1
|
Borrego-Ruiz A, Borrego JJ. Epigenetic Mechanisms in Aging: Extrinsic Factors and Gut Microbiome. Genes (Basel) 2024; 15:1599. [PMID: 39766866 PMCID: PMC11675900 DOI: 10.3390/genes15121599] [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: 11/23/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Aging is a natural physiological process involving biological and genetic pathways. Growing evidence suggests that alterations in the epigenome during aging result in transcriptional changes, which play a significant role in the onset of age-related diseases, including cancer, cardiovascular disease, diabetes, and neurodegenerative disorders. For this reason, the epigenetic alterations in aging and age-related diseases have been reviewed, and the major extrinsic factors influencing these epigenetic alterations have been identified. In addition, the role of the gut microbiome and its metabolites as epigenetic modifiers has been addressed. RESULTS Long-term exposure to extrinsic factors such as air pollution, diet, drug use, environmental chemicals, microbial infections, physical activity, radiation, and stress provoke epigenetic changes in the host through several endocrine and immune pathways, potentially accelerating the aging process. Diverse studies have reported that the gut microbiome plays a critical role in regulating brain cell functions through DNA methylation and histone modifications. The interaction between genes and the gut microbiome serves as a source of adaptive variation, contributing to phenotypic plasticity. However, the molecular mechanisms and signaling pathways driving this process are still not fully understood. CONCLUSIONS Extrinsic factors are potential inducers of epigenetic alterations, which may have important implications for longevity. The gut microbiome serves as an epigenetic effector influencing host gene expression through histone and DNA modifications, while bidirectional interactions with the host and the underexplored roles of microbial metabolites and non-bacterial microorganisms such as fungi and viruses highlight the need for further research.
Collapse
Affiliation(s)
- Alejandro Borrego-Ruiz
- Departamento de Psicología Social y de las Organizaciones, Universidad Nacional de Educación a Distancia (UNED), 28040 Madrid, Spain;
| | - Juan J. Borrego
- Departamento de Microbiología, Universidad de Málaga, 29071 Málaga, Spain
| |
Collapse
|
2
|
Pepke ML, Hansen SB, Limborg MT. Unraveling host regulation of gut microbiota through the epigenome-microbiome axis. Trends Microbiol 2024; 32:1229-1240. [PMID: 38839511 DOI: 10.1016/j.tim.2024.05.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
Recent studies of dynamic interactions between epigenetic modifications of a host organism and the composition or activity of its associated gut microbiota suggest an opportunity for the host to shape its microbiome through epigenetic alterations that lead to changes in gene expression and noncoding RNA activity. We use insights from microbiota-induced epigenetic changes to review the potential of the host to epigenetically regulate its gut microbiome, from which a bidirectional 'epigenome-microbiome axis' emerges. This axis embeds environmentally induced variation, which may influence the adaptive evolution of host-microbe interactions. We furthermore present our perspective on how the epigenome-microbiome axis can be understood and investigated within a holo-omic framework with potential applications in the applied health and food sciences.
Collapse
Affiliation(s)
- Michael L Pepke
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| | - Søren B Hansen
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark
| | - Morten T Limborg
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| |
Collapse
|
3
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
4
|
Holuka C, Grova N, Charalambous EG, Le Cléac H J, Turner JD, Mposhi A. Transgenerational impacts of early life adversity: from health determinants, implications to epigenetic consequences. Neurosci Biobehav Rev 2024; 164:105785. [PMID: 38945418 DOI: 10.1016/j.neubiorev.2024.105785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/09/2024] [Accepted: 06/23/2024] [Indexed: 07/02/2024]
Abstract
Exposure to different environmental factors, social and socioeconomic factors promotes development of the early-life adversity (ELA) phenotype. The persistence of this phenotype across generations is an interesting phenomenon that remains unexplored. Of late many studies have focused on disease-associated outcomes of ELA following exposure during childhood but the persistence of epigenetic imprints transmitted by ELA exposed parents to their offspring remains poorly described. It is possible that both parents are able to transmit ELA-associated genetic imprints to their offspring via transgenerational inheritance mechanisms. Here, we highlight the role of the mother and father in the biological process of conception, from epigenetic reprogramming cycles to later environmental exposures. We explain some of the known determinants of ELA (pollution, socioeconomic challenges, infections, etc.) and their disease-associated outcomes. Finally, we highlight the role of epigenetics, mitochondria and ncRNAs as mechanisms mediating transgenerational inheritance. Whether these transgenerational inheritance mechanisms occur in the human context remains unclear but there is a large body of suggestive evidence in non-human models that points out to its existence.
Collapse
Affiliation(s)
- Cyrielle Holuka
- Immune Endocrine Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg; Faculty of Science, University of Luxembourg, Belval L-4365, Luxembourg
| | - Nathalie Grova
- Immune Endocrine Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg; UMR Inserm 1256 nGERE, Nutrition-Génétique et exposition aux risques environnementaux, Institute of Medical Research (Pôle BMS) - University of Lorraine, B.P. 184, Nancy 54511, France
| | - Eleftheria G Charalambous
- Department of Psychiatry and Psychotherapy, University Medecine Greifswald, Ellernholzstr. 1-2, Greifswald 17489, Germany; Department of Psychology, University of Cyprus, Nicosia 2109, Cyprus
| | - Jeanne Le Cléac H
- Immune Endocrine Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg; Faculty of Science, University of Luxembourg, Belval L-4365, Luxembourg
| | - Jonathan D Turner
- Immune Endocrine Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg.
| | - Archibold Mposhi
- Immune Endocrine Epigenetics Research Group, Department of Infection and Immunity, Luxembourg Institute of Health, Luxembourg
| |
Collapse
|
5
|
Jacob T, Sindhu S, Hasan A, Malik MZ, Arefanian H, Al-Rashed F, Nizam R, Kochumon S, Thomas R, Bahman F, Shenouda S, Wilson A, Akther N, Al-Roub A, Abukhalaf N, Albeloushi S, Abu-Farha M, Al Madhoun A, Alzaid F, Thanaraj TA, Koistinen HA, Tuomilehto J, Al-Mulla F, Ahmad R. Soybean oil-based HFD induces gut dysbiosis that leads to steatosis, hepatic inflammation and insulin resistance in mice. Front Microbiol 2024; 15:1407258. [PMID: 39165573 PMCID: PMC11334085 DOI: 10.3389/fmicb.2024.1407258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 07/09/2024] [Indexed: 08/22/2024] Open
Abstract
High-fat diets (HFDs) shape the gut microbiome and promote obesity, inflammation, and liver steatosis. Fish and soybean are part of a healthy diet; however, the impact of these fats, in the absence of sucrose, on gut microbial dysbiosis and its association with liver steatosis remains unclear. Here, we investigated the effect of sucrose-free soybean oil-and fish oil-based high fat diets (HFDs) (SF-Soy-HFD and SF-Fish-HFD, respectively) on gut dysbiosis, obesity, steatosis, hepatic inflammation, and insulin resistance. C57BL/6 mice were fed these HFDs for 24 weeks. Both diets had comparable effects on liver and total body weights. But 16S-rRNA sequencing of the gut content revealed induction of gut dysbiosis at different taxonomic levels. The microbial communities were clearly separated, showing differential dysbiosis between the two HFDs. Compared with the SF-Fish-HFD control group, the SF-Soy-HFD group had an increased abundance of Bacteroidetes, Firmicutes, and Deferribacteres, but a lower abundance of Verrucomicrobia. The Clostridia/Bacteroidia (C/B) ratio was higher in the SF-Soy-HFD group (3.11) than in the SF-Fish-HFD group (2.5). Conversely, the Verrucomicrobiacae/S24_7 (also known as Muribaculaceae family) ratio was lower in the SF-Soy-HFD group (0.02) than that in the SF-Fish-HFD group (0.75). The SF-Soy-HFD group had a positive association with S24_7, Clostridiales, Allobaculum, Coriobacteriaceae, Adlercreutzia, Christensenellaceae, Lactococcus, and Oscillospira, but was related to a lower abundance of Akkermansia, which maintains gut barrier integrity. The gut microbiota in the SF-Soy-HFD group had predicted associations with host genes related to fatty liver and inflammatory pathways. Mice fed the SF-Soy-HFD developed liver steatosis and showed increased transcript levels of genes associated with de novo lipogenesis (Acaca, Fasn, Scd1, Elovl6) and cholesterol synthesis (Hmgcr) pathways compared to those in the SF-Fish-HFD-group. No differences were observed in the expression of fat uptake genes (Cd36 and Fabp1). The expression of the fat efflux gene (Mttp) was reduced in the SF-Soy-HFD group. Moreover, hepatic inflammation markers (Tnfa and Il1b) were notably expressed in SF-Soy-HFD-fed mice. In conclusion, SF-Soy-HFD feeding induced gut dysbiosis in mice, leading to steatosis, hepatic inflammation, and impaired glucose homeostasis.
Collapse
Affiliation(s)
- Texy Jacob
- Dasman Diabetes Institute, Dasman, Kuwait
| | | | - Amal Hasan
- Dasman Diabetes Institute, Dasman, Kuwait
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Fawaz Alzaid
- Dasman Diabetes Institute, Dasman, Kuwait
- INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| | | | - Heikki A. Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
| | | | | |
Collapse
|
6
|
Kochumon S, Malik MZ, Sindhu S, Arefanian H, Jacob T, Bahman F, Nizam R, Hasan A, Thomas R, Al-Rashed F, Shenouda S, Wilson A, Albeloushi S, Almansour N, Alhamar G, Al Madhoun A, Alzaid F, Thanaraj TA, Koistinen HA, Tuomilehto J, Al-Mulla F, Ahmad R. Gut Dysbiosis Shaped by Cocoa Butter-Based Sucrose-Free HFD Leads to Steatohepatitis, and Insulin Resistance in Mice. Nutrients 2024; 16:1929. [PMID: 38931284 PMCID: PMC11207001 DOI: 10.3390/nu16121929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND High-fat diets cause gut dysbiosis and promote triglyceride accumulation, obesity, gut permeability changes, inflammation, and insulin resistance. Both cocoa butter and fish oil are considered to be a part of healthy diets. However, their differential effects on gut microbiome perturbations in mice fed high concentrations of these fats, in the absence of sucrose, remains to be elucidated. The aim of the study was to test whether the sucrose-free cocoa butter-based high-fat diet (C-HFD) feeding in mice leads to gut dysbiosis that associates with a pathologic phenotype marked by hepatic steatosis, low-grade inflammation, perturbed glucose homeostasis, and insulin resistance, compared with control mice fed the fish oil based high-fat diet (F-HFD). RESULTS C57BL/6 mice (5-6 mice/group) were fed two types of high fat diets (C-HFD and F-HFD) for 24 weeks. No significant difference was found in the liver weight or total body weight between the two groups. The 16S rRNA sequencing of gut bacterial samples displayed gut dysbiosis in C-HFD group, with differentially-altered microbial diversity or relative abundances. Bacteroidetes, Firmicutes, and Proteobacteria were highly abundant in C-HFD group, while the Verrucomicrobia, Saccharibacteria (TM7), Actinobacteria, and Tenericutes were more abundant in F-HFD group. Other taxa in C-HFD group included the Bacteroides, Odoribacter, Sutterella, Firmicutes bacterium (AF12), Anaeroplasma, Roseburia, and Parabacteroides distasonis. An increased Firmicutes/Bacteroidetes (F/B) ratio in C-HFD group, compared with F-HFD group, indicated the gut dysbiosis. These gut bacterial changes in C-HFD group had predicted associations with fatty liver disease and with lipogenic, inflammatory, glucose metabolic, and insulin signaling pathways. Consistent with its microbiome shift, the C-HFD group showed hepatic inflammation and steatosis, high fasting blood glucose, insulin resistance, increased hepatic de novo lipogenesis (Acetyl CoA carboxylases 1 (Acaca), Fatty acid synthase (Fasn), Stearoyl-CoA desaturase-1 (Scd1), Elongation of long-chain fatty acids family member 6 (Elovl6), Peroxisome proliferator-activated receptor-gamma (Pparg) and cholesterol synthesis (β-(hydroxy β-methylglutaryl-CoA reductase (Hmgcr). Non-significant differences were observed regarding fatty acid uptake (Cluster of differentiation 36 (CD36), Fatty acid binding protein-1 (Fabp1) and efflux (ATP-binding cassette G1 (Abcg1), Microsomal TG transfer protein (Mttp) in C-HFD group, compared with F-HFD group. The C-HFD group also displayed increased gene expression of inflammatory markers including Tumor necrosis factor alpha (Tnfa), C-C motif chemokine ligand 2 (Ccl2), and Interleukin-12 (Il12), as well as a tendency for liver fibrosis. CONCLUSION These findings suggest that the sucrose-free C-HFD feeding in mice induces gut dysbiosis which associates with liver inflammation, steatosis, glucose intolerance and insulin resistance.
Collapse
Affiliation(s)
- Shihab Kochumon
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Md. Zubbair Malik
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Sardar Sindhu
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Hossein Arefanian
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Texy Jacob
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fatemah Bahman
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Rasheeba Nizam
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Amal Hasan
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Reeby Thomas
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fatema Al-Rashed
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Steve Shenouda
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ajit Wilson
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Shaima Albeloushi
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Nourah Almansour
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ghadeer Alhamar
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Ashraf Al Madhoun
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Fawaz Alzaid
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
- Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, Institut Necker Enfants Malades, F-75015 Paris, France
| | - Thangavel Alphonse Thanaraj
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Heikki A. Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, 00029 Helsinki, Finland;
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, 00271 Helsinki, Finland;
- Minerva Foundation Institute for Medical Research, 00290 Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, P.O. Box 30, 00271 Helsinki, Finland;
- Department of Public Health, University of Helsinki, 00014 Helsinki, Finland
| | - Fahd Al-Mulla
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| | - Rasheed Ahmad
- Dasman Diabetes Institute, Dasman 15462, Kuwait; (S.K.); (M.Z.M.); (S.S.); (H.A.); (T.J.); (F.B.); (R.N.); (A.H.); (R.T.); (F.A.-R.); (S.S.); (A.W.); (S.A.); (N.A.); (G.A.); (A.A.M.); (F.A.); (T.A.T.); (F.A.-M.)
| |
Collapse
|
7
|
Pirola CJ, Salatino A, Fernández Gianotti T, Castaño GO, Garaycoechea M, Sookoian S. Cross talk between the liver microbiome and epigenome in patients with metabolic dysfunction-associated steatotic liver disease. EBioMedicine 2024; 101:104996. [PMID: 38320344 PMCID: PMC10862506 DOI: 10.1016/j.ebiom.2024.104996] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUND The pathogenesis of MASLD (metabolic dysfunction-associated steatotic liver disease), including its severe clinical forms, involves complex processes at all levels of biological organization. This study examined the potential link between the liver microbiome profile and epigenetic factors. METHODS Liver microbial DNA composition was analysed using high throughput 16S rRNA gene sequencing in 116 individuals, with 55% being female, across the spectrum of liver disease severity. Total activity of histone deacetylases (HDACs) and acetyltransferases (HATs) was assayed in nuclear extracts from fresh liver samples. In addition, we measured the global 5-hydroxymethylcytosine (5-hmC) levels of liver DNA. FINDINGS Patients with MASLD showed a 2.07-fold increase (p = 0.013) in liver total HAT activity. Moreover, a correlation was observed between liver total HAT activity and the score for histological steatosis (Spearman's R = 0.60, p = 1.0E-3) and disease severity (R = 0.40, p = 2.0E-2). Liver HAT and HDAC activities also showed associations with the abundance of several liver bacterial DNAs. Additionally, liver global levels of 5-hmC showed negative correlation with the read number of Bacteroidetes (R = -0.62, p = 9.3E-4) and Gammaproteobacteria (R = -0.43, p = 3.2E-2), while it was positively correlated with the abundance of Acidobacteria (R = 0.42, p = 4.1E-2) and Actinobacteria (R = 0.47, p = 1.8E-2). INTERPRETATION The host liver epigenome, including the activity of enzymes involved in maintaining the balance between protein acetylation and deacetylation and the global DNA hydroxy-methylation status, may be the target of microbial signals. FUNDING Agencia Nacional de Promoción Científica y Tecnológica, FonCyT.
Collapse
Affiliation(s)
- Carlos Jose Pirola
- Systems Biology of Complex Diseases, Translational Health Research Center (CENITRES), Maimónides University, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Faculty of Health Science, Maimónides University, Buenos Aires, Argentina.
| | - Adrian Salatino
- Max Planck Institute for Immunobiology and Epigenetics, Bioinformatics Facility, Germany
| | - Tomas Fernández Gianotti
- Systems Biology of Complex Diseases, Translational Health Research Center (CENITRES), Maimónides University, Buenos Aires, Argentina
| | - Gustavo Osvaldo Castaño
- Liver Unit, Medicine and Surgery Department, Hospital Abel Zubizarreta, Ciudad Autónoma de Buenos Aires, Argentina
| | - Martin Garaycoechea
- Department of Surgery, Hospital de Alta Complejidad en Red "El Cruce", Florencio Varela, Buenos Aires, Argentina
| | - Silvia Sookoian
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina; Faculty of Health Science, Maimónides University, Buenos Aires, Argentina; Clinical and Molecular Hepatology, Translational Health Research Center (CENITRES), Maimónides University, Buenos Aires, Argentina.
| |
Collapse
|
8
|
Mohanan A, Harilal SL, Plakkot B, Pottakkat B, Kanakkaparambil R. Nutritional Epigenetics and Gut Microbiome. EPIGENETICS AND HUMAN HEALTH 2024:121-159. [DOI: 10.1007/978-3-031-54215-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
9
|
Zhang G, Le Souëf P. The influence of modern living conditions on the human microbiome and potential therapeutic opportunities for allergy prevention. World Allergy Organ J 2024; 17:100857. [PMID: 38235259 PMCID: PMC10793171 DOI: 10.1016/j.waojou.2023.100857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/01/2023] [Accepted: 12/09/2023] [Indexed: 01/19/2024] Open
Abstract
Modern living conditions and the recent surge in global urbanization have transformed the human microbiome. This transformation is believed to be a significant factor in the recent spike of common chronic inflammatory diseases like asthma and allergies worldwide, evident in both developed and developing nations. Immigrants from less developed regions who settle in highly urbanized and affluent areas present an ideal demographic for research. Investigating immigrant populations can yield valuable insights, particularly when studying microbiome changes that occur as individuals transition from areas with low asthma prevalence to regions with a high prevalence of the condition. The application of prebiotics and probiotics as potential treatments for asthma and allergies faces challenges. This is due to the complex interplay of numerous factors that contribute to their aetiology. Exploring the interaction between the human microbiome and potential epigenetic changes in specific populations, such as immigrants adapting to new, urbanized environments, may offer crucial insights. Such research could underscore the role of prebiotics and probiotics in preventing allergic conditions. Recognizing the changes in the human microbiome in the context of a Western/modern environment might be essential in addressing the increasing prevalence of allergic diseases. Persistent research in this domain is pivotal for devising effective interventions such as dietary supplementation with prebiotics and probiotics.
Collapse
Affiliation(s)
- Guicheng Zhang
- School of Population Health, Curtin University, Perth, 6102, Western Australia, Australia
- School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia 6008, Australia
| | - Peter Le Souëf
- School of Population Health, Curtin University, Perth, 6102, Western Australia, Australia
- School of Medicine, The University of Western Australia, Perth, Western Australia 6008, Australia
| |
Collapse
|
10
|
Giannì ML, Morniroli D, Mosca F, Rescigno M. Can Postbiotics Represent a New Strategy for NEC? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1449:43-57. [PMID: 39060730 DOI: 10.1007/978-3-031-58572-2_3] [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
Intestinal bacteria, also known as gut microbiota, are a rich ecology of microorganisms found in the human digestive tract. Extensive study has highlighted their critical relevance in preserving human health. New research has revealed that bacterial viability is not invariably necessary to induce health benefits. Postbiotics (defined soluble substances produced as a byproduct of the metabolic processes of living microbes) have thus emerged as an important topic of research. They contribute to shaping the gut microbiota, exert immune-modulation activity, and improve the integrity of the gut barrier.Alterations in preterm gut colonization associated with intestinal barrier immaturity and the increased reactivity of the intestinal mucosa to colonizing bacteria have been implicated in the pathogenesis of necrotizing enterocolitis. Postbiotics have shown promising outcomes in reducing the risk of developing NEC, lowering inflammation, encouraging the development of good bacteria, and strengthening the intestinal barrier. This is an important advancement in newborn care and highlights the potential of postbiotics to avoid severe intestinal disorders.
Collapse
Affiliation(s)
- Maria Lorella Giannì
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Daniela Morniroli
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy
| | - Fabio Mosca
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy.
- Department of Clinical Science and Community Health, University of Milan, Milan, Italy.
| | | |
Collapse
|
11
|
Desai S. Influence of pathogens on host genome and epigenome in development of head and neck cancer. Cancer Rep (Hoboken) 2023; 6:e1846. [PMID: 37322598 PMCID: PMC10644332 DOI: 10.1002/cnr2.1846] [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: 04/01/2023] [Revised: 05/11/2023] [Accepted: 06/05/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Head and neck cancer (HNSCC) is a heterogeneous group of cancers, affecting multiple regions such as oral cavity, pharynx, larynx, and nasal region, each showing a distinct molecular profile. HNSCC accounts for more than 6 million cases worldwide, soaring mainly in the developing countries. RECENT FINDINGS The aetiology of HNSCC is complex and multifactorial, involving both genetic and environmental factors. The critical role of microbiome, which includes bacteria, viruses, and fungi, is under spotlight due to the recent reports on their contribution in the development and progression of HNSCC. This review focuses on the effect of opportunistic pathogens on the host genome and epigenome, which contributes to the disease progression. Drawing parallels from the host-pathogen interactions observed in other tumour types arising from the epithelial tissue such as colorectal cancer, the review also calls attention to the potential explorations of the role of pathogens in HNSCC biology and discusses the clinical implications of microbiome research in detection and treatment of HNSCC. CONCLUSION Our understanding of the genomic effects of the microbes on the disease progression and the mechanistic insights of the host-pathogen interaction will pave way to novel treatment and preventive approaches in HNSCC.
Collapse
|
12
|
Wang AJ, Song D, Hong YM, Liu NN. Multi-omics insights into the interplay between gut microbiota and colorectal cancer in the "microworld" age. Mol Omics 2023; 19:283-296. [PMID: 36916422 DOI: 10.1039/d2mo00288d] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Colorectal cancer (CRC) is a multifactorial heterogeneous disease largely due to both genetic predisposition and environmental factors including the gut microbiota, a dynamic microbial ecosystem inhabiting the gastrointestinal tract. Elucidation of the molecular mechanisms by which the gut microbiota interacts with the host may contribute to the pathogenesis, diagnosis, and promotion of CRC. However, deciphering the influence of genetic variants and interactions with the gut microbial ecosystem is rather challenging. Despite recent advancements in single omics analysis, the application of multi-omics approaches to integrate multiple layers of information in the microbiome and host to introduce effective prevention, diagnosis, and treatment strategies is still in its infancy. Here, we integrate host- and microbe-based multi-omics studies, respectively, to provide a strategy to explore potential causal relationships between gut microbiota and colorectal cancer. Specifically, we summarize the recent multi-omics studies such as metagenomics combined with metabolomics and metagenomics combined with genomics. Meanwhile, the sample size and sample types commonly used in multi-omics research, as well as the methods of data analysis, were also generalized. We highlight multiple layers of information from multi-omics that need to be verified by different types of models. Together, this review provides new insights into the clinical diagnosis and treatment of colorectal cancer patients.
Collapse
Affiliation(s)
- An-Jun Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Dingka Song
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Yue-Mei Hong
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| | - Ning-Ning Liu
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China.
| |
Collapse
|
13
|
Wang L, Liang X, Chen H, Cao L, Liu L, Zhu F, Ding Y, Tang J, Xie Y. CDEMI: characterizing differences in microbial composition and function in microbiome data. Comput Struct Biotechnol J 2023; 21:2502-2513. [PMID: 37090432 PMCID: PMC10113763 DOI: 10.1016/j.csbj.2023.03.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/28/2023] Open
Abstract
Microbial communities influence host phenotypes through microbiota-derived metabolites and interactions between exogenous active substances (EASs) and the microbiota. Owing to the high dynamics of microbial community composition and difficulty in microbial functional analysis, the identification of mechanistic links between individual microbes and host phenotypes is complex. Thus, it is important to characterize variations in microbial composition across various conditions (for example, topographical locations, times, physiological and pathological conditions, and populations of different ethnicities) in microbiome studies. However, no web server is currently available to facilitate such characterization. Moreover, accurately annotating the functions of microbes and investigating the possible factors that shape microbial function are critical for discovering links between microbes and host phenotypes. Herein, an online tool, CDEMI, is introduced to discover microbial composition variations across different conditions, and five types of microbe libraries are provided to comprehensively characterize the functionality of microbes from different perspectives. These collective microbe libraries include (1) microbial functional pathways, (2) disease associations with microbes, (3) EASs associations with microbes, (4) bioactive microbial metabolites, and (5) human body habitats. In summary, CDEMI is unique in that it can reveal microbial patterns in distributions/compositions across different conditions and facilitate biological interpretations based on diverse microbe libraries. CDEMI is accessible at http://rdblab.cn/cdemi/.
Collapse
Affiliation(s)
- Lidan Wang
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing 401147, China
| | - Xiao Liang
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hao Chen
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lijie Cao
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lan Liu
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Feng Zhu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yubin Ding
- Department of Obstetrics and Gynecology, Women and Children’s Hospital of Chongqing Medical University, Chongqing 401147, China
- Corresponding authors.
| | - Jing Tang
- School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China
- Joint International Research Laboratory of Reproductive and Development, Department Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing 400016, China
- Corresponding author at: School of Basic Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Youlong Xie
- Joint International Research Laboratory of Reproductive and Development, Department Reproductive Biology, School of Public Health, Chongqing Medical University, Chongqing 400016, China
- Corresponding authors.
| |
Collapse
|
14
|
Extracellular vesicle microRNAs in celiac disease patients under a gluten-free diet, and in lactose intolerant individuals. BBA ADVANCES 2022; 2:100053. [PMID: 37082606 PMCID: PMC10074944 DOI: 10.1016/j.bbadva.2022.100053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Celiac disease (CD) is an autoimmune disorder triggered by an abnormal immunological response to gluten ingestion and is associated with deregulated expression of cellular microRNAs (miRNAs) of the gut mucosa. It is frequently misdiagnosed as lactose intolerance (LI) due to symptom resemblance. Microvilli loss may be counteracted by a rigorous gluten-free diet (GFD). AIMS To identify altered extracellular vesicle miRNAs from plasma among CD patients on GFD (n=34), lactose intolerant individuals on restrictive diet (n=14) and controls (n=23), and to predict biological pathways in which these altered miRNAs may play a part. METHODS Five different small RNA samples of each group were pooled twice and then screened by new-generation sequencing. Four miRNAs were selected to be quantified by RT-qPCR in the entire sample. RESULTS The levels of four miRNAs - miR-99b-3p, miR-197-3p, miR-223-3p, and miR-374b-5p - differed between CD patients and controls (P<0.05). Apart from miR-223-3p, all these miRNAs tended to have altered levels also between LI and controls (P<0.10). The results for miR-99b-3p and miR-197-3p between CD and controls were confirmed by RT-qPCR, which also indicated different levels of miR-99b-3p and miR-374b-5p between CD-associated LI and LI (P<0.05). CONCLUSIONS These miRNAs may have targets that affect cell death, cell communication, adhesion, and inflammation modulation pathways. Hence, altered miRNA levels could be associated with CD-related aspects and gut mucosa recovery.
Collapse
|