics through epigenetic modifiers and mediates various health-promoting effects. The present study was aimed to explore the temporal changes in DNA and histone modifiers (DNMT1, TET2, p300, HDAC1, KMT2A, KDM5B, EzH2 and JMJD3) in intestinal epithelial cells (Caco-2) by probiotic lactobacilli (Limosilactobacillus fermentum MTCC 5898 and Lacticaseibacillus rhamnosus MTCC 5897) in comparison to opportunistic commensal pathogen Escherichia coli (ATCC 14849). Cells were treated separately with probiotic strains and E. coli for different durations and temporal changes in gene expression among DNA and histone modifiers were measured. Time-dependent studies showed that L. fermentum enhanced the transcription of epigenetic modifiers at 12 h of treatment (P < 0.05) contrary to E. coli which reduced the expression of these genes during the same duration of treatment. On the other hand, probiotic L. rhamnosus was not able to induce any significant changes in gene expression of these modifiers. Furthermore, during the exclusion of E. coli by L. fermentum, the probiotic was found to resist the changes made by E. coli in the transcription of some of the epigenetic modifiers. Thus, it is concluded that the probiotics modulated the mRNA expression of DNA and histone modifiers contrarily to E. coli in a strain-specific manner.
Collapse
Affiliation(s)
- Ankita Kumari
- Division of Animal Biochemistry, National Dairy Research Institute (NDRI), Karnal, Haryana, 132001, India
| | - Shalaka Bhawal
- Division of Animal Biochemistry, National Dairy Research Institute (NDRI), Karnal, Haryana, 132001, India
| | - Suman Kapila
- Division of Animal Biochemistry, National Dairy Research Institute (NDRI), Karnal, Haryana, 132001, India
| | - Rajeev Kapila
- Division of Animal Biochemistry, National Dairy Research Institute (NDRI), Karnal, Haryana, 132001, India.
| |
Collapse
|
15
|
Zhang W, Zhang J, Liu T, Xing J, Zhang H, Wang D, Tang D. Bidirectional effects of intestinal microbiota and antibiotics: a new strategy for colorectal cancer treatment and prevention. J Cancer Res Clin Oncol 2022; 148:2387-2404. [PMID: 35661254 DOI: 10.1007/s00432-022-04081-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/19/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE Colorectal cancer (CRC) is the third most common cancer worldwide, and its incidence and mortality rates are increasing every year. The intestinal microbiota has been called the "neglected organ" and there is growing evidence that the intestinal microbiota and its metabolites can be used in combination with immunotherapy, radiotherapy and chemotherapy to greatly enhance the treatment of colorectal cancer and to address some of the side effects and adverse effects of these therapies. Antibiotics have great potential to eliminate harmful microbiota, control infection, and reduce colorectal cancer side effects. However, the use of antibiotics has been a highly controversial issue, and numerous retrospective studies have shown that the use of antibiotics affects the effectiveness of treatment (especially immunotherapy). Understanding the bi-directional role of the gut microbiota and antibiotics will further enhance our research into the diagnosis and treatment of cancer. METHODS We searched the "PubMed" database and selected the following keywords "intestinal microbiota, antibiotics, treatment, prevention, colorectal cancer". In this review, we discuss the role of the intestinal microbiota in immunotherapy, radiotherapy, chemotherapy, diagnosis, and prevention of CRC. We also conclude that the intestinal microbiota and antibiotics work together to promote the treatment of CRC through a bidirectional effect. RESULTS We found that the intestinal microbiota plays a key role in promoting immunotherapy, chemotherapy, radiotherapy, diagnosis and prevention of CRC. In addition, gut microbiota and antibiotic interactions could be a new strategy for CRC treatment. CONCLUSION The bi-directional role of the intestinal microbiota and antibiotics plays a key role in the prevention, diagnosis, and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Wenjie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Jie Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Tian Liu
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Juan Xing
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Huan Zhang
- Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu Province, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Clinical Medical College, Northern Jiangsu Province Hospital, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
16
|
Xiong T, Zheng X, Zhang K, Wu H, Dong Y, Zhou F, Cheng B, Li L, Xu W, Su J, Huang J, Jiang Z, Li B, Zhang B, Lv G, Chen S. Ganluyin ameliorates DSS-induced ulcerative colitis by inhibiting the enteric-origin LPS/TLR4/NF-κB pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115001. [PMID: 35085745 DOI: 10.1016/j.jep.2022.115001] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/24/2021] [Accepted: 01/12/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD), that is associated with a significantly increased risk of colon cancer. As a classic traditional Chinese medicine, Ganluyin (GLY) has a long history as an anti-inflammatory medication, but its impacts on UC has not been established. AIM OF THE STUDY This study aims to evaluate the protective effect and mechanism of GLY on a pathway involving enteric-origin lipopolysaccharide (LPS), toll-like receptor (TLR)4, and NF-κB in mice with dextran sulfate sodium (DSS)-induced UC. MATERIALS AND METHODS After three weeks of intragastric administration of GLY, a UC model was induced in mice by administration of 4% DSS in drinking water for one week. The disease activity index (DAI) was measured, and histological staining was used to detect histopathological changes of colon. LPS content of the serum was measured by ELISA, and the expression of tight junction proteins and proteins related to TLR4/NF-κB pathway in colon were analyzed by immunohistochemistry or Western Blotting. The intestinal flora was analyzed by 16S rRNA sequencing. RESULTS GLY improved the histological pathological changes of DSS-induced UC, as assessed by DAI, colonic mucosal damage, inflammatory cell infiltration, and goblet cell and mucus reduction. GLY also protected the intestinal mucosal barrier by increasing the expression of the tight junction proteins, occludin, claudin-1, and ZO-1 and by reducing the serum LPS content and decreasing the expression of TLR4, MyD88, NF-κB, IL-6, IL-1β, and TNF-α proteins in colon. Analyses of the intestinal flora showed that GLY restored the homeostasis of the intestinal flora through increases in the abundance of Firmicutes and decreases in the abundance of Proteobacteria and Bacteroidetes, which is associated with the production of LPS. CONCLUSION GLY might exert an anti-UC effect by improving the colonic mucosal barrier and inhibiting the enteric-origin LPS/TLR4/NF-κB inflammatory pathway, and restoring the homeostasis of the intestinal flora in UC mice. These discoveries lay a strong foundation for GLY as a UC treatment.
Collapse
Affiliation(s)
- Taoxiu Xiong
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Xiang Zheng
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Ke Zhang
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Hansong Wu
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Yingjie Dong
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Fuchen Zhou
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Bingbing Cheng
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Linzi Li
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Wanfeng Xu
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Jie Su
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jiahui Huang
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Zetian Jiang
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Bo Li
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China
| | - Beibei Zhang
- Center for Food Evaluation, State Administration for Market Regulation, Beijing, 100070, PR China.
| | - Guiyuan Lv
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Suhong Chen
- Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, PR China.
| |
Collapse
|
17
|
Holubekova V, Kolkova Z, Kasubova I, Samec M, Mazurakova A, Koklesova L, Kubatka P, Rokos T, Kozubik E, Biringer K, Kudela E. Interaction of cervical microbiome with epigenome of epithelial cells: Significance of inflammation to primary healthcare. Biomol Concepts 2022; 13:61-80. [PMID: 35245973 DOI: 10.1515/bmc-2022-0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
One pillar of the predictive, preventive, and personalized medicine framework strategies is the female health. The evaluation of women's lifestyle and dietary habits in context with genetic and modifiable risk factors may reflect the prevention of cervical cancer before the occurrence of clinical symptoms and prediction of cervical lesion behavior. The main aim of this review is to analyze publications in the field of precision medicine that allow the use of research knowledge of cervical microbiome, epigenetic modifications, and inflammation in potential application in clinical practice. Personalized approach in evaluating patient's risk of future development of cervical abnormality should consider the biomarkers of the local microenvironment characterized by the microbial composition, epigenetic pattern of cervical epithelium, and presence of chronic inflammation. Novel sequencing techniques enable a more detailed characterization of actual state in cervical epithelium. Better understanding of all changes in multiomics level enables a better assessment of disease prognosis and selects the eligible targeted therapy in personalized medicine. Restoring of healthy vaginal microflora and reversing the outbreak of cervical abnormality can be also achieved by dietary habits as well as uptake of prebiotics, probiotics, synbiotics, microbial transplantation, and others.
Collapse
Affiliation(s)
- Veronika Holubekova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Zuzana Kolkova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Ivana Kasubova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Marek Samec
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Alena Mazurakova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, SK-03601, Slovakia
| | - Tomas Rokos
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Erik Kozubik
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| | - Erik Kudela
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin University Hospital, Martin, SK-03601, Slovakia
| |
Collapse
|
18
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
19
|
Ali A, Hamzaid NH, Ismail NAS. The Interplay of Nutriepigenomics, Personalized Nutrition and Clinical Practice in Managing Food Allergy. Life (Basel) 2021; 11:1275. [PMID: 34833150 PMCID: PMC8623511 DOI: 10.3390/life11111275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Food allergy in children has been a common issue due to the challenges of prescribing personalized nutrition with a lack of nutriepigenomics data. This has indeed further influenced clinical practice for appropriate management. While allergen avoidance is still the main principle in food allergy management, we require more information to advance the science behind nutrition, genes, and the immune system. Many researchers have highlighted the importance of personalized nutrition but there is a lack of data on how the decision is made. Thus, this review highlights the relationship among these key players in identifying the solution to the clinical management of food allergy with current nutriepigenomics data. The discussion integrates various inputs, including clinical assessments, biomarkers, and epigenetic information pertaining to food allergy, to curate a holistic and personalized approach to food allergy management in particular.
Collapse
Affiliation(s)
- Adli Ali
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nur Hana Hamzaid
- Dietetic Program & Centre for Rehabilitation and Special Needs Studies (iCaRehab), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Kuala Lumpur 50300, Malaysia;
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
20
|
Gutierrez A, Demond H, Brebi P, Ili CG. Novel Methylation Biomarkers for Colorectal Cancer Prognosis. Biomolecules 2021; 11:1722. [PMID: 34827720 PMCID: PMC8615818 DOI: 10.3390/biom11111722] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) comprises the third most common cancer worldwide and the second regarding number of deaths. In order to make a correct and early diagnosis to predict metastasis formation, biomarkers are an important tool. Although there are multiple signaling pathways associated with cancer progression, the most recognized are the MAPK pathway, p53 pathway, and TGF-β pathway. These pathways regulate many important functions in the cell, such as cell cycle regulation, proliferation, differentiation, and metastasis formation, among others. Changes in expression in genes belonging to these pathways are drivers of carcinogenesis. Often these expression changes are caused by mutations; however, epigenetic changes, such as DNA methylation, are increasingly acknowledged to play a role in the deregulation of oncogenic genes. This makes DNA methylation changes an interesting biomarkers in cancer. Among the newly identified biomarkers for CRC metastasis INHBB, SMOC2, BDNF, and TBRG4 are included, all of which are highly deregulated by methylation and closely associated with metastasis. The identification of such biomarkers in metastasis of CRC may allow a better treatment and early identification of cancer formation in order to perform better diagnostics and improve the life expectancy.
Collapse
Affiliation(s)
| | | | - Priscilla Brebi
- Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (A.G.); (H.D.)
| | - Carmen Gloria Ili
- Millennium Institute on Immunology and Immunotherapy, Laboratory of Integrative Biology (LIBi), Centro de Excelencia en Medicina Traslacional (CEMT), Scientific and Technological Bioresource Nucleus (BIOREN), Universidad de La Frontera, Temuco 4810296, Chile; (A.G.); (H.D.)
| |
Collapse
|
21
|
Rajeev R, Dwivedi AP, Sinha A, Agarwaal V, Dev RR, Kar A, Khosla S. Epigenetic interaction of microbes with their mammalian hosts. J Biosci 2021. [PMID: 34728591 PMCID: PMC8550911 DOI: 10.1007/s12038-021-00215-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
The interaction of microbiota with its host has the ability to alter the cellular functions of both, through several mechanisms. Recent work, from many laboratories including our own, has shown that epigenetic mechanisms play an important role in the alteration of these cellular functions. Epigenetics broadly refers to change in the phenotype without a corresponding change in the DNA sequence. This change is usually brought by epigenetic modifications of the DNA itself, the histone proteins associated with the DNA in the chromatin, non-coding RNA or the modifications of the transcribed RNA. These modifications, also known as epigenetic code, do not change the DNA sequence but alter the expression level of specific genes. Microorganisms seem to have learned how to modify the host epigenetic code and modulate the host transcriptome in their favour. In this review, we explore the literature that describes the epigenetic interaction of bacteria, fungi and viruses, with their mammalian hosts.
Collapse
|
22
|
Probiotics and Trained Immunity. Biomolecules 2021; 11:biom11101402. [PMID: 34680035 PMCID: PMC8533468 DOI: 10.3390/biom11101402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022] Open
Abstract
The characteristics of innate immunity have recently been investigated in depth in several research articles, and original findings suggest that innate immunity also has a memory capacity, which has been named “trained immunity”. This notion has revolutionized our knowledge of the innate immune response. Thus, stimulation of trained immunity represents a therapeutic alternative that is worth exploring. In this context, probiotics, live microorganisms which when administered in adequate amounts confer a health benefit on the host, represent attractive candidates for the stimulation of trained immunity; however, although numerous studies have documented the beneficial proprieties of these microorganisms, their mechanisms of action are not yet fully understood. In this review, we propose to explore the putative connection between probiotics and stimulation of trained immunity.
Collapse
|
23
|
Gupta T, Kaur H, Kapila S, Kapila R. Potential probiotic Lacticaseibacillus rhamnosus MTCC-5897 attenuates Escherichia coli induced inflammatory response in intestinal cells. Arch Microbiol 2021; 203:5703-5713. [PMID: 34476513 DOI: 10.1007/s00203-021-02541-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/31/2021] [Accepted: 08/18/2021] [Indexed: 12/31/2022]
Abstract
Probiotics are microbes having tremendous potential to prevent gastrointestinal disorders. In current investigation, immunomodulatory action of probiotic Lacticaseibacillus rhamnosus MTCC-5897 was studied during exclusion, competition and displacement of Escherichia coli on intestinal epithelial (Caco-2) cells. The incubation of intestinal cells with Escherichia coli, enhanced downstream signalling and activated nuclear factor kappa B (NF-κB). This significantly increased (p < 0.01) the pro-inflammatory cytokines (IL-8, TNF-α, IFN-ϒ) expression. While, incubation of epithelial cells with Lacticaseibacillus rhamnosus during exclusion and competition with Escherichia coli, counteracted these enhanced expressions. The immunomodulatory feature of Lacticaseibacillus rhamnosus was also highlighted with increased (p < 0.05) transcription of toll-like receptor-2 (TLR-2) and single Ig IL-1-related receptor (SIGIRR) along with diminished expression of TLR-4. Likewise, attenuation (p < 0.05) of E. coli-mediated enhanced nuclear translocation of NF-κB p-65 subunit by Lacticaseibacillus rhamnosus during exclusion was confirmed with western blotting. Thus, present finding establishes the prophylactic potential of Lacticaseibacillus rhamnosus against exclusion of Escherichia coli in intestinal cells.
Collapse
Affiliation(s)
- Taruna Gupta
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Harpreet Kaur
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Suman Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Rajeev Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| |
Collapse
|
24
|
Dunislawska A, Slawinska A, Gryzinska M, Siwek M. Interaction between early in ovo stimulation of the gut microbiota and chicken host - splenic changes in gene expression and methylation. J Anim Sci Biotechnol 2021; 12:73. [PMID: 34229755 PMCID: PMC8262062 DOI: 10.1186/s40104-021-00602-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 05/09/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Epigenetic regulation of the gene expression results from interaction between the external environment and transcription of the genetic information encoded in DNA. Methylated CpG regions within the gene promoters lead to silencing of the gene expression in most cases. Factors contributing to epigenetic regulation include intestinal microbiota, which in chicken can be potently modified by in ovo stimulation. The main aim of this study was to determine global and specific methylation patterns of the spleen under the influence of host-microbiome interaction. RESULTS Fertilized eggs of two genotypes: Ross 308 and Green-legged Partridgelike were in ovo stimulated on d 12 of incubation. The injected compounds were as follows: probiotic - Lactococcus lactis subsp. cremoris IBB477, prebiotic - galactooligosaccharides, and synbiotic - combination of both. Chickens were sacrificed on d 42 post-hatching. Spleen was collected, RNA and DNA were isolated and intended to gene expression, gene methylation and global methylation analysis. We have proved that negative regulation of gene expression after administration of bioactive substances in ovo might have epigenetic character. Epigenetic changes depend on the genotype and the substance administered in ovo. CONCLUSION Epigenetic nature of microbial reprogramming in poultry and extension of issues related to host-microbiome interaction is a new direction of this research.
Collapse
Affiliation(s)
- A Dunislawska
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, 85-084, Bydgoszcz, Poland.
| | - A Slawinska
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, 85-084, Bydgoszcz, Poland
| | - M Gryzinska
- Institute of Biological Basis of Animal Production, Sub-Department of General and Molecular Genetics, University of Life Sciences in Lublin, 20-032, Lublin, Poland
| | - M Siwek
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, 85-084, Bydgoszcz, Poland
| |
Collapse
|
25
|
Rajeev R, Dwivedi AP, Sinha A, Agarwaal V, Dev RR, Kar A, Khosla S. Epigenetic interaction of microbes with their mammalian hosts. J Biosci 2021; 46:94. [PMID: 34728591 PMCID: PMC8550911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/20/2021] [Indexed: 02/11/2023]
Abstract
The interaction of microbiota with its host has the ability to alter the cellular functions of both, through several mechanisms. Recent work, from many laboratories including our own, has shown that epigenetic mechanisms play an important role in the alteration of these cellular functions. Epigenetics broadly refers to change in the phenotype without a corresponding change in the DNA sequence. This change is usually brought by epigenetic modifications of the DNA itself, the histone proteins associated with the DNA in the chromatin, non-coding RNA or the modifications of the transcribed RNA. These modifications, also known as epigenetic code, do not change the DNA sequence but alter the expression level of specific genes. Microorganisms seem to have learned how to modify the host epigenetic code and modulate the host transcriptome in their favour. In this review, we explore the literature that describes the epigenetic interaction of bacteria, fungi and viruses, with their mammalian hosts.
Collapse
Affiliation(s)
- Ramisetti Rajeev
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Graduate Studies, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Ambey Prasad Dwivedi
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Graduate Studies, Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Anunay Sinha
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Graduate Studies, Regional Centre for Biotechnology (RCB), Faridabad, India
| | - Viplove Agarwaal
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| | | | - Anjana Kar
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
| | - Sanjeev Khosla
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Hyderabad, India
- Institute of Microbial Technology (IMTech), Chandigarh, India
| |
Collapse
|
26
|
Elhag DA, Kumar M, Al Khodor S. Exploring the Triple Interaction between the Host Genome, the Epigenome, and the Gut Microbiome in Type 1 Diabetes. Int J Mol Sci 2020; 22:ijms22010125. [PMID: 33374418 PMCID: PMC7795494 DOI: 10.3390/ijms22010125] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/13/2020] [Accepted: 12/15/2020] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes (T1D) is an auto-immune disorder characterized by a complex interaction between the host immune system and various environmental factors in genetically susceptible individuals. Genome-wide association studies (GWAS) identified different T1D risk and protection alleles, however, little is known about the environmental factors that can be linked to these alleles. Recent evidence indicated that, among those environmental factors, dysbiosis (imbalance) in the gut microbiota may play a role in the pathogenesis of T1D, affecting the integrity of the gut and leading to systemic inflammation and auto-destruction of the pancreatic β cells. Several studies have identified changes in the gut microbiome composition in humans and animal models comparing T1D subjects with controls. Those changes were characterized by a higher abundance of Bacteroides and a lower abundance of the butyrate-producing bacteria such as Clostridium clusters IV and XIVa. The mechanisms by which the dysbiotic bacteria and/or their metabolites interact with the genome and/or the epigenome of the host leading to destructive autoimmunity is still not clear. As T1D is a multifactorial disease, understanding the interaction between different environmental factors such as the gut microbiome, the genetic and the epigenetic determinants that are linked with the early appearance of autoantibodies can expand our knowledge about the disease pathogenesis. This review aims to provide insights into the interaction between the gut microbiome, susceptibility genes, epigenetic factors, and the immune system in the pathogenesis of T1D.
Collapse
|
27
|
Chen M, Zhu H, Mao YJ, Cao N, Yu YL, Li LY, Zhao Q, Wu M, Ye M. Regulation of IL12B Expression in Human Macrophages by TALEN-mediated Epigenome Editing. Curr Med Sci 2020; 40:900-909. [PMID: 33123904 DOI: 10.1007/s11596-020-2249-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 06/22/2020] [Indexed: 10/23/2022]
Abstract
Although the exact etiology of inflammatory bowel disease (IBD) remains unclear, exaggerated immune response in genetically predisposed individuals has been reported. Th1 and Th17 cells mediate IBD development. Macrophages produce IL-12 and IL-23 that share p40 subunit encoded by IL12B gene as heteromer partner to drive Th1 and Th17 differentiation. The available animal and human data strongly support the pathogenic role of IL-12/IL-23 in IBD development and suggest that blocking p40 might be the potential strategy for IBD treatment. Furthermore, aberrant alteration of some cytokines expression via epigenetic mechanisms is involved in pathogenesis of IBD. In this study, we analyzed core promoter region of IL12B gene and investigated whether IL12B expression could be regulated through targeted epigenetic modification with gene editing technology. Transcription activator-like effectors (TALEs) are widely used in the field of genome editing and can specifically target DNA sequence in the host genome. We synthesized the TALE DNA-binding domains that target the promoter of human IL12B gene and fused it with the functional catalytic domains of epigenetic enzymes. Transient expression of these engineered enzymes demonstrated that the TALE-DNMT3A targeted the selected IL12B promoter region, induced loci-specific DNA methylation, and down-regulated IL-12B expression in various human cell lines. Collectively, our data suggested that epigenetic editing of IL12B through methylating DNA on its promoter might be developed as a potential therapeutic strategy for IBD treatment.
Collapse
Affiliation(s)
- Meng Chen
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Hua Zhu
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Yu-Juan Mao
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Nan Cao
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Ya-Li Yu
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Lian-Yun Li
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital, Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Lab of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Min Wu
- College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Mei Ye
- College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| |
Collapse
|
28
|
Kumari A, Bhawal S, Kapila S, Yadav H, Kapila R. Health-promoting role of dietary bioactive compounds through epigenetic modulations: a novel prophylactic and therapeutic approach. Crit Rev Food Sci Nutr 2020; 62:619-639. [PMID: 33081489 DOI: 10.1080/10408398.2020.1825286] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The epigenome is an overall epigenetic state of an organism, which is as important as that of the genome for normal development and functioning of an individual. Epigenetics involves heritable but reversible changes in gene expression through alterations in DNA methylation, histone modifications and regulation of non-coding RNAs in cells, without any change in the DNA sequence. Epigenetic changes are owned by various environmental factors including pollution, microbiota and diet, which have profound effects on epigenetic modifiers. The bioactive compounds present in the diet mainly include curcumin, resveratrol, catechins, quercetin, genistein, sulforaphane, epigallocatechin-3-gallate, alkaloids, vitamins, and peptides. Bioactive compounds released during fermentation by the action of microbes also have a significant effect on the host epigenome. Besides, recent studies have explored the new insights in vitamin's functions through epigenetic regulation. These bioactive compounds exert synergistic, preventive and therapeutic effects when combined as well as when used with chemotherapeutic agents. Therefore, these compounds have potential of therapeutic agents that could be used as "Epidrug" to treat many inflammatory diseases and various cancers where chemotherapy results have many side effects. In this review, the effect of diet derived bioactive compounds through epigenetic modulations on in vitro and in vivo models is discussed.
Collapse
Affiliation(s)
- Ankita Kumari
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Shalaka Bhawal
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Suman Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Hariom Yadav
- Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Rajeev Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
29
|
Wu J, Zhao Y, Wang X, Kong L, Johnston LJ, Lu L, Ma X. Dietary nutrients shape gut microbes and intestinal mucosa via epigenetic modifications. Crit Rev Food Sci Nutr 2020; 62:783-797. [PMID: 33043708 DOI: 10.1080/10408398.2020.1828813] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jianmin Wu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Ying Zhao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xian Wang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Lingchang Kong
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Lee J. Johnston
- West Central Research & Outreach Centre, University of Minnesota, Morris, Minnesota, USA
| | - Lin Lu
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
30
|
Cuna A, Yu W, Menden H, Feng L, Srinivasan P, Chavez-Bueno S, Ahmed I, Umar S, Sampath V. NEC-like intestinal injury is ameliorated by Lactobacillus rhamnosus GG in parallel with SIGIRR and A20 induction in neonatal mice. Pediatr Res 2020; 88:546-555. [PMID: 32053825 PMCID: PMC8213439 DOI: 10.1038/s41390-020-0797-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 01/13/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Exaggerated Toll-like receptor (TLR) signaling and intestinal dysbiosis are key contributors to necrotizing enterocolitis (NEC). Lactobacillus rhamnosus GG (LGG) decreases NEC in preterm infants, but underlying mechanisms of protection remain poorly understood. We hypothesized that LGG alleviates dysbiosis and upregulates TLR inhibitors to protect against TLR-mediated gut injury. METHODS Effects of LGG (low- and high-dose) on intestinal pro-inflammatory TLR signaling and injury in neonatal mice subjected to formula feeding (FF) and NEC were determined. 16S sequencing of stool and expression of anti-TLR mediators SIGIRR (single immunoglobulin interleukin-1-related receptor) and A20 were analyzed. RESULTS FF induced mild intestinal injury with increased expression of interleukin-1β (IL-1β) and Kupffer cell (KC) (mouse homolog of IL-8) compared to controls. LGG decreased IL-1β and KC in association with attenuated TLR signaling and increased SIGIRR and A20 expression in a dose-dependent manner. Low- and high-dose LGG had varying effects on gut microbiome despite both doses providing gut protection. Subsequent experiments of LGG on NEC revealed that pro-inflammatory TLR signaling and intestinal injury were also decreased, and SIGIRR and A20 expression increased, in a dose-dependent manner with LGG pre-treatment. CONCLUSIONS LGG protects against intestinal TLR-mediated injury by upregulating TLR inhibitors without major changes in gut microbiome composition.
Collapse
Affiliation(s)
- Alain Cuna
- University of Missouri-Kansas City, Kansas City MO,Children’s Mercy Kansas City, Kansas City MO
| | - Wei Yu
- Children’s Mercy Kansas City, Kansas City MO
| | | | - Linda Feng
- Children’s Mercy Kansas City, Kansas City MO
| | | | - Susana Chavez-Bueno
- University of Missouri-Kansas City, Kansas City MO,Children’s Mercy Kansas City, Kansas City MO
| | - Ishfaq Ahmed
- University of Kansas Medical Center, Kansas City KS
| | - Shahid Umar
- University of Kansas Medical Center, Kansas City KS
| | - Venkatesh Sampath
- University of Missouri-Kansas City, Kansas City, MO, USA. .,Children's Mercy Kansas City, Kansas City, MO, USA.
| |
Collapse
|
31
|
Nataraj BH, Ali SA, Behare PV, Yadav H. Postbiotics-parabiotics: the new horizons in microbial biotherapy and functional foods. Microb Cell Fact 2020; 19:168. [PMID: 32819443 PMCID: PMC7441679 DOI: 10.1186/s12934-020-01426-w] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 08/13/2020] [Indexed: 12/20/2022] Open
Abstract
Probiotics have several health benefits by modulating gut microbiome; however, techno-functional limitations such as viability controls have hampered their full potential applications in the food and pharmaceutical sectors. Therefore, the focus is gradually shifting from viable probiotic bacteria towards non-viable paraprobiotics and/or probiotics derived biomolecules, so-called postbiotics. Paraprobiotics and postbiotics are the emerging concepts in the functional foods field because they impart an array of health-promoting properties. Although, these terms are not well defined, however, for time being these terms have been defined as here. The postbiotics are the complex mixture of metabolic products secreted by probiotics in cell-free supernatants such as enzymes, secreted proteins, short chain fatty acids, vitamins, secreted biosurfactants, amino acids, peptides, organic acids, etc. While, the paraprobiotics are the inactivated microbial cells of probiotics (intact or ruptured containing cell components such as peptidoglycans, teichoic acids, surface proteins, etc.) or crude cell extracts (i.e. with complex chemical composition)". However, in many instances postbiotics have been used for whole category of postbiotics and parabiotics. These elicit several advantages over probiotics like; (i) availability in their pure form, (ii) ease in production and storage, (iii) availability of production process for industrial-scale-up, (iv) specific mechanism of action, (v) better accessibility of Microbes Associated Molecular Pattern (MAMP) during recognition and interaction with Pattern Recognition Receptors (PRR) and (vi) more likely to trigger only the targeted responses by specific ligand-receptor interactions. The current review comprehensively summarizes and discussed various methodologies implied to extract, purify, and identification of paraprobiotic and postbiotic compounds and their potential health benefits.
Collapse
Affiliation(s)
- Basavaprabhu H Nataraj
- Technofunctional Starters Lab, National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Syed Azmal Ali
- Proteomics and Cell Biology Lab, Animal Biotechnology Center, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India
| | - Pradip V Behare
- Technofunctional Starters Lab, National Collection of Dairy Cultures (NCDC), Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, Haryana, 132001, India.
| | - Hariom Yadav
- Department of Internal Medicine-Molecular Medicine and Microbiology and Immunology, Wake Forest School of Medicine, Biotech Place, Room 2E-034, 575 North Patterson Ave, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
32
|
Leccese G, Bibi A, Mazza S, Facciotti F, Caprioli F, Landini P, Paroni M. Probiotic Lactobacillus and Bifidobacterium Strains Counteract Adherent-Invasive Escherichia coli (AIEC) Virulence and Hamper IL-23/Th17 Axis in Ulcerative Colitis, but Not in Crohn's Disease. Cells 2020; 9:cells9081824. [PMID: 32752244 PMCID: PMC7464949 DOI: 10.3390/cells9081824] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Hypersecretion of proinflammatory cytokines and dysregulated activation of the IL-23/Th17 axis in response to intestinal microbiota dysbiosis are key factors in the pathogenesis of inflammatory bowel diseases (IBD). In this work, we studied how Lactobacillus and Bifidobacterium strains affect AIEC-LF82 virulence mechanisms and the consequent inflammatory response linked to the CCR6–CCL20 and IL-23/Th17 axes in Crohn’s disease (CD) and ulcerative colitis (UC) patients. All Lactobacillus and Bifidobacterium strains significantly reduced the LF82 adhesion and persistence within HT29 intestinal epithelial cells, inhibiting IL-8 secretion while not affecting the CCR6–CCL20 axis. Moreover, they significantly reduced LF82 survival within macrophages and dendritic cells, reducing the secretion of polarizing cytokines related to the IL-23/Th17 axis, both in healthy donors (HD) and UC patients. In CD patients, however, only B. breve Bbr8 strain was able to slightly reduce the LF82 persistence within dendritic cells, thus hampering the IL-23/Th17 axis. In addition, probiotic strains were able to modulate the AIEC-induced inflammation in HD, reducing TNF-α and increasing IL-10 secretion by macrophages, but failed to do so in IBD patients. Interestingly, the probiotic strains studied in this work were all able to interfere with the IL-23/Th17 axis in UC patients, but not in CD patients. The different interaction mechanisms of probiotic strains with innate immune cells from UC and CD patients compared to HD suggest that testing on CD-derived immune cells may be pivotal for the identification of novel probiotic strains that could be effective also for CD patients.
Collapse
Affiliation(s)
- Gabriella Leccese
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.L.); (A.B.); (P.L.)
| | - Alessia Bibi
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.L.); (A.B.); (P.L.)
| | - Stefano Mazza
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.M.); (F.C.)
| | - Federica Facciotti
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, 20139 Milan, Italy;
| | - Flavio Caprioli
- Gastroenterology and Endoscopy Unit, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.M.); (F.C.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20135 Milan, Italy
| | - Paolo Landini
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.L.); (A.B.); (P.L.)
| | - Moira Paroni
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy; (G.L.); (A.B.); (P.L.)
- Correspondence:
| |
Collapse
|
33
|
Gut microbiota modulation: a novel strategy for prevention and treatment of colorectal cancer. Oncogene 2020; 39:4925-4943. [PMID: 32514151 PMCID: PMC7314664 DOI: 10.1038/s41388-020-1341-1] [Citation(s) in RCA: 331] [Impact Index Per Article: 82.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/17/2020] [Accepted: 05/27/2020] [Indexed: 02/08/2023]
Abstract
Research about the role of gut microbiome in colorectal cancer (CRC) is a newly emerging field of study. Gut microbiota modulation, with the aim to reverse established microbial dysbiosis, is a novel strategy for prevention and treatment of CRC. Different strategies including probiotics, prebiotics, postbiotics, antibiotics, and fecal microbiota transplantation (FMT) have been employed. Although these strategies show promising results, mechanistically by correcting microbiota composition, modulating innate immune system, enhancing gut barrier function, preventing pathogen colonization and exerting selective cytotoxicity against tumor cells, it should be noted that they are accompanied by risks and controversies that can potentially introduce clinical complications. During bench-to-bedside translation, evaluation of risk-and-benefit ratio, as well as patient selection, should be carefully performed. In view of the individualized host response to gut microbiome intervention, developing personalized microbiome therapy may be the key to successful clinical treatment.
Collapse
|
34
|
Evans LW, Athukorala M, Martinez-Guryn K, Ferguson BS. The Role of Histone Acetylation and the Microbiome in Phytochemical Efficacy for Cardiovascular Diseases. Int J Mol Sci 2020; 21:E4006. [PMID: 32503339 PMCID: PMC7313062 DOI: 10.3390/ijms21114006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/12/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases (CVD) are the main cause of death worldwide and create a substantial financial burden. Emerging studies have begun to focus on epigenetic targets and re-establishing healthy gut microbes as therapeutic options for the treatment and prevention of CVD. Phytochemicals, commonly found in fruits and vegetables, have been shown to exert a protective effect against CVD, though their mechanisms of action remain incompletely understood. Of interest, phytochemicals such as curcumin, resveratrol and epigallocatechin gallate (EGCG) have been shown to regulate both histone acetylation and microbiome re-composition. The purpose of this review is to highlight the microbiome-epigenome axis as a therapeutic target for food bioactives in the prevention and/or treatment of CVD. Specifically, we will discuss studies that highlight how the three phytochemicals above alter histone acetylation leading to global changes in gene expression and CVD protection. Then, we will expand upon these phytochemicals to discuss the impact of phytochemical-microbiome-histone acetylation interaction in CVD.
Collapse
Affiliation(s)
- Levi W. Evans
- Department of Nutrition, University of Nevada Reno, Reno, NV 89557, USA; (L.W.E.); (M.A.)
| | - Maheshi Athukorala
- Department of Nutrition, University of Nevada Reno, Reno, NV 89557, USA; (L.W.E.); (M.A.)
| | | | - Bradley S. Ferguson
- Department of Nutrition, University of Nevada Reno, Reno, NV 89557, USA; (L.W.E.); (M.A.)
- Center of Biomedical Research Excellence for Molecular and Cellular Signal Transduction in the Cardiovascular System, University of Nevada Reno, Reno, NV 89557, USA
| |
Collapse
|
35
|
Bannister S, Messina NL, Novakovic B, Curtis N. The emerging role of epigenetics in the immune response to vaccination and infection: a systematic review. Epigenetics 2020; 15:555-593. [PMID: 31914857 PMCID: PMC7574386 DOI: 10.1080/15592294.2020.1712814] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
Extensive research has highlighted the role of infection-induced epigenetic events in the development of cancer. More recently, attention has focused on the ability of non-carcinogenic infections, as well as vaccines, to modify the human epigenome and modulate the immune response. This review explores this rapidly evolving area of investigation and outlines the many and varied ways in which vaccination and natural infection can influence the human epigenome from modulation of the innate and adaptive immune response, to biological ageing and modification of disease risk. The implications of these epigenetic changes on immune regulation and their potential application to the diagnosis and treatment of chronic infection and vaccine development are also discussed.
Collapse
Affiliation(s)
- Samantha Bannister
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Infectious Diseases Unit, Royal Children’s Hospital Melbourne, Parkville, Australia
| | - Nicole L. Messina
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
| | - Boris Novakovic
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Epigenetics Research Group, Murdoch Children’s Research Institute, Parkville, Australia
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne, Parkville, Australia
- Infectious Diseases Research Group, Murdoch Children’s Research Institute, Parkville, Australia
- Infectious Diseases Unit, Royal Children’s Hospital Melbourne, Parkville, Australia
| |
Collapse
|
36
|
Evans LW, Stratton MS, Ferguson BS. Dietary natural products as epigenetic modifiers in aging-associated inflammation and disease. Nat Prod Rep 2020; 37:653-676. [PMID: 31993614 PMCID: PMC7577396 DOI: 10.1039/c9np00057g] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Covering: up to 2020Chronic, low-grade inflammation is linked to aging and has been termed "inflammaging". Inflammaging is considered a key contributor to the development of metabolic dysfunction and a broad spectrum of diseases or disorders including declines in brain and heart function. Genome-wide association studies (GWAS) coupled with epigenome-wide association studies (EWAS) have shown the importance of diet in the development of chronic and age-related diseases. Moreover, dietary interventions e.g. caloric restriction can attenuate inflammation to delay and/or prevent these diseases. Common themes in these studies entail the use of phytochemicals (plant-derived compounds) or the production of short chain fatty acids (SCFAs) as epigenetic modifiers of DNA and histone proteins. Epigenetic modifications are dynamically regulated and as such, serve as potential therapeutic targets for the treatment or prevention of age-related disease. In this review, we will focus on the role for natural products that include phytochemicals and short chain fatty acids (SCFAs) as regulators of these epigenetic adaptations. Specifically, we discuss regulators of methylation, acetylation and acylation, in the protection from chronic inflammation driven metabolic dysfunction and deterioration of neurocognitive and cardiac function.
Collapse
Affiliation(s)
- Levi W Evans
- Department of Nutrition, University of Nevada, Reno, NV 89557, USA.
| | | | | |
Collapse
|
37
|
Jeffrey MP, MacPherson CW, Mathieu O, Tompkins TA, Green-Johnson JM. Secretome-Mediated Interactions with Intestinal Epithelial Cells: A Role for Secretome Components from Lactobacillus rhamnosus R0011 in the Attenuation of Salmonella enterica Serovar Typhimurium Secretome and TNF-α-Induced Proinflammatory Responses. THE JOURNAL OF IMMUNOLOGY 2020; 204:2523-2534. [PMID: 32238458 DOI: 10.4049/jimmunol.1901440] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
Recent evidence suggests that lactic acid bacteria communicate with host cells via secretome components to influence immune responses but less is known about gut-pathogen secretomes, impact of lactic acid bacteria secretomes on host-pathogen interactions, and the mechanisms underlying these interactions. Genome-wide microarrays and cytokine profiling were used to interrogate the impact of the Lactobacillus rhamnosus R0011 secretome (LrS) on TNF-α and Salmonella enterica subsp. enterica serovar Typhimurium secretome (STS)-induced outcomes in human intestinal epithelial cells. The LrS attenuated both TNF-α- and STS-induced gene expression involved in NF-κB and MAPK activation, as well as expression of genes involved in other immune-related signaling pathways. Specifically, the LrS induced the expression of dual specificity phosphatase 1 (DUSP1), activating transcription factor 3 (ATF3), and tribbles pseudokinase 3 (TRIB3), negative regulators of innate immune signaling, in HT-29 intestinal epithelial cells challenged with TNF-α or STS. TNF-α- and STS-induced acetylation of H3 and H4 histones was attenuated by the LrS, as was the production of TNF-α- and STS-induced proinflammatory cytokines and chemokines. Interestingly, the LrS induced production of macrophage migration inhibitory factor (MIF), a cytokine involved in host-microbe interactions at the gut interface. We propose that the LrS attenuates proinflammatory mediator expression through increased transcription of negative regulators of innate immune activity and changes in global H3 and H4 histone acetylation. To our knowledge, these findings provide novel insights into the complex multifaceted mechanisms of action behind secretome-mediated interdomain communication at the gut-mucosal interface.
Collapse
Affiliation(s)
- Michael P Jeffrey
- Applied Bioscience Graduate Program and the Faculty of Science, Ontario Technical University, Oshawa, Ontario L1G 0C5, Canada; and
| | - Chad W MacPherson
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec H4P 2R2, Canada
| | - Olivier Mathieu
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec H4P 2R2, Canada
| | - Thomas A Tompkins
- Rosell Institute for Microbiome and Probiotics, Montreal, Quebec H4P 2R2, Canada
| | - Julia M Green-Johnson
- Applied Bioscience Graduate Program and the Faculty of Science, Ontario Technical University, Oshawa, Ontario L1G 0C5, Canada; and
| |
Collapse
|
38
|
DeKuiper JL, Coussens PM. Inflammatory Th17 responses to infection with Mycobacterium avium subspecies paratuberculosis (MAP) in cattle and their potential role in development of Johne's disease. Vet Immunol Immunopathol 2019; 218:109954. [PMID: 31733610 DOI: 10.1016/j.vetimm.2019.109954] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/24/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
Chronic intestinal inflammation typically associated with late stage Johne's disease (JD) in cattle occurs despite a lack of significant expression of the typical proinflammatory cytokines IFNγ and TNFα derived from Th1- like T cells. In contrast, these cytokines appear to be relatively abundant during early infections with Mycobacterium avium subspecies paratuberculosis (MAP), the causative agent of JD in cattle. The roles of non-classical immune responses, such as those associated with Th17 cells, in response to MAP infection and development of clinical JD are less clear. In this review, we examine literature suggesting that Mycobacterial infections, including Mycobacterium tuberculosis, Mycobacterium bovis, and MAP, are all associated with expression of Th17 promoting cytokines (IL-23, IL-22, IL-17a). We discuss the possibility that Th17 associated cytokines, particularly IL-23, may act as contributing factors in development and maintenance of inflammation characteristic of clinical JD. An as yet relatively unexplored source of chronic inflammation due to over expression of IL-1α and IL-1β is also presented. We further discuss the fact that, as with the typical Th1-like cytokines IFNγ and TNFα , IL-17a is not significantly expressed in CD4+ T cells from cows with clinical JD, possibly due to T cell exhaustion. Finally, we present the notion that the Th17 driving cytokine IL-23 expressed by infected macrophages and associated epithelial cells may contribute to chronic inflammation during later stages of JD.
Collapse
Affiliation(s)
- Justin L DeKuiper
- Michigan State University, 3385A Anthony Hall, 474 S. Shaw Lane, 48824, East Lansing, MI, United States
| | - Paul M Coussens
- Michigan State University, 3385A Anthony Hall, 474 S. Shaw Lane, 48824, East Lansing, MI, United States.
| |
Collapse
|
39
|
Bifidobacteria and Mucosal-Associated Invariant T (MAIT) Cells: A New Approach to Colorectal Cancer Prevention? GASTROINTESTINAL DISORDERS 2019. [DOI: 10.3390/gidisord1020022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer is the most preventable form of cancer worldwide. The pathogenesis of colorectal cancer includes gut inflammation, genetic and microbial composition factors. İmpairment of the gut microbiota has been associated with development of colorectal cancer. The genus Bifidobacterium is an important component of the commensal gut microbiota. Bifidobacteria are considered to have important roles in multiple homeostatic functions: immunologic, hormonal and metabolic. Mucosal-associated invariant T cells (MAIT) are components of the immune system involved in protection against infectious pathogens and regulate the pathogenesis of various inflammatory diseases and, potentially, colorectal cancer. Engagement between Bifidobacterium and MAIT cells could exert a beneficial effect on colorectal cancer prevention and treatment.
Collapse
|
40
|
Dovrolis N, Filidou E, Kolios G. Systems biology in inflammatory bowel diseases: on the way to precision medicine. Ann Gastroenterol 2019; 32:233-246. [PMID: 31040620 PMCID: PMC6479645 DOI: 10.20524/aog.2019.0373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel diseases (IBD) are chronic and recurrent inflammatory disorders of the gastrointestinal tract. The elucidation of their etiopathology requires complex and multiple approaches. Systems biology has come to fulfill this need in approaching the pathogenetic mechanisms of IBD and its etiopathology, in a comprehensive way, by combining data from different scientific sources. In combination with bioinformatics and network medicine, it uses principles from computer science, mathematics, physics, chemistry, biology, medicine and computational tools to achieve its purposes. Systems biology utilizes scientific sources that provide data from omics studies (e.g., genomics, transcriptomics, etc.) and clinical observations, whose combined analysis leads to network formation and ultimately to a more integrative image of disease etiopathogenesis. In this review, we analyze the current literature on the methods and the tools utilized by systems biology in order to cover an innovative and exciting field: IBD-omics.
Collapse
Affiliation(s)
- Nikolas Dovrolis
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Eirini Filidou
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Kolios
- Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
- Correspondence to: Prof. George Kolios, MD PhD, Laboratory of Pharmacology, Faculty of Medicine, Democritus University of Thrace, Dragana, Alexandroupolis, 68100, Greece, e-mail:
| |
Collapse
|
41
|
Yacon (Smallanthus sonchifolius)-based product increases fecal short-chain fatty acids and enhances regulatory T cells by downregulating RORγt in the colon of BALB/c mice. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.02.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
42
|
Probiotic lactobacilli mediated changes in global epigenetic signatures of human intestinal epithelial cells during Escherichia coli challenge. ANN MICROBIOL 2019. [DOI: 10.1007/s13213-019-01451-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
43
|
Alagón Fernández Del Campo P, De Orta Pando A, Straface JI, López Vega JR, Toledo Plata D, Niezen Lugo SF, Alvarez Hernández D, Barrientos Fortes T, Gutiérrez-Kobeh L, Solano-Gálvez SG, Vázquez-López R. The Use of Probiotic Therapy to Modulate the Gut Microbiota and Dendritic Cell Responses in Inflammatory Bowel Diseases. ACTA ACUST UNITED AC 2019; 7:medsci7020033. [PMID: 30813381 PMCID: PMC6410300 DOI: 10.3390/medsci7020033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/28/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022]
Abstract
Recent investigations have shown that different conditions such as diet, the overuse of antibiotics or the colonization of pathogenic microorganisms can alter the population status of the intestinal microbiota. This modification can produce a change from homeostasis to a condition known as imbalance or dysbiosis; however, the role-played by dysbiosis and the development of inflammatory bowel diseases (IBD) has been poorly understood. It was actually not until a few years ago that studies started to develop regarding the role that dendritic cells (DC) of intestinal mucosa play in the sensing of the gut microbiota population. The latest studies have focused on describing the DC modulation, specifically on tolerance response involving T regulatory cells or on the inflammatory response involving reactive oxygen species and tissue damage. Furthermore, the latest studies have also focused on the protective and restorative effect of the population of the gut microbiota given by probiotic therapy, targeting IBD and other intestinal pathologies. In the present work, the authors propose and summarize a recently studied complex axis of interaction between the population of the gut microbiota, the sensing of the DC and its modulation towards tolerance and inflammation, the development of IBD and the protective and restorative effect of probiotics on other intestinal pathologies.
Collapse
Affiliation(s)
- Pablo Alagón Fernández Del Campo
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - Alejandro De Orta Pando
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - Juan Ignacio Straface
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - José Ricardo López Vega
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - Diego Toledo Plata
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - Sebastian Felipe Niezen Lugo
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - Diego Alvarez Hernández
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| | - Tomás Barrientos Fortes
- Director Facultad de Ciencias de la Salud, Universidad Anáhuac México, 52786 Cuidad de México, Mexico.
| | - Laila Gutiérrez-Kobeh
- Unidad de Investigación UNAM-INC, División Investigación, Facultad de Medicina, Universidad Nacional Autónoma de México-Instituto Nacional de Cardiología "Ignacio Chávez," Mexico City 14080, Mexico.
| | - Sandra Georgina Solano-Gálvez
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico.
| | - Rosalino Vázquez-López
- Departamento de Microbiología del Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México Campus Norte, 52786 Cuidad de México, Mexico.
| |
Collapse
|
44
|
Berni Canani R, Paparo L, Nocerino R, Di Scala C, Della Gatta G, Maddalena Y, Buono A, Bruno C, Voto L, Ercolini D. Gut Microbiome as Target for Innovative Strategies Against Food Allergy. Front Immunol 2019; 10:191. [PMID: 30828329 PMCID: PMC6384262 DOI: 10.3389/fimmu.2019.00191] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
The dramatic increase in food allergy prevalence and severity globally requires effective strategies. Food allergy derives from a defect in immune tolerance mechanisms. Immune tolerance is modulated by gut microbiota function and structure, and microbiome alterations (dysbiosis) have a pivotal role in the development of food allergy. Environmental factors, including a low-fiber/high-fat diet, cesarean delivery, antiseptic agents, lack of breastfeeding, and drugs can induce gut microbiome dysbiosis, and have been associated with food allergy. New experimental tools and technologies have provided information regarding the role of metabolites generated from dietary nutrients and selected probiotic strains that could act on immune tolerance mechanisms. The mechanisms are multiple and still not completely defined. Increasing evidence has provided useful information on optimal bacterial species/strains, dosage, and timing for intervention. The increased knowledge of the crucial role played by nutrients and gut microbiota-derived metabolites is opening the way to a post-biotic approach in the stimulation of immune tolerance through epigenetic regulation. This review focused on the potential role of gut microbiome as the target for innovative strategies against food allergy.
Collapse
Affiliation(s)
- Roberto Berni Canani
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- European Laboratory for the Investigation of Food-Induced Diseases, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
- Task Force on Microbiome Studies, University of Naples “Federico II”, Naples, Italy
| | - Lorella Paparo
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Rita Nocerino
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Carmen Di Scala
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Giusy Della Gatta
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Ylenia Maddalena
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
| | - Aniello Buono
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Cristina Bruno
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
- ImmunoNutritionLab at CEINGE-Advanced Biotechnologies, University of Naples “Federico II”, Naples, Italy
| | - Luana Voto
- Department of Translational Medical Science – Pediatric Section, University of Naples “Federico II”, Naples, Italy
| | - Danilo Ercolini
- Task Force on Microbiome Studies, University of Naples “Federico II”, Naples, Italy
- Department of Agricultural Sciences, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
45
|
Bozkurt HS, Quigley EM, Kara B. Bifidobacterium animalis subspecies lactis engineered to produce mycosporin-like amino acids in colorectal cancer prevention. SAGE Open Med 2019; 7:2050312119825784. [PMID: 30719295 PMCID: PMC6348500 DOI: 10.1177/2050312119825784] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 12/31/2018] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer is the third most common cancer and the third leading cause of cancer-related death. The pathogensesis of colorectal cancer involves a multi-step and multi-factorial process. Disruption of the gut microbiota has been associated with gastrointestinal diseases such as colorectal cancer. The genus Bifidobacterium is considered an important component of the commensal microbiota and plays important roles in several homeostatic functions: immune, neurohormonal, and metabolic. Bifidobacterium animalis subsp. lactis is a well-documented probiotic within the species Bifidobacterium. Mycosporin-like amino acids are low molecular weight amino acids demonstrated to exert prebiotic effects and to modulate host immunity by regulating the proliferation and differentiation of intestinal epithelial cells, macrophages and lymphocytes, as well as cytokine production.Their modulation of the metabolism of the immune system and transcription factors could exert a beneficial effect on colorectal cancer. B. animalis does not produce mycosporin-like amino acids. If one could create a B. animalis–producing mycosporin-like amino acids via genetic open reading frame engineering it should exert more potent immuno-stimulatory properties and, thereby, become a potent strain-specific microbial based therapy in colorectal cancer prevention.
Collapse
Affiliation(s)
| | - Eamonn Mm Quigley
- Division of Gastroenterology and Hepatology, Lynda K and David M Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA
| | - Banu Kara
- Clinic of Gastroenterology, Adana Numune Research and Education Hospital, University of Health Sciences, Adana, Turkey
| |
Collapse
|
46
|
Paparo L, Nocerino R, Di Scala C, Della Gatta G, Di Costanzo M, Buono A, Bruno C, Berni Canani R. Targeting Food Allergy with Probiotics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1125:57-68. [DOI: 10.1007/5584_2018_316] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
47
|
Commensal lactic acid-producing bacteria affect host cellular lipid metabolism through various cellular metabolic pathways: Role of mTOR, FOXO1, and autophagy machinery system. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2018.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
48
|
Immunomodulatory effects of probiotics: Can they be used to treat allergies and autoimmune diseases? Maturitas 2018; 119:25-38. [PMID: 30502748 DOI: 10.1016/j.maturitas.2018.11.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/06/2018] [Accepted: 11/08/2018] [Indexed: 12/12/2022]
Abstract
As a person ages, physiological, immunological and gut microbiome changes collectively result in an array of chronic conditions. According to the 'hygiene hypothesis' the increasing prevalence of immune-mediated disorders may be related to intestinal dysbiosis, leading to immune dysfunction and associated conditions such as eczema, asthma, allergies and autoimmune diseases. Beneficial probiotic bacteria can be utilized by increasing their abundance within the gastrointestinal lumen, which in turn will modulate immune cells, such as, T helper (Th)-1, Th2, Th17, regulatory T (Treg) cells and B cells, which have direct relevance to human health and the pathogenesis of immune disorders. Here, we describe the cross-talk between probiotics and the gastrointestinal immune system, and their effects in relation to inflammatory bowel disease, multiple sclerosis, allergies and atopic dermatitis.
Collapse
|
49
|
Mardani F, Mahmoudi M, Esmaeili SA, Khorasani S, Tabasi N, Rastin M. In vivo study: Th1-Th17 reduction in pristane-induced systemic lupus erythematosus mice after treatment with tolerogenic Lactobacillus probiotics. J Cell Physiol 2018; 234:642-649. [PMID: 30078223 DOI: 10.1002/jcp.26819] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2018] [Accepted: 04/30/2018] [Indexed: 12/17/2022]
Abstract
Uncontrolled inflammation in systemic lupus erythematosus (SLE) could cause dysfunction in multiple organs. T helper 17 (Th17) cells are a main branch of inflammatory responses in the pathogenesis of SLE, and by producing interleukin 17 (IL-17), represent a major functional tool in the progression of inflammation. Animal models provide a special field for better studies of the pathogenesis of diseases. Tolergenic probiotics could decrease inflammation in autoimmune diseases by modulating the immune system and maintaining homeostasis. The aim of this project was to evaluate the effects of Lactobacillus rhamnosus and Lactobacillus delbrueckii on Th17 cells and their related mediators in a pristane-induced BALB/c mice model of SLE. The mice were divided into pretreatment groups, which received probiotics or prednisolone at Day 0, and treatment groups, which received probiotics and prednisolone 2 months after injection. The presence of antinuclear antibody (ANA), anti-double-stranded DNA (anti-dsDNA), and anti-ribonucleoprotein (anti-RNP) and lipogranuloma was evaluated; also, the population of Th1-Th17 cells as well as interferon γ (IFN-γ), IL-17, and IL-10 levels, and the expression of RAR-related orphan related receptor gamma (RORγt) and IL-17 were determined. We observed that probiotics and prednisolone could delay SLE in pretreatment and treatment mice groups, with a reduction in ANA, anti-dsDNA, anti-RNP, and mass of lipogranuloma. Probiotics and prednisolone decreased the population of Th1-Th17 cells and reduced IFN-γ and IL-17 as inflammatory cytokines in the pretreatment and treatment groups in comparison with SLE-induced mice. Our results indicated that, due to their anti-inflammatory properties and reduction of Th17, Th1, and cytotoxic T lymphocyte (CTL) cells, the use of these probiotics could probably represent a new tool for the better management of SLE.
Collapse
Affiliation(s)
- Fatemeh Mardani
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committeen, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed-Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committeen, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Khorasani
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committeen, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nafiseh Tabasi
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Rastin
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Department, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
50
|
Esmaeili S, Mahmoudi M, Rezaieyazdi Z, Sahebari M, Tabasi N, Sahebkar A, Rastin M. Generation of tolerogenic dendritic cells using
Lactobacillus rhamnosus
and
Lactobacillus delbrueckii
as tolerogenic probiotics. J Cell Biochem 2018; 119:7865-7872. [DOI: 10.1002/jcb.27203] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/24/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Seyed‐Alireza Esmaeili
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
- Student Research Committee, Mashhad University of Medical Sciences Mashhad Iran
| | - Mahmoud Mahmoudi
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | - Zahra Rezaieyazdi
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Sahebari
- Rheumatic Diseases Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Nafiseh Tabasi
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
| | - Amirhossein Sahebkar
- Neurogenic inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| | - Maryam Rastin
- Immunology Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Immunology Department Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|