1801
|
Gerbeth L, Glauben R. Histone Deacetylases in the Inflamed Intestinal Epithelium-Promises of New Therapeutic Strategies. Front Med (Lausanne) 2021; 8:655956. [PMID: 33842512 PMCID: PMC8032862 DOI: 10.3389/fmed.2021.655956] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 03/05/2021] [Indexed: 12/22/2022] Open
Abstract
The intestinal epithelium is a complex, dynamic barrier that separates luminal contents from the immune compartment while mediating nutrient absorption and controlled passage of antigens to convey oral tolerance. A compromised epithelial barrier often leads to inflammation because immune cells in the lamina propria come into direct contact with luminal antigens. Defects in epithelial cell function were also shown to be involved in the etiology of inflammatory bowel diseases. These are severe, chronically relapsing inflammatory conditions of the gastrointestinal tract that also increase the risk of developing colorectal cancer. Despite major efforts of the scientific community, the precise causes and drivers of these conditions still remain largely obscured impeding the development of a permanent cure. Current therapeutic approaches mostly focus on alleviating symptoms by targeting immune cell signaling. The protein family of histone deacetylases (HDACs) has gained increasing attention over the last years, as HDAC inhibitors were shown to be potent tumor cell suppressors and also alleviate morbid inflammatory responses. Recent research continuously identifies new roles for specific HDACs suggesting that HDACs influence the cell signaling network from many different angles. This makes HDACs very interesting targets for therapeutic approaches but predicting effects after system manipulations can be difficult. In this review, we want to provide a comprehensive overview of current knowledge about the individual roles of HDACs in the intestinal epithelium to evaluate their therapeutic potential for inflammatory conditions of the gut.
Collapse
Affiliation(s)
- Lorenz Gerbeth
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Medical Biotechnology, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Rainer Glauben
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
1802
|
Roles of interstitial fluid pH and weak organic acids in development and amelioration of insulin resistance. Biochem Soc Trans 2021; 49:715-726. [PMID: 33769491 DOI: 10.1042/bst20200667] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022]
Abstract
Type 2 diabetes mellitus (T2DM) is one of the most common lifestyle-related diseases (metabolic disorders) due to hyperphagia and/or hypokinesia. Hyperglycemia is the most well-known symptom occurring in T2DM patients. Insulin resistance is also one of the most important symptoms, however, it is still unclear how insulin resistance develops in T2DM. Detailed understanding of the pathogenesis primarily causing insulin resistance is essential for developing new therapies for T2DM. Insulin receptors are located at the plasma membrane of the insulin-targeted cells such as myocytes, adipocytes, etc., and insulin binds to the extracellular site of its receptor facing the interstitial fluid. Thus, changes in interstitial fluid microenvironments, specially pH, affect the insulin-binding affinity to its receptor. The most well-known clinical condition regarding pH is systemic acidosis (arterial blood pH < 7.35) frequently observed in severe T2DM associated with insulin resistance. Because the insulin-binding site of its receptor faces the interstitial fluid, we should recognize the interstitial fluid pH value, one of the most important factors influencing the insulin-binding affinity. It is notable that the interstitial fluid pH is unstable compared with the arterial blood pH even under conditions that the arterial blood pH stays within the normal range, 7.35-7.45. This review article introduces molecular mechanisms on unstable interstitial fluid pH value influencing the insulin action via changes in insulin-binding affinity and ameliorating actions of weak organic acids on insulin resistance via their characteristics as bases after absorption into the body even with sour taste at the tongue.
Collapse
|
1803
|
Guo W, Zhou X, Li X, Zhu Q, Peng J, Zhu B, Zheng X, Lu Y, Yang D, Wang B, Wang J. Depletion of Gut Microbiota Impairs Gut Barrier Function and Antiviral Immune Defense in the Liver. Front Immunol 2021; 12:636803. [PMID: 33841420 PMCID: PMC8027085 DOI: 10.3389/fimmu.2021.636803] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 03/08/2021] [Indexed: 02/06/2023] Open
Abstract
Commensal gut microbiota protects the immune defense of extra-intestinal organs. Gut microbiota depletion by antibiotics can impair host antiviral immune responses and alter hepatitis B virus (HBV) infection outcomes. However, how gut microbiota modulates antiviral immune response in the liver remains unclear. Here, mice were treated with broad-spectrum antibiotics to deplete gut microbiota. Gut integrity was evaluated, and translocation of live commensal gut bacteria and their components into the liver was investigated. An HBV infection model was established to evaluate impairment of antiviral immune response in the liver after gut microbiota depletion. We found that gut microbiota depletion was associated with impairment of colon epithelial integrity, and live commensal gut microbiota could translocate to the liver. Further, T cell antiviral function in the liver was impaired, partially relying on enhanced PD-1 expression, and HBV immune clearance was hampered. In conclusion, gut microbiota depletion by antibiotics can impair gut barrier function and suppress T cell antiviral immune response in the liver.
Collapse
Affiliation(s)
- Weina Guo
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zhou
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoran Li
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingfeng Zhu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Peng
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Zhu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Zheng
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinping Lu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dongliang Yang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Baoju Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junzhong Wang
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
1804
|
Ojeda J, Ávila A, Vidal PM. Gut Microbiota Interaction with the Central Nervous System throughout Life. J Clin Med 2021; 10:1299. [PMID: 33801153 PMCID: PMC8004117 DOI: 10.3390/jcm10061299] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 02/08/2023] Open
Abstract
During the last years, accumulating evidence has suggested that the gut microbiota plays a key role in the pathogenesis of neurodevelopmental and neurodegenerative diseases via the gut-brain axis. Moreover, current research has helped to elucidate different communication pathways between the gut microbiota and neural tissues (e.g., the vagus nerve, tryptophan production, extrinsic enteric-associated neurons, and short chain fatty acids). On the other hand, altering the composition of gut microbiota promotes a state known as dysbiosis, where the balance between helpful and pathogenic bacteria is disrupted, usually stimulating the last ones. Herein, we summarize selected findings of the recent literature concerning the gut microbiome on the onset and progression of neurodevelopmental and degenerative disorders, and the strategies to modulate its composition in the search for therapeutical approaches, focusing mainly on animal models studies. Readers are advised that this is a young field, based on early studies, that is rapidly growing and being updated as the field advances.
Collapse
Affiliation(s)
- Jorge Ojeda
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción 4090541, Chile;
| | - Ariel Ávila
- Developmental Neurobiology Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción 4090541, Chile;
| | - Pía M. Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, Concepción 4090541, Chile;
| |
Collapse
|
1805
|
Impaired Butyrate Induced Regulation of T Cell Surface Expression of CTLA-4 in Patients with Ulcerative Colitis. Int J Mol Sci 2021; 22:ijms22063084. [PMID: 33802979 PMCID: PMC8002718 DOI: 10.3390/ijms22063084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 01/08/2023] Open
Abstract
Patients with ulcerative colitis (UC) have reduced intestinal levels of short-chain fatty acids (SCFAs), including butyrate, which are important regulators of host–microbiota crosstalk. The aim was therefore to determine effects of butyrate on blood and intestinal T cells from patients with active UC. T cells from UC patients and healthy subjects were polyclonally stimulated together with SCFAs and proliferation, activation, cytokine secretion, and surface expression of cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) were analyzed. Butyrate induced comparable, dose dependent inhibition of activation and proliferation in blood T cells and activation in intestinal T cells from UC patients and healthy subjects. However, surface expression of the inhibitory molecule CTLA-4 on stimulated blood and intestinal T cells was impaired in UC patients and was not restored following butyrate treatment. Furthermore, unlike intestinal T cells from healthy subjects, butyrate was unable to downregulate secretion of interferon gamma (IFNγ), interleukin (IL)-4, IL-17A, and IL-10 in UC patients. Although seemingly normal inhibitory effects on T cell activation and proliferation, butyrate has an impaired ability to reduce cytokine secretion and induce surface expression of CTLA-4 in T cells from UC patients with active disease. Overall, these observations indicate a dysfunction in butyrate induced immune regulation linked to CTLA-4 signaling in T cells from UC patients during a flare.
Collapse
|
1806
|
Huschtscha Z, Parr A, Porter J, Costa RJS. The Effects of a High-Protein Dairy Milk Beverage With or Without Progressive Resistance Training on Fat-Free Mass, Skeletal Muscle Strength and Power, and Functional Performance in Healthy Active Older Adults: A 12-Week Randomized Controlled Trial. Front Nutr 2021; 8:644865. [PMID: 33816540 PMCID: PMC8010144 DOI: 10.3389/fnut.2021.644865] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022] Open
Abstract
The study aimed to investigate the independent and combined effects of consuming a high-protein dairy milk beverage, twice daily, with or without a progressive resistance training (PRT) program on outcomes of age-related sarcopenia, in healthy active older (≥50 years) adults. In this 12-week, 2 × 2 factorial study, participants were randomly allocated into one of four groups: dairy milk beverage (DM), exercise and dairy milk beverage (EX+DM), exercise alone (EX), and control (CON). The EX group underwent a 12-week whole-body PRT schedule (three sessions/week) and a high-protein dairy milk beverage (DM) was consumed twice daily (30 g protein/day). At weeks 0, 6, and 12, body composition (iDXA), strength [one-repetition maximum (1RM): leg press, chest press, lateral (lat) pull-down, and handgrip], power (countermovement jump), cardiorespiratory fitness (VO2), and physical performance (gait speed) were measured. Before measurements, blood samples were collected to determine the immune (i.e., leukocyte trafficking and inflammatory cytokines) and hormonal (i.e., insulin, cortisol, IGF-1, testosterone, and estradiol) profiles. Participants (n = 37) completed the study within the controlled experimental conditions. Protein intake increased in the EX+DM [mean ± SD, 1.2 ± 0.2 to 1.8 ± 0.4 g/kg body mass (BM) per day-1] and DM (1.3 ± 0.5 to 1.8 ± 0.6 g kg-1 BM day-1) groups during the intervention. Absolute fat-free mass increased in the EX+DM [mean (95% confidence interval) = 0.65 (0.25-1.0) kg] and EX [0.49 (-0.44 to 1.40) kg] groups (P < 0.001) compared to DM [-0.54 (-1.6 to 0.05) kg]. Relative fat mass decreased (group*time, P = 0.018) in DM [-1.8% (-3.3 to -0.35%)] and EX+DM [-1.3% (-2.3 to -0.31%)], which was a greater reduction than that in the CON [0.10% (-0.80 to 1.0%)] group (P < 0.01). Relative maximal strength increased in both the EX and EX+DM (≥35%, P < 0.05) groups, but not in the DM and CON groups. The change in 1RM strength outcomes was higher in EX+DM compared to all other groups (53-78%, P < 0.01). There was an increase in resting plasma IL-10 concentration in EX+DM (88%), compared to all the other groups (P = 0.016). No other differences in systemic inflammatory cytokines were observed. There were no significant changes in all hormone concentrations measured among all groups. In conclusion, a high-protein dairy milk beverage providing additional protein did not further enhance the effects of PRT on outcomes of fat-free mass, power, or physical performance. However, there was a significant augmentative effect for high-protein dairy milk consumption on changes to maximal strength outcomes during PRT in healthy active older adults.
Collapse
Affiliation(s)
- Zoya Huschtscha
- Department of Nutrition Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Alexandra Parr
- Department of Nutrition Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| | - Judi Porter
- Department of Nutrition Dietetics & Food, Monash University, Notting Hill, VIC, Australia
- School of Exercise and Nutrition Sciences, Institute for Physical Activity and Nutrition (IPAN), Deakin University, Geelong, VIC, Australia
| | - Ricardo J. S. Costa
- Department of Nutrition Dietetics & Food, Monash University, Notting Hill, VIC, Australia
| |
Collapse
|
1807
|
Molino S, Lerma-Aguilera A, Jiménez-Hernández N, Gosalbes MJ, Rufián-Henares JÁ, Francino MP. Enrichment of Food With Tannin Extracts Promotes Healthy Changes in the Human Gut Microbiota. Front Microbiol 2021; 12:625782. [PMID: 33796085 PMCID: PMC8008114 DOI: 10.3389/fmicb.2021.625782] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/24/2021] [Indexed: 01/04/2023] Open
Abstract
Food and food bioactive components are major drivers of modulation of the human gut microbiota. Tannin extracts consist of a mix of bioactive compounds, which are already exploited in the food industry for their chemical and sensorial properties. The aim of our study was to explore the viability of associations between tannin wood extracts of different origin and food as gut microbiota modulators. 16S rRNA amplicon next-generation sequencing (NGS) was used to test the effects on the gut microbiota of tannin extracts from quebracho, chestnut, and tara associated with commercial food products with different composition in macronutrients. The different tannin-enriched and non-enriched foods were submitted to in vitro digestion and fermentation by the gut microbiota of healthy subjects. The profile of the short chain fatty acids (SCFAs) produced by the microbiota was also investigated. The presence of tannin extracts in food promoted an increase of the relative abundance of the genus Akkermansia, recognized as a marker of a healthy gut, and of various members of the Lachnospiraceae and Ruminococcaceae families, involved in SCFA production. The enrichment of foods with tannin extracts had a booster effect on the production of SCFAs, without altering the profile given by the foods alone. These preliminary results suggest a positive modulation of the gut microbiota with potential benefits for human health through the enrichment of foods with tannin extracts.
Collapse
Affiliation(s)
- Silvia Molino
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
| | - Alberto Lerma-Aguilera
- Area de Genòmica i Salut, Fundació per al Foment de la Investigació Sanitària i Biomèdica de la Comunitat Valenciana (FISABIO-Salut Pública), València, Spain
| | - Nuria Jiménez-Hernández
- Area de Genòmica i Salut, Fundació per al Foment de la Investigació Sanitària i Biomèdica de la Comunitat Valenciana (FISABIO-Salut Pública), València, Spain.,CIBER en Epidemiología y Salud Pública, Madrid, Spain
| | - María José Gosalbes
- Area de Genòmica i Salut, Fundació per al Foment de la Investigació Sanitària i Biomèdica de la Comunitat Valenciana (FISABIO-Salut Pública), València, Spain.,CIBER en Epidemiología y Salud Pública, Madrid, Spain
| | - José Ángel Rufián-Henares
- Departamento de Nutrición y Bromatología, Instituto de Nutrición y Tecnología de los Alimentos, Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain.,Instituto de Investigación Biosanitaria ibs.Granada, Granada, Spain
| | - M Pilar Francino
- Area de Genòmica i Salut, Fundació per al Foment de la Investigació Sanitària i Biomèdica de la Comunitat Valenciana (FISABIO-Salut Pública), València, Spain.,CIBER en Epidemiología y Salud Pública, Madrid, Spain
| |
Collapse
|
1808
|
Kaur AP, Bhardwaj S, Dhanjal DS, Nepovimova E, Cruz-Martins N, Kuča K, Chopra C, Singh R, Kumar H, Șen F, Kumar V, Verma R, Kumar D. Plant Prebiotics and Their Role in the Amelioration of Diseases. Biomolecules 2021; 11:440. [PMID: 33809763 PMCID: PMC8002343 DOI: 10.3390/biom11030440] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Prebiotics are either natural or synthetic non-digestible (non-)carbohydrate substances that boost the proliferation of gut microbes. Undigested fructooligosaccharides in the large intestine are utilised by the beneficial microorganisms for the synthesis of short-chain fatty acids for their own growth. Although various food products are now recognized as having prebiotic properties, several others, such as almonds, artichoke, barley, chia seeds, chicory, dandelion greens, flaxseeds, garlic, and oats, are being explored and used as functional foods. Considering the benefits of these prebiotics in mineral absorption, metabolite production, gut microbiota modulation, and in various diseases such as diabetes, allergy, metabolic disorders, and necrotising enterocolitis, increasing attention has been focused on their applications in both food and pharmaceutical industries, although some of these food products are actually used as food supplements. This review aims to highlight the potential and need of these prebiotics in the diet and also discusses data related to the distinct types, sources, modes of action, and health benefits.
Collapse
Affiliation(s)
- Amrit Pal Kaur
- School of Bioengineering and Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India; (A.P.K.); (H.K.)
| | - Sonali Bhardwaj
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.B.); (D.S.D.); (C.C.); (R.S.)
| | - Daljeet Singh Dhanjal
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.B.); (D.S.D.); (C.C.); (R.S.)
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Laboratory of Neuropsychophysiology, Faculty of Psychology and Education Sciences, University of Porto, 4200-135 Porto, Portugal
| | - Kamil Kuča
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic;
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.B.); (D.S.D.); (C.C.); (R.S.)
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab 144411, India; (S.B.); (D.S.D.); (C.C.); (R.S.)
| | - Harsh Kumar
- School of Bioengineering and Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India; (A.P.K.); (H.K.)
| | - Fatih Șen
- Sen Research Group, Biochemistry Department, Faculty of Arts and Science, EvliyaÇelebi Campus, Dumlupınar University, Kütahya 43100, Turkey;
| | - Vinod Kumar
- School of Water, Energy and Environment, Cranfield University, Cranfield MK430AL, UK;
| | - Rachna Verma
- School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India;
| | - Dinesh Kumar
- School of Bioengineering and Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India; (A.P.K.); (H.K.)
| |
Collapse
|
1809
|
Lorente-Picón M, Laguna A. New Avenues for Parkinson's Disease Therapeutics: Disease-Modifying Strategies Based on the Gut Microbiota. Biomolecules 2021; 11:433. [PMID: 33804226 PMCID: PMC7998286 DOI: 10.3390/biom11030433] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a multifactorial neurodegenerative disorder that currently affects 1% of the population over the age of 60 years, and for which no disease-modifying treatments exist. Neurodegeneration and neuropathology in different brain areas are manifested as both motor and non-motor symptoms in patients. Recent interest in the gut-brain axis has led to increasing research into the gut microbiota changes in PD patients and their impact on disease pathophysiology. As evidence is piling up on the effects of gut microbiota in disease development and progression, another front of action has opened up in relation to the potential usage of microbiota-based therapeutic strategies in treating gastrointestinal alterations and possibly also motor symptoms in PD. This review provides status on the different strategies that are in the front line (i.e., antibiotics; probiotics; prebiotics; synbiotics; dietary interventions; fecal microbiota transplantation, live biotherapeutic products), and discusses the opportunities and challenges the field of microbiome research in PD is facing.
Collapse
Affiliation(s)
- Marina Lorente-Picón
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| | - Ariadna Laguna
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
1810
|
Zhang Y, Yin C, Schroyen M, Everaert N, Ma T, Zhang H. Effects of the Inclusion of Fermented Mulberry Leaves and Branches in the Gestational Diet on the Performance and Gut Microbiota of Sows and Their Offspring. Microorganisms 2021; 9:604. [PMID: 33804202 PMCID: PMC7998242 DOI: 10.3390/microorganisms9030604] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 01/02/2023] Open
Abstract
Fermented feed mulberry (FFM), being rich in dietary fiber, has not been fully evaluated to be used in sow's diet. In this study, we investigated the effects of 25.5% FFM supplemented in gestation diets on the performance and gut microbiota of sows and their offspring. Results showed that the serum concentration of glucose, progesterone, and estradiol were not affected by the dietary treatment, while the level of serum insulin and fecal short chain fatty acid were both reduced in FFM group on gestation day 60 (G60, p < 0.05). Additionally, FFM increased both voluntary feed intake and weaning litter weight (p < 0.05), while decreased the losses of both Backfat thickness and bodyweight throughout lactation (p < 0.05). 16S rRNA sequencing showed FFM supplementation significantly increased the diversity and relative abundance of sows' fecal microbiota on G60 (p < 0.05). The differential microbiota for sows from FFM group was that Bacteroidetes was increased on G60 while Firmicutes were decreased on Lactation day 7 (L7, p < 0.05), and which for the FFM piglets was that both unclassified_f_Lachnospiraceae on L0 and norank_f_Ruminococcaceae on L7 were increased (p < 0.05). In short, FFM can be recognized as a potential feed ingredient used in sow's diet.
Collapse
Affiliation(s)
- Yuping Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.Z.); (C.Y.); (H.Z.)
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium; (M.S.); (N.E.)
| | - Chang Yin
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.Z.); (C.Y.); (H.Z.)
| | - Martine Schroyen
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium; (M.S.); (N.E.)
| | - Nadia Everaert
- Precision Livestock and Nutrition Laboratory, TERRA Teaching and Research Centre, Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium; (M.S.); (N.E.)
| | - Teng Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.Z.); (C.Y.); (H.Z.)
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (Y.Z.); (C.Y.); (H.Z.)
| |
Collapse
|
1811
|
Mu Y, Kinashi Y, Li J, Yoshikawa T, Kishimura A, Tanaka M, Matsui T, Mori T, Hase K, Katayama Y. Polyvinyl Butyrate Nanoparticles as Butyrate Donors for Colitis Treatment. ACS APPLIED BIO MATERIALS 2021; 4:2335-2341. [DOI: 10.1021/acsabm.0c01105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Yunmei Mu
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Yusuke Kinashi
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
| | - Jinting Li
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takuma Yoshikawa
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Akihiro Kishimura
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- International Research Center for Molecular System, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Mitsuru Tanaka
- Research and Development Center for Five-Sense Devices, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Toshiro Matsui
- Department of Bioscience and Biotechnology, Faculty of Agriculture, Graduate School of Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Takeshi Mori
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan
- Division of Mucosal Barrierology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yoshiki Katayama
- Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Center for Future Chemistry, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- International Research Center for Molecular System, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
- Department of Biomedical Engineering, Chung Yuan Christian University, 200 Chung Pei Rd., Chung Li, Taoyuan 32023, Taiwan
| |
Collapse
|
1812
|
Wong CW, Yost SE, Lee JS, Gillece JD, Folkerts M, Reining L, Highlander SK, Eftekhari Z, Mortimer J, Yuan Y. Analysis of Gut Microbiome Using Explainable Machine Learning Predicts Risk of Diarrhea Associated With Tyrosine Kinase Inhibitor Neratinib: A Pilot Study. Front Oncol 2021; 11:604584. [PMID: 33796451 PMCID: PMC8008168 DOI: 10.3389/fonc.2021.604584] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/22/2021] [Indexed: 01/22/2023] Open
Abstract
Neratinib has great efficacy in treating HER2+ breast cancer but is associated with significant gastrointestinal toxicity. The objective of this pilot study was to understand the association of gut microbiome and neratinib-induced diarrhea. Twenty-five patients (age ≥ 60) were enrolled in a phase II trial evaluating safety and tolerability of neratinib in older adults with HER2+ breast cancer (NCT02673398). Fifty stool samples were collected from 11 patients at baseline and during treatment. 16S rRNA analysis was performed and relative abundance data were generated. Shannon's diversity was calculated to examine gut microbiome dysbiosis. An explainable tree-based approach was utilized to classify patients who might experience neratinib-related diarrhea (grade ≥ 1) based on pre-treatment baseline microbial relative abundance data. The hold-out Area Under Receiver Operating Characteristic and Area Under Precision-Recall Curves of the model were 0.88 and 0.95, respectively. Model explanations showed that patients with a larger relative abundance of Ruminiclostridium 9 and Bacteroides sp. HPS0048 may have reduced risk of neratinib-related diarrhea and was confirmed by Kruskal-Wallis test (p ≤ 0.05, uncorrected). Our machine learning model identified microbiota associated with reduced risk of neratinib-induced diarrhea and the result from this pilot study will be further verified in a larger study. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov, identifier NCT02673398.
Collapse
Affiliation(s)
- Chi Wah Wong
- Department of Applied AI and Data Science, City of Hope National Medical Center, Duarte, CA, United States
| | - Susan E. Yost
- Department of Medical Oncology & Therapeutic Research, City of Hope National Medical Center, Duarte, CA, United States
| | - Jin Sun Lee
- Department of Medical Oncology & Therapeutic Research, City of Hope National Medical Center, Duarte, CA, United States
| | - John D. Gillece
- Pathogen and Microbiome Division, Translational Genomics Research Institute North, Flagstaff, AZ, United States
| | - Megan Folkerts
- Pathogen and Microbiome Division, Translational Genomics Research Institute North, Flagstaff, AZ, United States
| | - Lauren Reining
- Pathogen and Microbiome Division, Translational Genomics Research Institute North, Flagstaff, AZ, United States
| | - Sarah K. Highlander
- Pathogen and Microbiome Division, Translational Genomics Research Institute North, Flagstaff, AZ, United States
| | - Zahra Eftekhari
- Department of Applied AI and Data Science, City of Hope National Medical Center, Duarte, CA, United States
| | - Joanne Mortimer
- Department of Medical Oncology & Therapeutic Research, City of Hope National Medical Center, Duarte, CA, United States
| | - Yuan Yuan
- Department of Medical Oncology & Therapeutic Research, City of Hope National Medical Center, Duarte, CA, United States
| |
Collapse
|
1813
|
Abbasi A, Rad AH, Ghasempour Z, Sabahi S, Kafil HS, Hasannezhad P, Rahbar Saadat Y, Shahbazi N. The biological activities of postbiotics in gastrointestinal disorders. Crit Rev Food Sci Nutr 2021; 62:5983-6004. [PMID: 33715539 DOI: 10.1080/10408398.2021.1895061] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
According to outcomes from clinical studies, an intricate relationship occurs between the beneficial microbiota, gut homeostasis, and the host's health status. Numerous studies have confirmed the health-promoting effects of probiotics, particularly in gastrointestinal diseases. On the other hand, the safety issues regarding the consumption of some probiotics are still a matter of debate, thus to overcome the problems related to the application of live probiotic cells in terms of clinical, technological, and economic aspects, microbial-derived biomolecules (postbiotics) were introducing as a potential alternative agent. Presently scientific literature confirms that the postbiotic components can be used as promising tools for both prevention and treatment strategies in gastrointestinal disorders with less undesirable side-effects, particularly in infants and children. Future head-to-head trials are required to distinguish appropriate strains of parent cells, optimal dosages of postbiotics, and assessment of the cost-effectiveness of postbiotics compared to alternative drugs. This review provides an overview of the concept and safety issues regarding postbiotics, with emphasis on their biological role in the treatment of some important gastrointestinal disorders.
Collapse
Affiliation(s)
- Amin Abbasi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Homayouni Rad
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Ghasempour
- Department of Food Science and Technology, Faculty of Nutrition & Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sahar Sabahi
- Department of Nutritional Sciences, School of Paramedical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Paniz Hasannezhad
- Department of Medical Engineering Science, University College of Rouzbahan, Sari, Iran
| | - Yalda Rahbar Saadat
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nayyer Shahbazi
- Faculty of Agriculture Engineering, Department of Food Science, Shahrood University of Technology, Shahrood, Iran
| |
Collapse
|
1814
|
Park HJ, Jeong OY, Chun SH, Cheon YH, Kim M, Kim S, Lee SI. Butyrate Improves Skin/Lung Fibrosis and Intestinal Dysbiosis in Bleomycin-Induced Mouse Models. Int J Mol Sci 2021; 22:ijms22052765. [PMID: 33803282 PMCID: PMC7967124 DOI: 10.3390/ijms22052765] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disorder characterized by fibrosis of the skin and internal organs. Despite several studies on SSc treatments, effective treatments for SSc are still lacking. Since evidence suggests an association between intestinal microbiota and SSc, we focused on butyrate, which has beneficial effects in autoimmune diseases as a bacterial metabolite. Here, we investigated the therapeutic potential of sodium butyrate (SB) using a bleomycin-induced fibrosis mouse model of SSc and human dermal fibroblasts (HDFs). SB attenuated bleomycin-induced dermal and lung fibrosis in mice. SB influenced fecal microbiota composition (phyla Actinobacteria and Bacteroidetes, genera Bifidobacterium and Ruminococcus_g2). SB controlled macrophage differentiation in mesenteric lymph nodes, spleen, and bronchoalveolar lavage cells of mice with bleomycin-induced skin fibrosis. Profibrotic and proinflammatory gene expression was suppressed by SB administration in skin. Furthermore, SB inhibited transforming growth factor β1-responsive proinflammatory expression with increased acetylation of histone 3 in HDFs. Subcutaneous SB application had antifibrogenic effects on the skin. Butyrate ameliorated skin and lung fibrosis by improving anti-inflammatory activity in a mouse model of SSc. Butyrate may exhibit indirect and direct anti-fibrogenic action on fibroblasts by regulating macrophage differentiation and inhibition of histone deacetylase 3. These findings suggest butyrate as an SSc treatment.
Collapse
Affiliation(s)
- Hee Jin Park
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
| | - Ok-Yi Jeong
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
| | - Sung Hak Chun
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
| | - Yun Hong Cheon
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
| | - Mingyo Kim
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
| | - Suhee Kim
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
- Correspondence: (S.K.); (S.-I.L.)
| | - Sang-Il Lee
- Department of Internal Medicine and Institute of Health Science, College of Medicine, Gyeongsang National University and Hospital, Jinju 52727, Korea; (H.J.P.); (O.-Y.J.); (S.H.C.); (Y.H.C.); (M.K.)
- Department of Convergence Medical Science, Gyeongsang National University, Jinju 52727, Korea
- Correspondence: (S.K.); (S.-I.L.)
| |
Collapse
|
1815
|
Guo M, Peng J, Huang X, Xiao L, Huang F, Zuo Z. Gut Microbiome Features of Chinese Patients Newly Diagnosed with Alzheimer's Disease or Mild Cognitive Impairment. J Alzheimers Dis 2021; 80:299-310. [PMID: 33523001 DOI: 10.3233/jad-201040] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Patients with Alzheimer's disease (AD) have gut microbiome alterations compared with healthy controls. However, previous studies often assess AD patients who have been on medications or other interventions for the disease. Also, simultaneous determination of gut microbiome in patients with mild cognitive impairment (MCI) or AD in a study is rare. OBJECTIVE To determine whether there was a gut microbiome alteration in patients newly diagnosed with AD or MCI and whether the degree of gut microbiome alteration was more severe in patients with AD than patients with MCI. METHODS Fecal samples of 18 patients with AD, 20 patients with MCI, and 18 age-matched healthy controls were collected in the morning for 16S ribosomal RNA sequencing. No patient had medications or interventions for AD or MCI before the samples were collected. RESULTS Although there was no difference in the microbial α-diversity among the three groups, patients with AD or MCI had increased β-diversity compared with healthy controls. Patients with AD had decreased Bacteroides, Lachnospira, and Ruminiclostridium_9 and increased Prevotella at the genus level compared with healthy controls. The changing direction of these genera in patients with MCI was the same as patients with AD. However, Lachnospira was the only genus whose abundance in patients with MCI was statistically significantly lower than healthy controls. Bacteroides, Lachnospira, and Ruminiclostridium_9 were positively associated with better cognitive functions whereas Prevotella was on the contrary when subjects of all three groups were considered. The negative correlation of Prevotella with cognitive functions remained among patients with MCI. CONCLUSION Patients newly diagnosed with AD or MCI have gut dysbiosis that includes the decrease of potentially protective microbiome, such as Bacteroides, and the increase of microbiome that can promote inflammation, such as Prevotella. Our results support a novel idea that the degree of gut dysbiosis is worsened with the disease stage from MCI to AD.
Collapse
Affiliation(s)
- Mingyan Guo
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Peng
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyan Huang
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lingjun Xiao
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fenyan Huang
- Department of Rehabilitation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia Health System, Charlottesville, VA, USA
| |
Collapse
|
1816
|
Alterations in the gut microbiota and metabolic profiles coincide with intestinal damage in mice with a bloodborne Candida albicans infection. Microb Pathog 2021; 154:104826. [PMID: 33689815 DOI: 10.1016/j.micpath.2021.104826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/26/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022]
Abstract
Candida albicans is an opportunistic fungus that can threaten life especially in patients with candidemia. The morbidity and mortality of candidemia originating from a central venous catheter (CVC) and illicit intravenous drug use (IVDU) are increasing. However, the mechanism underlying the bloodborne C. albicans infection remains unclear. Herein, we evaluated the gut microbiome, metabolites and intestinal mucosa by constructing the mouse models with candidemia. Model mice were injected with C. albicans via tail vein. Control mice underwent sham procedures. We observed basic life characteristics, intestinal damage-related alterations using hematoxylin and eosin (H&E) staining, intestinal tight junction protein levels, and intestinal permeability in these mice. Fecal samples were analyzed by performing 16S rRNA gene sequencing of the microbiota and LC-MS metabolomics to reveal the perturbations in intestinal flora and metabolism exacerbating intestinal damage. Weight loss, a decreased survival rate, C. albicans infection spread, and colonic epithelial damage occurred in the model group. Furthermore, the intestinal flora abundance was reduced. Several probiotics, such as Lactobacillus, and butyrate-producing bacteria, including Roseburia, Lachnospiraceae, and Clostridia, were depleted, and some pathogenic bacteria, such as Escherichia-Shigella and Proteus, belonging to the Proteobacteria phylum, and the inflammation mediators Ruminococcus and Parabacteroides were enriched in model mice. Multiple differentially altered metabolic pathways were observed and mainly related to bile acid, arachidonic acid, bile secretion, and arachidonic acid metabolism. This study illustrated the effects of a bloodborne C. albicans on the intestinal microbiota, metabolites, and intestinal barrier, which may provide new insights into tests or treatments for candidemia originating from CVC or IVDU.
Collapse
|
1817
|
Nuzzo A, Saha S, Berg E, Jayawickreme C, Tocker J, Brown JR. Expanding the drug discovery space with predicted metabolite-target interactions. Commun Biol 2021; 4:288. [PMID: 33674782 PMCID: PMC7935942 DOI: 10.1038/s42003-021-01822-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolites produced in the human gut are known modulators of host immunity. However, large-scale identification of metabolite-host receptor interactions remains a daunting challenge. Here, we employed computational approaches to identify 983 potential metabolite-target interactions using the Inflammatory Bowel Disease (IBD) cohort dataset of the Human Microbiome Project 2 (HMP2). Using a consensus of multiple machine learning methods, we ranked metabolites based on importance to IBD, followed by virtual ligand-based screening to identify possible human targets and adding evidence from compound assay, differential gene expression, pathway enrichment, and genome-wide association studies. We confirmed known metabolite-target pairs such as nicotinic acid-GPR109a or linoleoyl ethanolamide-GPR119 and inferred interactions of interest including oleanolic acid-GABRG2 and alpha-CEHC-THRB. Eleven metabolites were tested for bioactivity in vitro using human primary cell-types. By expanding the universe of possible microbial metabolite-host protein interactions, we provide multiple drug targets for potential immune-therapies.
Collapse
Affiliation(s)
- Andrea Nuzzo
- GlaxoSmithKline Pharma R&D, 1250 S. Collegeville Rd, Collegeville, PA, 19426-0989, USA.
| | - Somdutta Saha
- GlaxoSmithKline Pharma R&D, 1250 S. Collegeville Rd, Collegeville, PA, 19426-0989, USA
- EMD Serono Research & Development Institute, Inc. 45A Middlesex Turnpike, Billerica, MA, 01821, USA
| | - Ellen Berg
- Eurofins Discovery, 111 Anza Boulevard, Burlingame, CA, 94010, USA
| | - Channa Jayawickreme
- GlaxoSmithKline Pharma R&D, 1250 S. Collegeville Rd, Collegeville, PA, 19426-0989, USA
| | - Joel Tocker
- GlaxoSmithKline Pharma R&D, 1250 S. Collegeville Rd, Collegeville, PA, 19426-0989, USA
| | - James R Brown
- GlaxoSmithKline Pharma R&D, 1250 S. Collegeville Rd, Collegeville, PA, 19426-0989, USA.
- Kaleido Biosciences, Inc. 65 Hayden Avenue, Lexington, MA, 02421, USA.
| |
Collapse
|
1818
|
Host genetics exerts lifelong effects upon hindgut microbiota and its association with bovine growth and immunity. ISME JOURNAL 2021; 15:2306-2321. [PMID: 33649551 PMCID: PMC8319427 DOI: 10.1038/s41396-021-00925-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 01/26/2021] [Accepted: 02/03/2021] [Indexed: 12/22/2022]
Abstract
The gut microbiota is a complex ecological community that plays multiple critical roles within a host. Known intrinsic and extrinsic factors affect gut microbiota structure, but the influence of host genetics is understudied. To investigate the role of host genetics upon the gut microbiota structure, we performed a longitudinal study in which we evaluated the hindgut microbiota and its association with animal growth and immunity across life. We evaluated three different growth stages in an Angus-Brahman multibreed population with a graduated spectrum of genetic variation, raised under variable environmental conditions and diets. We found the gut microbiota structure was changed significantly during growth when preweaning, and fattening calves experienced large variations in diet and environmental changes. However, regardless of the growth stage, we found gut microbiota is significantly influenced by breed composition throughout life. Host genetics explained the relative abundances of 52.2%, 40.0%, and 37.3% of core bacterial taxa at the genus level in preweaning, postweaning, and fattening calves, respectively. Sutterella, Oscillospira, and Roseburia were consistently associated with breed composition at these three growth stages. Especially, butyrate-producing bacteria, Roseburia and Oscillospira, were associated with nine single-nucleotide polymorphisms (SNPs) located in genes involved in the regulation of host immunity and metabolism in the hindgut. Furthermore, minor allele frequency analysis found breed-associated SNPs in the short-chain fatty acids (SCFAs) receptor genes that promote anti-inflammation and enhance intestinal epithelial barrier functions. Our findings provide evidence of dynamic and lifelong host genetic effects upon gut microbiota, regardless of growth stages. We propose that diet, environmental changes, and genetic components may explain observed variation in critical hindgut microbiota throughout life.
Collapse
|
1819
|
Chleilat F, Schick A, Reimer RA. Microbiota Changes in Fathers Consuming a High Prebiotic Fiber Diet Have Minimal Effects on Male and Female Offspring in Rats. Nutrients 2021; 13:820. [PMID: 33801321 PMCID: PMC8001975 DOI: 10.3390/nu13030820] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/25/2021] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Consuming a diet high in prebiotic fiber has been associated with improved metabolic and gut microbial parameters intergenerationally, although studies have been limited to maternal intake with no studies examining this effect in a paternal model. METHOD Male Sprague Dawley rats were allocated to either (1) control or (2) oligofructose-supplemented diet for nine weeks and then mated. Offspring consumed control diet until 16 weeks of age. Bodyweight, body composition, glycemia, hepatic triglycerides, gastrointestinal hormones, and gut microbiota composition were measured in fathers and offspring. RESULTS Paternal energy intake was reduced, while satiety inducing peptide tyrosine tyrosine (PYY) gut hormone was increased in prebiotic versus control fathers. Increased serum PYY persisted in female prebiotic adult offspring. Hepatic triglycerides were decreased in prebiotic fathers with a similar trend (p = 0.07) seen in female offspring. Gut microbial composition showed significantly reduced alpha diversity in prebiotic fathers at 9 and 12 weeks of age (p < 0.001), as well as concurrent differences in beta diversity (p < 0.001), characterized by differences in Bifidobacteriaceae, Lactobacillaceae and Erysipelotrichaceae, and particularly Bifidobacterium animalis. Female prebiotic offspring had higher alpha diversity at 3 and 9 weeks of age (p < 0.002) and differences in beta diversity at 15 weeks of age (p = 0.04). Increases in Bacteroidetes in female offspring and Christensenellaceae in male offspring were seen at nine weeks of age. CONCLUSIONS Although paternal prebiotic intake before conception improves metabolic and microbiota outcomes in fathers, effects on offspring were limited with increased serum satiety hormone levels and changes to only select gut bacteria.
Collapse
Affiliation(s)
- Faye Chleilat
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Alana Schick
- International Microbiome Centre, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Raylene A. Reimer
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
- Department of Biochemistry & Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
1820
|
Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends Microbiol 2021; 29:700-712. [PMID: 33674141 DOI: 10.1016/j.tim.2021.02.001] [Citation(s) in RCA: 519] [Impact Index Per Article: 129.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/31/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Our ancestral diet consisted of much more nondigestible fiber than that of many societies today. Thus, from an evolutionary perspective the human genome and its physiological and nutritional requirements are not well aligned to modern dietary habits. Fiber reaching the colon is anaerobically fermented by the gut bacteria, which produce short-chain fatty acids (SCFAs) as metabolic by-products. SCFAs play a role in intestinal homeostasis, helping to explain why changes in the microbiota can contribute to the pathophysiology of human diseases. Recent research has shown that SCFAs can also have effects on tissues and organs beyond the gut, through their circulation in the blood. SCFAs not only signal through binding to cognate G-protein-coupled receptors on endocrine and immune cells in the body but also induce epigenetic changes in the genome through effects on the activity of histone acetylase and histone deacetylase enzymes. Furthermore, epigenetic imprinting likely occurs in utero, highlighting the importance of the maternal diet in early life. Here we review current understanding of how SCFAs impact on human and animal physiology and discuss the potential applications of SCFAs in the prevention and treatment of human diseases.
Collapse
|
1821
|
Identification of bioactive polysaccharide from Pseudostellaria heterophylla with its anti-inflammatory effects. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104353] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
1822
|
Alemao CA, Budden KF, Gomez HM, Rehman SF, Marshall JE, Shukla SD, Donovan C, Forster SC, Yang IA, Keely S, Mann ER, El Omar EM, Belz GT, Hansbro PM. Impact of diet and the bacterial microbiome on the mucous barrier and immune disorders. Allergy 2021; 76:714-734. [PMID: 32762040 DOI: 10.1111/all.14548] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/10/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022]
Abstract
The prevalence of chronic immune and metabolic disorders is increasing rapidly. In particular, inflammatory bowel diseases, obesity, diabetes, asthma and chronic obstructive pulmonary disease have become major healthcare and economic burdens worldwide. Recent advances in microbiome research have led to significant discoveries of associative links between alterations in the microbiome and health, as well as these chronic supposedly noncommunicable, immune/metabolic disorders. Importantly, the interplay between diet, microbiome and the mucous barrier in these diseases has gained significant attention. Diet modulates the mucous barrier via alterations in gut microbiota, resulting in either disease onset/exacerbation due to a "poor" diet or protection against disease with a "healthy" diet. In addition, many mucosa-associated disorders possess a specific gut microbiome fingerprint associated with the composition of the mucous barrier, which is further influenced by host-microbiome and inter-microbial interactions, dietary choices, microbe immigration and antimicrobials. Our review focuses on the interactions of diet (macronutrients and micronutrients), gut microbiota and mucous barriers (gastrointestinal and respiratory tract) and their importance in the onset and/or progression of major immune/metabolic disorders. We also highlight the key mechanisms that could be targeted therapeutically to prevent and/or treat these disorders.
Collapse
Affiliation(s)
- Charlotte A. Alemao
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Kurtis F. Budden
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Henry M. Gomez
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Saima F. Rehman
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Jacqueline E. Marshall
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| | - Shakti D. Shukla
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
| | - Chantal Donovan
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| | - Samuel C. Forster
- Department of Molecular and Translational Sciences Hudson Institute of Medical Research Centre for Innate Immunity and Infectious Diseases Monash University Clayton VIC Australia
| | - Ian A. Yang
- Thoracic Program The Prince Charles Hospital Metro North Hospital and Health Service Brisbane QLD Australia
- Faculty of Medicine UQ Thoracic Research Centre The University of Queensland Brisbane QLD Australia
| | - Simon Keely
- Hunter Medical Research Institute Priority Research Centre for Digestive Health and Neurogastroenterology University of Newcastle New Lambton Heights NSW Australia
| | - Elizabeth R. Mann
- Lydia Becker Institute of Immunology and Inflammation University of Manchester Manchester UK
- Faculty of Biology Medicine and Health Manchester Collaborative Centre for Inflammation Research Manchester Academic Health Science Centre University of Manchester Manchester UK
| | - Emad M. El Omar
- St George & Sutherland Clinical School Microbiome Research Centre University of New South Wales Sydney NSW Australia
| | - Gabrielle T. Belz
- Diamantina Institute University of Queensland Woolloongabba QLD Australia
- Department of Medical Biology Walter and Eliza Hall Institute of Medical Research University of Melbourne Parkville VIC Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs Hunter Medical Research Institute New Lambton, Newcastle NSW Australia
- The University of Newcastle Newcastle NSW Australia
- Faculty of Science Centre for Inflammation Centenary Institute University of Technology Sydney Sydney NSW Australia
| |
Collapse
|
1823
|
Saint-Criq V, Lugo-Villarino G, Thomas M. Dysbiosis, malnutrition and enhanced gut-lung axis contribute to age-related respiratory diseases. Ageing Res Rev 2021; 66:101235. [PMID: 33321253 DOI: 10.1016/j.arr.2020.101235] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/23/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022]
Abstract
Older people are at an increased risk of developing respiratory diseases such as chronic obstructive pulmonary diseases, asthma, idiopathic pulmonary fibrosis or lung infections. Susceptibility to these diseases is partly due to the intrinsic ageing process, characterized by genomic, cellular and metabolic hallmarks and immunosenescence, and is associated with changes in the intestinal microbiota. Importantly, in the lungs, ageing is also associated with a dysbiosis and loss of resilience of the resident microbiota and alterations of the gut-lung axis. Notably, as malnutrition is often observed in the elderly, nutrition is one of the most accessible modifiable factors affecting both senescence and microbiota. This article reviews the changes affecting the lung and its resident microbiota during ageing, as well as the interconnections between malnutrition, senescence, microbiota, gut-lung axis and respiratory health. As the communication along the gut-lung axis becomes more permissive with ageing, this review also explores the evidence that the gut and lung microbiota are key players in the maintenance of healthy lungs, and as such, are potential targets for nutrition-based preventive strategies against lung disease in elderly populations.
Collapse
|
1824
|
Yang K, Hou Y, Zhang Y, Liang H, Sharma A, Zheng W, Wang L, Torres R, Tatebe K, Chmura SJ, Pitroda SP, Gilbert JA, Fu YX, Weichselbaum RR. Suppression of local type I interferon by gut microbiota-derived butyrate impairs antitumor effects of ionizing radiation. J Exp Med 2021; 218:e20201915. [PMID: 33496784 PMCID: PMC7844434 DOI: 10.1084/jem.20201915] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/02/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022] Open
Abstract
The antitumor effects of ionizing radiation (IR) are mediated in part through activation of innate and adaptive immunity. Here we report that gut microbiota influences tumor control following IR. Vancomycin decreased the abundance of butyrate-producing gut bacteria and enhanced antitumor responses to IR. Oral administration of Lachnospiraceae, a family of vancomycin-sensitive bacteria, was associated with increased systemic and intratumoral butyric acid levels and impaired the efficacy of IR in germ-free (GF) mice. Local butyrate inhibited STING-activated type I IFN expression in dendritic cells (DCs) through blockade of TBK1 and IRF3 phosphorylation, which abrogated IR-induced tumor-specific cytotoxic T cell immune responses without directly protecting tumor cells from radiation. Our findings demonstrate that the selective targeting of butyrate-producing microbiota may provide a novel therapeutic option to enhance tumor radiation sensitivity.
Collapse
Affiliation(s)
- Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Yuzhu Hou
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Yuan Zhang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Hua Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Anukriti Sharma
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA
| | - Wenxin Zheng
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Rolando Torres
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
| | - Ken Tatebe
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
| | - Steven J. Chmura
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
| | - Sean P. Pitroda
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| | - Jack A. Gilbert
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
- Scripps Institution of Oceanography, University of California, San Diego, La Jolla, CA
| | - Yang-Xin Fu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, IL
- The Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL
| |
Collapse
|
1825
|
Lamousé-Smith E, Kelly D, De Cremoux I. Designing bugs as drugs: exploiting the gut microbiome. Am J Physiol Gastrointest Liver Physiol 2021; 320:G295-G303. [PMID: 33264062 PMCID: PMC8609565 DOI: 10.1152/ajpgi.00381.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The extensive investigation of the human microbiome and the accumulating evidence regarding its critical relationship to human health and disease has advanced recognition of its potential as the next frontier of drug development. The rapid development of technologies, directed at understanding the compositional and functional dynamics of the human microbiome, and the ability to mine for novel therapeutic targets and biomarkers are leading innovative efforts to develop microbe-derived drugs that can prevent and treat autoimmune, metabolic, and infectious diseases. Increasingly, academics, biotechs, investors, and large pharmaceutical companies are partnering to collectively advance various therapeutic modalities ranging from live bacteria to small molecules. We review the leading platforms in current development focusing on live microbial consortia, engineered microbes, and microbial-derived metabolites. We will also touch on how the field is addressing and challenging the traditional definitions of pharmacokinetics and pharmacodynamics, dosing, toxicity, and safety to advance the development of these novel and cutting-edge therapeutics into the clinic.
Collapse
|
1826
|
Quin C, Ghosh S, Dai C, Barnett JA, Garner AM, Yoo RKH, Zandberg WF, Botta A, Gorzelak MA, Gibson DL. Maternal Intake of Dietary Fat Pre-Programs Offspring's Gut Ecosystem Altering Colonization Resistance and Immunity to Infectious Colitis in Mice. Mol Nutr Food Res 2021; 65:e2000635. [PMID: 33559319 DOI: 10.1002/mnfr.202000635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 01/02/2021] [Indexed: 11/10/2022]
Abstract
SCOPE The transgenerational impact of dietary fat remains unclear. Here, the role of maternal fat consumption as a modulator of gut microbial communities and infectious disease outcomes in their offspring is explored. METHODS AND RESULTS C57BL/6 mice are fed isocaloric high-fat diets throughout breeding, gestation and lactation. Diets contained either milk fat (MF), olive oil (OO) or corn oil (CO), with or without fish oil. The pups born to maternally exposed mice are weaned on to chow and raised into adulthood. At 8 weeks, the offsprings are either euthanized for colonic 16S rRNA analysis or challenged with the enteric pathogen, Citrobacter rodentium. Maternal CO exposure resulted in unique clustering of bacterial communities in offspring compared with MF and OO. Diets rich in CO reduced survival in offspring challenged with C. rodentium. The addition of fish oil did not improve mortality caused by CO and worsened disease outcomes when combined with OO. Unlike the unsaturated diets, MF is protective with and without fish oil. CONCLUSIONS Overall, these data reveal that maternal intake of fatty acids do have transgenerational impacts on their offspring's bacteriome and enteric infection risk. Based on this study, saturated fats should be included in maternal diets.
Collapse
Affiliation(s)
- Candice Quin
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Sanjoy Ghosh
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Chuanbin Dai
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Jacqueline A Barnett
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Alexander M Garner
- Department of Chemistry Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Rachael K H Yoo
- Department of Chemistry Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Wesley F Zandberg
- Department of Chemistry Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Amy Botta
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Monika A Gorzelak
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
| | - Deanna L Gibson
- Department of Biology Okanagan campus, University of British Columbia, Kelowna, British Columbia, Canada
- Department of Medicine, Faculty of Medicine, University of British Columbia, Kelowna, British Columbia, Canada
| |
Collapse
|
1827
|
Wang G, Zhong D, Liu H, Yang T, Liang Q, Wang J, Zhang R, Zhang Y. Water soluble dietary fiber from walnut meal as a prebiotic in preventing metabolic syndrome. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
1828
|
Wong MK, Barbulescu P, Coburn B, Reguera-Nuñez E. Therapeutic interventions and mechanisms associated with gut microbiota-mediated modulation of immune checkpoint inhibitor responses. Microbes Infect 2021; 23:104804. [PMID: 33652120 DOI: 10.1016/j.micinf.2021.104804] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023]
Abstract
The link between the gut microbiome and responsiveness to immune checkpoint inhibitor (ICI) therapy is now well established. New therapeutic opportunities exploiting this relationship are being developed with the goal of augmenting ICI efficacy. In this review, we summarize the foundational research establishing these interactions and discuss the mechanisms and novel therapeutic options associated with this gut microbiome-ICI connection.
Collapse
Affiliation(s)
- Matthew K Wong
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Philip Barbulescu
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Bryan Coburn
- Department of Immunology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada; Department of Medicine, Division of Infectious Diseases, University Health Network, Toronto, M5G 0A3, Canada.
| | - Elaine Reguera-Nuñez
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, M5G 0A3, Canada.
| |
Collapse
|
1829
|
Baldi S, Menicatti M, Nannini G, Niccolai E, Russo E, Ricci F, Pallecchi M, Romano F, Pedone M, Poli G, Renzi D, Taddei A, Calabrò AS, Stingo FC, Bartolucci G, Amedei A. Free Fatty Acids Signature in Human Intestinal Disorders: Significant Association between Butyric Acid and Celiac Disease. Nutrients 2021; 13:742. [PMID: 33652681 PMCID: PMC7996737 DOI: 10.3390/nu13030742] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/12/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Altered circulating levels of free fatty acids (FFAs), namely short chain fatty acids (SCFAs), medium chain fatty acids (MCFAs), and long chain fatty acids (LCFAs), are associated with metabolic, gastrointestinal, and malignant diseases. Hence, we compared the serum FFA profile of patients with celiac disease (CD), adenomatous polyposis (AP), and colorectal cancer (CRC) to healthy controls (HC). We enrolled 44 patients (19 CRC, 9 AP, 16 CD) and 16 HC. We performed a quantitative FFA evaluation with the gas chromatography-mass spectrometry method (GC-MS), and we performed Dirichlet-multinomial regression in order to highlight disease-specific FFA signature. HC showed a different composition of FFAs than CRC, AP, and CD patients. Furthermore, the partial least squares discriminant analysis (PLS-DA) confirmed perfect overlap between the CRC and AP patients and separation of HC from the diseased groups. The Dirichlet-multinomial regression identified only strong positive association between CD and butyric acid. Moreover, CD patients showed significant interactions with age, BMI, and gender. In addition, among patients with the same age and BMI, being male compared to being female implies a decrease of the CD effect on the (log) prevalence of butyric acid in FFA composition. Our data support GC-MS as a suitable method for the concurrent analysis of circulating SCFAs, MCFAs, and LCFAs in different gastrointestinal diseases. Furthermore, and notably, we suggest for the first time that butyric acid could represent a potential biomarker for CD screening.
Collapse
Affiliation(s)
- Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (S.B.); (G.N.); (E.N.); (E.R.); (A.T.)
| | - Marta Menicatti
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, 50139 Florence, Italy; (M.M.); (M.P.); (G.B.)
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (S.B.); (G.N.); (E.N.); (E.R.); (A.T.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (S.B.); (G.N.); (E.N.); (E.R.); (A.T.)
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (S.B.); (G.N.); (E.N.); (E.R.); (A.T.)
| | - Federica Ricci
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, 50134 Florence, Italy; (F.R.); (D.R.); (A.S.C.)
| | - Marco Pallecchi
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, 50139 Florence, Italy; (M.M.); (M.P.); (G.B.)
| | - Francesca Romano
- Laboratory of Clinical Pathology, Versilia Hospital, 55041 Lido di Camaiore, Italy;
| | - Matteo Pedone
- Department of Statistics, Computer Science, Applications “G. Parenti”, 50134 Florence, Italy; (M.P.); (G.P.); (F.C.S.)
| | - Giovanni Poli
- Department of Statistics, Computer Science, Applications “G. Parenti”, 50134 Florence, Italy; (M.P.); (G.P.); (F.C.S.)
| | - Daniela Renzi
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, 50134 Florence, Italy; (F.R.); (D.R.); (A.S.C.)
| | - Antonio Taddei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (S.B.); (G.N.); (E.N.); (E.R.); (A.T.)
| | - Antonino S. Calabrò
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio” University of Florence, 50134 Florence, Italy; (F.R.); (D.R.); (A.S.C.)
| | - Francesco C. Stingo
- Department of Statistics, Computer Science, Applications “G. Parenti”, 50134 Florence, Italy; (M.P.); (G.P.); (F.C.S.)
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences University of Florence, 50139 Florence, Italy; (M.M.); (M.P.); (G.B.)
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (S.B.); (G.N.); (E.N.); (E.R.); (A.T.)
- SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50134 Florence, Italy
| |
Collapse
|
1830
|
Interplay between severities of COVID-19 and the gut microbiome: implications of bacterial co-infections? Gut Pathog 2021; 13:14. [PMID: 33632296 PMCID: PMC7906082 DOI: 10.1186/s13099-021-00407-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/16/2021] [Indexed: 01/07/2023] Open
Abstract
COVID-19 is an acute respiratory distress syndrome and is often accompanied by gastrointestinal symptoms. The SARS-CoV-2 has been traced not only in nasopharyngeal and mid-nasal swabs but also in stool and rectal swabs of COVID-19 patients. The gut microbiota is important for an effective immune response as it ensures that unfavorable immune reactions in lungs and other vital organs are regulated. The human gut-lung microbiota interplay provides a framework for therapies in the treatment and management of several pulmonary diseases and infections. Here, we have collated data from COVID-19 studies, which suggest that bacterial co-infections as well as the gut-lung cross talk may be important players in COVID-19 disease prognosis. Our analyses suggests a role of gut microbiome in pathogen infections as well as in an array of excessive immune reactions during and post COVID-19 infection recovery period.
Collapse
|
1831
|
Huang Y, Yang Q, Mi X, Qiu L, Tao X, Zhang Z, Xia J, Wu Q, Wei H. Ripened Pu-erh Tea Extract Promotes Gut Microbiota Resilience against Dextran Sulfate Sodium Induced Colitis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:2190-2203. [PMID: 33570405 DOI: 10.1021/acs.jafc.0c07537] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Ripened Pu-erh tea (RPT) has been shown to be an effective natural ingredient to defend against experimentally induced colitis. We hypothesized that RPT would alleviate dextran sulfate sodium (DSS) induced colitis via modulating intestinal microbiota. The effect of RPT on mice gut microbiota was evaluated using 16S rRNA gene amplicon sequencing, broad-spectrum antibiotic (ABX) treatment, and fecal microbiota transplantation (FMT). Pretreatment with RPT enhanced intestinal barrier function, reduced colonic and serum proinflammatory cytokine and macrophage infiltration, and preserved the resilience of gut microbiota in mice during a DSS challenge. Administration of either RPT-regulated or healthy control-derived gut microbiota showed similar protection against colitis, and such protection could not be recapitulated with fecal microbiota from ABX-treated mice, suggesting a key role of protective consortium in the disease protection. Mechanistically, cecal contents of short-chain fatty acids (SCFAs) and colonic peroxisome proliferator activated receptor-γ (PPAR-γ) expression in colitis mice increased significantly by RPT intervention. Collectively, RPT treatment improved DSS-induced colitis by partially reversing the dysbiosis state of gut microbiota, which might be associated with an increase in SCFA level and PPAR-γ expression.
Collapse
Affiliation(s)
- Yina Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| | - Qin Yang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| | - Xuan Mi
- Wanlongshan Tea Plantation, Pingxiang, Jiangxi 337000, People's Republic of China
| | - Liang Qiu
- Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, People's Republic of China
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| | - Zhihong Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
| | - Jun Xia
- Veterinary Medicine and Feed Supervision Institute, Nanchang, Jiangxi 330096, People's Republic of China
| | - Qinglong Wu
- Wanlongshan Tea Plantation, Pingxiang, Jiangxi 337000, People's Republic of China
- Texas Children's Microbiome Center, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi 330031, People's Republic of China
- Wanlongshan Tea Plantation, Pingxiang, Jiangxi 337000, People's Republic of China
| |
Collapse
|
1832
|
Cao Y, Liu Y, Dong Q, Wang T, Niu C. Alterations in the gut microbiome and metabolic profile in rats acclimated to high environmental temperature. Microb Biotechnol 2021; 15:276-288. [PMID: 33620148 PMCID: PMC8719808 DOI: 10.1111/1751-7915.13772] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/26/2021] [Accepted: 01/30/2021] [Indexed: 12/29/2022] Open
Abstract
Heat acclimation (HA) is the best strategy to improve heat stress tolerance by inducing positive physiological adaptations. Evidence indicates that the gut microbiome plays a fundamental role in the development of HA, and modulation of gut microbiota can improve tolerance to heat exposure and decrease the risks of heat illness. In this study, for the first time, we applied 16S rRNA gene sequencing and untargeted liquid chromatography–mass spectrometry (LC‐MS) metabolomics to explore variations in the gut microbiome and faecal metabolic profiles in rats after HA. The gut microbiota of HA subjects exhibited higher diversity and richer microbes. HA altered the gut microbiota composition with significant increases in the genera Lactobacillus (a major probiotic) and Oscillospira alongside significant decreases in the genera Blautia and Allobaculum. The faecal metabolome was also significantly changed after HA, and among the 13 perturbed metabolites, (S)‐AL 8810 and celastrol were increased. Moreover, the two increased genera were positively correlated with the two upregulated metabolites and negatively correlated with the other 11 downregulated metabolites, while the correlations between the two decreased genera and the upregulated/downregulated metabolites were completely contrary. In summary, both the structure of the gut microbiome community and the faecal metabolome were improved after 28 days of HA. These findings provide novel insights regarding the improvement of the gut microbiome and its functions as a potential mechanism by which HA confers protection against heat stress.
Collapse
Affiliation(s)
- Yang Cao
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Ying Liu
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Qingyang Dong
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Tao Wang
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| | - Chao Niu
- Department of Environmental Medicine, Tianjin Institute of Environmental and Operational Medicine, Tianjin, 300050, China
| |
Collapse
|
1833
|
Zhang N, Li C, Niu Z, Kang H, Wang M, Zhang B, Tian H. Colonization and immunoregulation of Lactobacillus plantarum BF_15, a novel probiotic strain from the feces of breast-fed infants. Food Funct 2021; 11:3156-3166. [PMID: 32207765 DOI: 10.1039/c9fo02745a] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immunosuppression is a manifestation imbalance in the immune system, often during unhealthy states. In recent years, lactic acid bacteria (LAB) have been found to be important components of the body's innate immune system, and indispensable to maintaining normal immune function. Lactobacillus plantarum BF_15, a novel strain isolated from the feces of breast-fed infants, which has shown potential as an immunomodulator in vitro. In the present study, with the Polymerase Chain Reaction-Denaturing Gradient Gel Electrophoresis (PCR-DGGE) based on RNA-polymerase beta subunit encoding gene (rpoB) to analyze the colonization of L. plantarum BF_15 in the intestine of mice. In addition, Lactobacillus rhamnosus GG (LGG) as a positive control strain, by measuring immune-related indexes and the diversity of intestinal microbiota, the effects of BF_15 on immunoregulation and intestinal microbiota dysbiosis were investigated to elucidate whether the attenuation of immunosuppression is related to the modulation of intestinal microbiota. Results did indeed support this notion that BF_15 did colonize murine intestines well, in which it could still be detected in mice feces 14 days after stopping the probiotic administration. Moreover, BF_15 found to protect mice against reduction in the levels of several immune-related indicators, including the thymus and spleen indexes, splenic lymphocyte proliferation, toe swelling degree, serum hemolysin-antibody level, and macrophage phagocytosis index, triggered by high-dose (200 mg kg-1) intraperitoneal administration of cyclophosphamide (CTX). In addition, the strain was also found to effectively balance intestinal microbiota dysbiosis in the mice. Collectively, these results indicated that L. plantarum BF_15 can not only successfully colonize murine intestines, but also can effectively alleviate CTX-induced immunosuppression, once established, by rebalancing the intestinal microbiota. This, therefore, provides strong evidence for the view that BF_15 has the potential to become a highly effective immunomodulating probiotic in human microbiota as well.
Collapse
Affiliation(s)
- Na Zhang
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China. and College of Biochemistry and Environmental Engineering, Baoding University, Baoding, Hebei 071000, China
| | - Chen Li
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Zhihua Niu
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Hongyan Kang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, Hebei 071000, China
| | - Miaoshu Wang
- New Hope Tensun (Hebei) Dairy Co., Ltd, Baoding, Hebei 071000, China
| | - Bo Zhang
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China.
| | - Hongtao Tian
- College of Food Science and Technology, Agricultural University of Hebei, Baoding, Hebei 071000, China. and National Engineering Research Center for Agriculture in Northern Mountainous Areas, Baoding, Hebei 071000, China
| |
Collapse
|
1834
|
Thavamani A, Salem I, Sferra TJ, Sankararaman S. Impact of Altered Gut Microbiota and Its Metabolites in Cystic Fibrosis. Metabolites 2021; 11:123. [PMID: 33671639 PMCID: PMC7926988 DOI: 10.3390/metabo11020123] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is the most common lethal, multisystemic genetic disorder in Caucasians. Mutations in the gene encoding the cystic fibrosis transmembrane regulator (CFTR) protein are responsible for impairment of epithelial anionic transport, leading to impaired fluid regulation and pH imbalance across multiple organs. Gastrointestinal (GI) manifestations in CF may begin in utero and continue throughout the life, resulting in a chronic state of an altered intestinal milieu. Inherent dysfunction of CFTR leads to dysbiosis of the gut. This state of dysbiosis is further perpetuated by acquired factors such as use of antibiotics for recurrent pulmonary exacerbations. Since the gastrointestinal microbiome and their metabolites play a vital role in nutrition, metabolic, inflammatory, and immune functions, the gut dysbiosis will in turn impact various manifestations of CF-both GI and extra-GI. This review focuses on the consequences of gut dysbiosis and its metabolic implications on CF disease and possible ways to restore homeostasis.
Collapse
Affiliation(s)
- Aravind Thavamani
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| | - Iman Salem
- Center for Medial Mycology, Case Western Reserve University School of Medicine, UH Cleveland Medical Center, Cleveland, OH 44106, USA;
| | - Thomas J. Sferra
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| | - Senthilkumar Sankararaman
- Department of Pediatrics, Division of Pediatric Gastroenterology, UH Rainbow Babies & Children’s Hospital, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA; (A.T.); (T.J.S.)
| |
Collapse
|
1835
|
Pane S, Sacco A, Iorio A, Romani L, Putignani L. Strongyloides stercoralis Infestation in a Child: How a Nematode Can Affect Gut Microbiota. Int J Mol Sci 2021; 22:ijms22042131. [PMID: 33669932 PMCID: PMC7924877 DOI: 10.3390/ijms22042131] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 02/17/2021] [Accepted: 02/19/2021] [Indexed: 12/15/2022] Open
Abstract
Background: Strongyloidiasis is a neglected tropical disease caused by the intestinal nematode Strongyloides stercoralis and characterized by gastrointestinal and pulmonary involvement. We report a pediatric case of strongyloidiasis to underline the response of the host microbiota to the perturbation induced by the nematode. Methods: We performed a 16S rRNA-metagenomic analysis of the gut microbiota of a 7-year-old female during and after S. stercolaris infection, investigating three time-point of stool samples’ ecology: T0- during parasite infection, T1- a month after parasite infection, and T2- two months after parasite infection. Targeted-metagenomics were used to investigate ecology and to predict the functional pathways of the gut microbiota. Results: an increase in the alpha-diversity indices in T0-T1 samples was observed compared to T2 and healthy controls (CTRLs). Beta-diversity analysis showed a shift in the relative abundance of specific gut bacterial species from T0 to T2 samples. Moreover, the functional prediction of the targeted-metagenomics profiles suggested an enrichment of microbial glycan and carbohydrate metabolisms in the T0 sample compared with CTRLs. Conclusions: The herein report reinforces the literature suggestion of a putative direct or immune-mediated ability of S. stercolaris to promote the increase in bacterial diversity.
Collapse
Affiliation(s)
- Stefania Pane
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.P.); (A.S.)
| | - Anna Sacco
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (S.P.); (A.S.)
| | - Andrea Iorio
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Lorenza Romani
- Immunology and Infectious Diseases Unit, University Department of Pediatrics, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Lorenza Putignani
- Department of Diagnostic and Laboratory Medicine, Unit of Parasitology and Multimodal Laboratory Medicine Research Area, Unit of Human Microbiome, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
- Correspondence: ; Tel.: +39-06-6859-4127
| |
Collapse
|
1836
|
Wipperman MF, Bhattarai SK, Vorkas CK, Maringati VS, Taur Y, Mathurin L, McAulay K, Vilbrun SC, Francois D, Bean J, Walsh KF, Nathan C, Fitzgerald DW, Glickman MS, Bucci V. Gastrointestinal microbiota composition predicts peripheral inflammatory state during treatment of human tuberculosis. Nat Commun 2021; 12:1141. [PMID: 33602926 PMCID: PMC7892575 DOI: 10.1038/s41467-021-21475-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/21/2021] [Indexed: 12/15/2022] Open
Abstract
The composition of the gastrointestinal microbiota influences systemic immune responses, but how this affects infectious disease pathogenesis and antibiotic therapy outcome is poorly understood. This question is rarely examined in humans due to the difficulty in dissociating the immunologic effects of antibiotic-induced pathogen clearance and microbiome alteration. Here, we analyze data from two longitudinal studies of tuberculosis (TB) therapy (35 and 20 individuals) and a cross sectional study from 55 healthy controls, in which we collected fecal samples (for microbiome analysis), sputum (for determination of Mycobacterium tuberculosis (Mtb) bacterial load), and peripheral blood (for transcriptomic analysis). We decouple microbiome effects from pathogen sterilization by comparing standard TB therapy with an experimental TB treatment that did not reduce Mtb bacterial load. Random forest regression to the microbiome-transcriptome-sputum data from the two longitudinal datasets reveals that renormalization of the TB inflammatory state is associated with Mtb pathogen clearance, increased abundance of Clusters IV and XIVa Clostridia, and decreased abundance of Bacilli and Proteobacteria. We find similar associations when applying machine learning to peripheral gene expression and microbiota profiling in the independent cohort of healthy individuals. Our findings indicate that antibiotic-induced reduction in pathogen burden and changes in the microbiome are independently associated with treatment-induced changes of the inflammatory response of active TB, and the response to antibiotic therapy may be a combined effect of pathogen killing and microbiome driven immunomodulation. Antibiotic therapy can lead to pathogen clearance, but also to alterations in the gut microbiota and systemic immune responses. Here, the authors analyze data from patients with tuberculosis and healthy subjects to show that pathogen clearance and gut microbiota alterations are independently associated with antibiotic-induced changes of the inflammatory response of active tuberculosis.
Collapse
Affiliation(s)
- Matthew F Wipperman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Clinical and Translational Science Center, Weill Cornell Medicine, New York, NY, USA
| | - Shakti K Bhattarai
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Charles Kyriakos Vorkas
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA
| | - Venkata Suhas Maringati
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ying Taur
- Division of Infectious Diseases, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laurent Mathurin
- Haitian Study Group for Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | | | - Stalz Charles Vilbrun
- Haitian Study Group for Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Daphie Francois
- Haitian Study Group for Kaposi's Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - James Bean
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kathleen F Walsh
- Center for Global Health, Weill Cornell Medicine, New York, NY, USA
| | - Carl Nathan
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, NY, USA
| | | | - Michael S Glickman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA. .,Division of Infectious Diseases, Weill Cornell Medicine, New York, NY, USA. .,Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, NY, USA.
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, USA. .,Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA, USA. .,Program in Systems Biology, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
1837
|
Resistant Starch Type 2 from Wheat Reduces Postprandial Glycemic Response with Concurrent Alterations in Gut Microbiota Composition. Nutrients 2021; 13:nu13020645. [PMID: 33671147 PMCID: PMC7922998 DOI: 10.3390/nu13020645] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 01/03/2023] Open
Abstract
The majority of research on the physiological effects of dietary resistant starch type 2 (RS2) has focused on sources derived from high-amylose maize. In this study, we conduct a double-blind, randomized, placebo-controlled, crossover trial investigating the effects of RS2 from wheat on glycemic response, an important indicator of metabolic health, and the gut microbiota. Overall, consumption of RS2-enriched wheat rolls for one week resulted in reduced postprandial glucose and insulin responses relative to conventional wheat when participants were provided with a standard breakfast meal containing the respective treatment rolls (RS2-enriched or conventional wheat). This was accompanied by an increase in the proportions of bacterial taxa Ruminococcus and Gemmiger in the fecal contents, reflecting the composition in the distal intestine. Additionally, fasting breath hydrogen and methane were increased during RS2-enriched wheat consumption. However, although changes in fecal short-chain fatty acid (SCFA) concentrations were not significant between control and RS-enriched wheat roll consumption, butyrate and total SCFAs were positively correlated with relative abundance of Faecalibacterium, Ruminoccocus, Roseburia, and Barnesiellaceae. These effects show that RS2-enriched wheat consumption results in a reduction in postprandial glycemia, altered gut microbial composition, and increased fermentation activity relative to wild-type wheat.
Collapse
|
1838
|
Daliri EBM, Ofosu FK, Chelliah R, Lee BH, Oh DH. Challenges and Perspective in Integrated Multi-Omics in Gut Microbiota Studies. Biomolecules 2021; 11:300. [PMID: 33671370 PMCID: PMC7922017 DOI: 10.3390/biom11020300] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/10/2021] [Accepted: 02/14/2021] [Indexed: 12/14/2022] Open
Abstract
The advent of omic technology has made it possible to identify viable but unculturable micro-organisms in the gut. Therefore, application of multi-omic technologies in gut microbiome studies has become invaluable for unveiling a comprehensive interaction between these commensals in health and disease. Meanwhile, despite the successful identification of many microbial and host-microbial cometabolites that have been reported so far, it remains difficult to clearly identify the origin and function of some proteins and metabolites that are detected in gut samples. However, the application of single omic techniques for studying the gut microbiome comes with its own challenges which may be overcome if a number of different omics techniques are combined. In this review, we discuss our current knowledge about multi-omic techniques, their challenges and future perspective in this field of gut microbiome studies.
Collapse
Affiliation(s)
- Eric Banan-Mwine Daliri
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 200-701, Korea; (E.B.-M.D.); (F.K.O.); (R.C.)
| | - Fred Kwame Ofosu
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 200-701, Korea; (E.B.-M.D.); (F.K.O.); (R.C.)
| | - Ramachandran Chelliah
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 200-701, Korea; (E.B.-M.D.); (F.K.O.); (R.C.)
| | - Byong H. Lee
- SportBiomics, Sacramento Inc., California, CA 95660, USA;
| | - Deog-Hwan Oh
- Department of Food Science and Biotechnology, Kangwon National University, Chuncheon 200-701, Korea; (E.B.-M.D.); (F.K.O.); (R.C.)
| |
Collapse
|
1839
|
Lo Sasso G, Khachatryan L, Kondylis A, Battey JND, Sierro N, Danilova NA, Grigoryeva TV, Markelova MI, Khusnutdinova DR, Laikov AV, Salafutdinov II, Romanova YD, Siniagina MN, Vasiliev IY, Boulygina EA, Solovyeva VV, Garanina EE, Kitaeva KV, Ivanov KY, Chulpanova DS, Kletenkov KS, Valeeva AR, Odintsova AK, Ardatskaya MD, Abdulkhakov RA, Ivanov NV, Peitsch MC, Hoeng J, Abdulkhakov SR. Inflammatory Bowel Disease-Associated Changes in the Gut: Focus on Kazan Patients. Inflamm Bowel Dis 2021; 27:418-433. [PMID: 32766755 PMCID: PMC7885336 DOI: 10.1093/ibd/izaa188] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Several studies have highlighted the role of host-microbiome interactions in the pathogenesis of inflammatory bowel disease (IBD), resulting in an increasing amount of data mainly focusing on Western patients. Because of the increasing prevalence of IBD in newly industrialized countries such as those in Asia, the Middle East, and South America, there is mounting interest in elucidating the gut microbiota of these populations. We present a comprehensive analysis of several IBD-related biomarkers and gut microbiota profiles and functions of a unique population of patients with IBD and healthy patients from Kazan (Republic of Tatarstan, Russia). METHODS Blood and fecal IBD biomarkers, serum cytokines, and fecal short-chain fatty acid (SCFA) content were profiled. Finally, fecal microbiota composition was analyzed by 16S and whole-genome shotgun sequencing. RESULTS Fecal microbiota whole-genome sequencing confirmed the presence of classic IBD dysbiotic features at the phylum level, with increased abundance of Proteobacteria, Actinobacteria, and Fusobacteria and decreased abundance of Firmicutes, Bacteroidetes, and Verrucomicrobia. At the genus level, the abundance of both fermentative (SCFA-producing and hydrogen (H2)-releasing) and hydrogenotrophic (H2-consuming) microbes was affected in patients with IBD. This imbalance was confirmed by the decreased abundance of SCFA species in the feces of patients with IBD and the change in anaerobic index, which mirrors the redox status of the intestine. CONCLUSIONS Our analyses highlighted how IBD-related dysbiotic microbiota-which are generally mainly linked to SCFA imbalance-may affect other important metabolic pathways, such as H2 metabolism, that are critical for host physiology and disease development.
Collapse
Affiliation(s)
- Giuseppe Lo Sasso
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Lusine Khachatryan
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Athanasios Kondylis
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - James N D Battey
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Nicolas Sierro
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Natalia A Danilova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Tatiana V Grigoryeva
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Maria I Markelova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Dilyara R Khusnutdinova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Alexander V Laikov
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Ilnur I Salafutdinov
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Yulia D Romanova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Mariia N Siniagina
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Ilya Yu Vasiliev
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Eugenia A Boulygina
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Valeriya V Solovyeva
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Ekaterina E Garanina
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Kristina V Kitaeva
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Konstantin Y Ivanov
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Darja S Chulpanova
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Konstantin S Kletenkov
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| | - Alina R Valeeva
- Department of Clinical Immunology and Allergology, Kazan State Medical University, Kazan, Tatarstan, Russian Federation
| | - Alfiya Kh Odintsova
- Department of Gastroenterology, Republican Clinical Hospital of Tatarstan Republic, Kazan, Tatarstan, Russian Federation
| | - Maria D Ardatskaya
- Central State Medical Academy of Administrative Department of the President of the Russian Federation, Moscow, Russian Federation
| | - Rustam A Abdulkhakov
- Department of Clinical Immunology and Allergology, Kazan State Medical University, Kazan, Tatarstan, Russian Federation
| | - Nikolai V Ivanov
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Manuel C Peitsch
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Julia Hoeng
- Philip Morris International Research and Development, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Sayar R Abdulkhakov
- Kazan Federal University, Institute of Fundamental Medicine and Biology, Kazan, Tatarstan, Russian Federation
| |
Collapse
|
1840
|
Yip W, Hughes MR, Li Y, Cait A, Hirst M, Mohn WW, McNagny KM. Butyrate Shapes Immune Cell Fate and Function in Allergic Asthma. Front Immunol 2021; 12:628453. [PMID: 33659009 PMCID: PMC7917140 DOI: 10.3389/fimmu.2021.628453] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/25/2021] [Indexed: 12/19/2022] Open
Abstract
The microbiome plays a fundamental role in how the immune system develops and how inflammatory responses are shaped and regulated. The “gut-lung axis” is a relatively new term that highlights a crucial biological crosstalk between the intestinal microbiome and lung. A growing body of literature suggests that dysbiosis, perturbation of the gut microbiome, is a driving force behind the development, and severity of allergic asthma. Animal models have given researchers new insights into how gut microbe-derived components and metabolites, such as short-chain fatty acids (SCFAs), influence the development of asthma. While the full understanding of how SCFAs influence allergic airway disease remains obscure, a recurring theme of epigenetic regulation of gene expression in several immune cell compartments is emerging. This review will address our current understanding of how SCFAs, and specifically butyrate, orchestrates cell behavior, and epigenetic changes and will provide a detailed overview of the effects of these modifications on immune cells in the context of allergic airway disease.
Collapse
Affiliation(s)
- William Yip
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.,The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Michael R Hughes
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.,The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Yicong Li
- The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada
| | - Alissa Cait
- Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - Martin Hirst
- Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada.,Michael Smith Laboratories, The University of British Columbia, Vancouver, BC, Canada
| | - William W Mohn
- Life Sciences Institute, The University of British Columbia, Vancouver, BC, Canada.,Department of Microbiology and Immunology, The University of British Columbia, Vancouver, BC, Canada
| | - Kelly M McNagny
- School of Biomedical Engineering, The University of British Columbia, Vancouver, BC, Canada.,The Biomedical Research Centre, The University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
1841
|
Yu K, Choi I, Yun CH. Immunosecurity: immunomodulants enhance immune responses in chickens. Anim Biosci 2021; 34:321-337. [PMID: 33705619 PMCID: PMC7961195 DOI: 10.5713/ab.20.0851] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
The global population has increased with swift urbanization in developing countries, and it is likely to result in a high demand for animal-derived protein-rich foods. Animal farming has been constantly affected by various stressful conditions, which can be categorized into physical, environmental, nutritional, and biological factors. Such conditions could be exacerbated by banning on the use of antibiotics as a growth promoter together with a pandemic situation including, but not limited to, African swine fever, avian influenza, and foot-and-mouth disease. To alleviate these pervasive tension, various immunomodulants have been suggested as alternatives for antibiotics. Various studies have investigated how stressors (i.e., imbalanced nutrition, dysbiosis, and disease) could negatively affect nutritional physiology in chickens. Importantly, the immune system is critical for host protective activity against pathogens, but at the same time excessive immune responses negatively affect its productivity. Yet, comprehensive review articles addressing the impact of such stress factors on the immune system of chickens are scarce. In this review, we categorize these stressors and their effects on the immune system of chickens and attempt to provide immunomodulants which can be a solution to the aforementioned problems facing the chicken industry.
Collapse
Affiliation(s)
- Keesun Yu
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Inhwan Choi
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea
| | - Cheol-Heui Yun
- Department of Agricultural Biotechnology and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul 08826, Korea.,Institute of Green-Bio Science and Technology, Seoul National University, Pyeongchang 25354, Korea.,Center for Food Bioconvergence, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
1842
|
Millman JF, Okamoto S, Teruya T, Uema T, Ikematsu S, Shimabukuro M, Masuzaki H. Extra-virgin olive oil and the gut-brain axis: influence on gut microbiota, mucosal immunity, and cardiometabolic and cognitive health. Nutr Rev 2021; 79:1362-1374. [PMID: 33576418 PMCID: PMC8581649 DOI: 10.1093/nutrit/nuaa148] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Extra-virgin olive oil (EVOO), a popular functional food and major source of fat in the Mediterranean diet, possesses a variety of healthful components, including monounsaturated fatty acids and bioactive phenolic compounds that, individually and collectively, exert beneficial effects on cardiometabolic markers of health and act as neuroprotective agents through their anti-inflammatory and antioxidant activities. The gut microbiota and health of the intestinal environment are now considered important factors in the development of obesity, metabolic disease, and even certain neurodegenerative conditions via the gut-brain axis. Recently, data are emerging which demonstrate that the health-promoting benefits of EVOO may also extend to the gut microbiota. In this review, we aimed to examine findings from recent studies regarding the impact of EVOO on gut microbiota and intestinal health and explore how modulations in composition of gut microbiota, production of microbially produced products, and activity and functioning of the mucosal immune system may lead to favorable outcomes in cardiovascular, metabolic, and cognitive health.
Collapse
Affiliation(s)
- Jasmine F Millman
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Shiki Okamoto
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Taiki Teruya
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Tsugumi Uema
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Shinya Ikematsu
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Michio Shimabukuro
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| | - Hiroaki Masuzaki
- J.F. Millman, S. Okamoto, T. Taiki, T. Uema, and H. Masuzaki are with the Division of Endocrinology, Diabetes and Metabolism, Hematology, Rheumatology (Second Department of Internal Medicine), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. S. Ikematsu is with the Department of Bioresources Engineering, National Institute of Technology, Okinawa College, Okinawa, Japan. M. Shimabukuro is with the Department of Diabetes, Endocrinology and Metabolism, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
1843
|
Carranza-Naval MJ, Vargas-Soria M, Hierro-Bujalance C, Baena-Nieto G, Garcia-Alloza M, Infante-Garcia C, del Marco A. Alzheimer's Disease and Diabetes: Role of Diet, Microbiota and Inflammation in Preclinical Models. Biomolecules 2021; 11:biom11020262. [PMID: 33578998 PMCID: PMC7916805 DOI: 10.3390/biom11020262] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Epidemiological studies show the association between AD and type 2 diabetes (T2DM), although the mechanisms are not fully understood. Dietary habits and lifestyle, that are risk factors in both diseases, strongly modulate gut microbiota composition. Also, the brain-gut axis plays a relevant role in AD, diabetes and inflammation, through products of bacterial metabolism, like short-chain fatty acids. We provide a comprehensive review of current literature on the relation between dysbiosis, altered inflammatory cytokines profile and microglia in preclinical models of AD, T2DM and models that reproduce both diseases as commonly observed in the clinic. Increased proinflammatory cytokines, such as IL-1β and TNF-α, are widely detected. Microbiome analysis shows alterations in Actinobacteria, Bacteroidetes or Firmicutes phyla, among others. Altered α- and β-diversity is observed in mice depending on genotype, gender and age; therefore, alterations in bacteria taxa highly depend on the models and approaches. We also review the use of pre- and probiotic supplements, that by favoring a healthy microbiome ameliorate AD and T2DM pathologies. Whereas extensive studies have been carried out, further research would be necessary to fully understand the relation between diet, microbiome and inflammation in AD and T2DM.
Collapse
Affiliation(s)
- Maria Jose Carranza-Naval
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Salus Infirmorum, Universidad de Cadiz, 11005 Cadiz, Spain
| | - Maria Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
| | - Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
| | - Gloria Baena-Nieto
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Department of Endocrinology, Jerez Hospital, Jerez de la Frontera, 11407 Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
| | - Carmen Infante-Garcia
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Correspondence: (C.I.-G.); (A.d.M.)
| | - Angel del Marco
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Correspondence: (C.I.-G.); (A.d.M.)
| |
Collapse
|
1844
|
Kurilenko N, Fatkhullina AR, Mazitova A, Koltsova EK. Act Locally, Act Globally-Microbiota, Barriers, and Cytokines in Atherosclerosis. Cells 2021; 10:cells10020348. [PMID: 33562334 PMCID: PMC7915371 DOI: 10.3390/cells10020348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is characterized by the formation and progressive growth of atherosclerotic plaques in the wall of arteries. Atherosclerosis is a major predisposing factor for stroke and heart attack. Various immune-mediated mechanisms are implicated in the disease initiation and progression. Cytokines are key mediators of the crosstalk between innate and adaptive immune cells as well as non-hematopoietic cells in the aortic wall and are emerging players in the regulation of atherosclerosis. Progression of atherosclerosis is always associated with increased local and systemic levels of pro-inflammatory cytokines. The role of cytokines within atherosclerotic plaque has been extensively investigated; however, the cell-specific role of cytokine signaling, particularly the role of cytokines in the regulation of barrier tissues tightly associated with microbiota in the context of cardiovascular diseases has only recently come to light. Here, we summarize the knowledge about the function of cytokines at mucosal barriers and the interplay between cytokines, barriers, and microbiota and discuss their known and potential implications for atherosclerosis development.
Collapse
Affiliation(s)
- Natalia Kurilenko
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
| | | | - Aleksandra Mazitova
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
| | - Ekaterina K. Koltsova
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
- Correspondence:
| |
Collapse
|
1845
|
Bauset C, Gisbert-Ferrándiz L, Cosín-Roger J. Metabolomics as a Promising Resource Identifying Potential Biomarkers for Inflammatory Bowel Disease. J Clin Med 2021; 10:jcm10040622. [PMID: 33562024 PMCID: PMC7915257 DOI: 10.3390/jcm10040622] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a relapsing chronic disorder of the gastrointestinal tract characterized by disruption of epithelial barrier function and excessive immune response to gut microbiota. The lack of biomarkers providing early diagnosis or defining the status of the pathology difficulties an accurate assessment of the disease. Given the different metabolomic profiles observed in IBD patients, metabolomics may reveal prime candidates to be studied, which may help in understanding the pathology and identifying novel therapeutic targets. In this review, we summarize the most current advances describing the promising metabolites such as lipids or amino acids found through untargeted metabolomics from serum, faecal, urine and biopsy samples.
Collapse
Affiliation(s)
- Cristina Bauset
- Department of Pharmacology and CIBER, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.B.); (L.G.-F.)
| | - Laura Gisbert-Ferrándiz
- Department of Pharmacology and CIBER, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain; (C.B.); (L.G.-F.)
| | - Jesús Cosín-Roger
- Hospital Dr. Peset, Fundación para la Investigación Sanitaria y Biomédica de la Comunitat Valenciana, FISABIO, 46017 Valencia, Spain
- Correspondence: ; Tel.: +34-963851234
| |
Collapse
|
1846
|
Banfi D, Moro E, Bosi A, Bistoletti M, Cerantola S, Crema F, Maggi F, Giron MC, Giaroni C, Baj A. Impact of Microbial Metabolites on Microbiota-Gut-Brain Axis in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:1623. [PMID: 33562721 PMCID: PMC7915037 DOI: 10.3390/ijms22041623] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The complex bidirectional communication system existing between the gastrointestinal tract and the brain initially termed the "gut-brain axis" and renamed the "microbiota-gut-brain axis", considering the pivotal role of gut microbiota in sustaining local and systemic homeostasis, has a fundamental role in the pathogenesis of Inflammatory Bowel Disease (IBD). The integration of signals deriving from the host neuronal, immune, and endocrine systems with signals deriving from the microbiota may influence the development of the local inflammatory injury and impacts also more distal brain regions, underlying the psychophysiological vulnerability of IBD patients. Mood disorders and increased response to stress are frequently associated with IBD and may affect the disease recurrence and severity, thus requiring an appropriate therapeutic approach in addition to conventional anti-inflammatory treatments. This review highlights the more recent evidence suggesting that alterations of the microbiota-gut-brain bidirectional communication axis may concur to IBD pathogenesis and sustain the development of both local and CNS symptoms. The participation of the main microbial-derived metabolites, also defined as "postbiotics", such as bile acids, short-chain fatty acids, and tryptophan metabolites in the development of IBD-associated gut and brain dysfunction will be discussed. The last section covers a critical evaluation of the main clinical evidence pointing to the microbiome-based therapeutic approaches for the treatment of IBD-related gastrointestinal and neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Davide Banfi
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Elisabetta Moro
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Annalisa Bosi
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Michela Bistoletti
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Silvia Cerantola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti 2, 35131 Padova, Italy; (S.C.); (M.C.G.)
| | - Francesca Crema
- Department of Internal Medicine and Therapeutics, Section of Pharmacology, University of Pavia, via Ferrata 9, 27100 Pavia, Italy; (E.M.); (F.C.)
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| | - Maria Cecilia Giron
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Largo Meneghetti 2, 35131 Padova, Italy; (S.C.); (M.C.G.)
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
- Centre of Neuroscience, University of Insubria, 21100 Varese, Italy
| | - Andreina Baj
- Department of Medicine and Surgery, University of Insubria, via H Dunant 5, 21100 Varese, Italy; (D.B.); (A.B.); (M.B.); (F.M.); (A.B.)
| |
Collapse
|
1847
|
Basson AR, Chen C, Sagl F, Trotter A, Bederman I, Gomez-Nguyen A, Sundrud MS, Ilic S, Cominelli F, Rodriguez-Palacios A. Regulation of Intestinal Inflammation by Dietary Fats. Front Immunol 2021; 11:604989. [PMID: 33603741 PMCID: PMC7884479 DOI: 10.3389/fimmu.2020.604989] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
With the epidemic of human obesity, dietary fats have increasingly become a focal point of biomedical research. Epidemiological studies indicate that high-fat diets (HFDs), especially those rich in long-chain saturated fatty acids (e.g., Western Diet, National Health Examination survey; NHANES 'What We Eat in America' report) have multi-organ pro-inflammatory effects. Experimental studies have confirmed some of these disease associations, and have begun to elaborate mechanisms of disease induction. However, many of the observed effects from epidemiological studies appear to be an over-simplification of the mechanistic complexity that depends on dynamic interactions between the host, the particular fatty acid, and the rather personalized genetics and variability of the gut microbiota. Of interest, experimental studies have shown that certain saturated fats (e.g., lauric and myristic fatty acid-rich coconut oil) could exert the opposite effect; that is, desirable anti-inflammatory and protective mechanisms promoting gut health by unanticipated pathways. Owing to the experimental advantages of laboratory animals for the study of mechanisms under well-controlled dietary settings, we focus this review on the current understanding of how dietary fatty acids impact intestinal biology. We center this discussion on studies from mice and rats, with validation in cell culture systems or human studies. We provide a scoping overview of the most studied diseases mechanisms associated with the induction or prevention of Inflammatory Bowel Disease in rodent models relevant to Crohn's Disease and Ulcerative Colitis after feeding either high-fat diet (HFD) or feed containing specific fatty acid or other target dietary molecule. Finally, we provide a general outlook on areas that have been largely or scarcely studied, and assess the effects of HFDs on acute and chronic forms of intestinal inflammation.
Collapse
Affiliation(s)
- Abigail R. Basson
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Christy Chen
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Filip Sagl
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Ashley Trotter
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Department of Hospital Medicine, Pritzker School of Medicine, NorthShore University Health System, Chicago, IL, United States
| | - Ilya Bederman
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Adrian Gomez-Nguyen
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Mark S. Sundrud
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, FL, United States
| | - Sanja Ilic
- Department of Human Sciences, Human Nutrition, College of Education and Human Ecology, The Ohio State University, Columbus, OH, United States
| | - Fabio Cominelli
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| | - Alex Rodriguez-Palacios
- Division of Gastroenterology and Liver Diseases, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Digestive Health Research Institute, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Cleveland Digestive Diseases Research Core, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
- Digestive Health Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
- University Hospitals Research and Education Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
1848
|
Chen KJ, Chen YL, Ueng SH, Hwang TL, Kuo LM, Hsieh PW. Neutrophil elastase inhibitor (MPH-966) improves intestinal mucosal damage and gut microbiota in a mouse model of 5-fluorouracil-induced intestinal mucositis. Biomed Pharmacother 2021; 134:111152. [PMID: 33373916 DOI: 10.1016/j.biopha.2020.111152] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 12/03/2020] [Accepted: 12/14/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND 5-Fluorouracil (5-FU)-based chemotherapy is first-line chemotherapy for colorectal cancer. However, 5-FU-induced intestinal mucositis (FUIIM) is a common adverse effect that severely impairs drug tolerance and results in poor patient health. METHODS Male C57BL/6 mice were given 5-FU (50 mg/kg/day, i.p.) and treated with MPH-966 (5 and 7.5 mg/kg/day, p.o.) for five days. The body weight loss and the amount of food intake, and histopathological findings were recorded and analyzed. In addition, the neutrophil infiltration, levels of neutrophil serine proteases and pro-inflammatory cytokines, and tight junction proteins expression in intestinal tissues were determined. The ecology of gut microbiota was performed through next-generation sequencing technologies. RESULTS Neutrophil elastase (NE) overexpression is a key feature in FUIIM. This study showed that treatment with the specific NE inhibitor MPH-966 (7.5 mg/kg/day, p.o.) significantly reversed 5-FU-induced loss in body weight and food intake; reversed villous atrophy; significantly suppressed myeloperoxidase, NE, and proteinase 3 activity; and reduced pro-inflammatory cytokine expression in an FUIIM mouse model. In addition, MPH-966 prevented 5-FU-induced intestinal barrier dysfunction, as indicated by the modulated expression of the tight junction proteins zonula occludin-1 and occludin. MPH-966 also reversed 5-FU-induced changes in gut microbiota diversity and abundances, specifically the Firmicutes-to-Bacteroidetes ratio; Muribaculaceae, Ruminococcaceae, and Eggerthellaceae abundances at the family level; and Candidatus Arthromitus abundance at the genus level. CONCLUSION These data indicate that NE inhibitor is a key treatment candidate to alleviate FUIIM by regulating abnormal inflammatory responses, intestinal barrier dysfunction, and gut microbiota imbalance.
Collapse
Affiliation(s)
- Kung-Ju Chen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Li Chen
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shir-Hwa Ueng
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan; Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan
| | - Liang-Mou Kuo
- Department of General Surgery, Chang Gung Memorial Hospital, Chiayi, 613, Taiwan.
| | - Pei-Wen Hsieh
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Natural Products, School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Linkou, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
1849
|
Gao Y, Ding M, Dong X, Zhang J, Kursat Azkur A, Azkur D, Gan H, Sun Y, Fu W, Li W, Liang H, Cao Y, Yan Q, Cao C, Gao H, Brüggen M, Veen W, Sokolowska M, Akdis M, Akdis CA. Risk factors for severe and critically ill COVID-19 patients: A review. Allergy 2021; 76:428-455. [PMID: 33185910 DOI: 10.1111/all.14657] [Citation(s) in RCA: 852] [Impact Index Per Article: 213.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023]
Abstract
The pandemic of coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused an unprecedented global social and economic impact, and high numbers of deaths. Many risk factors have been identified in the progression of COVID-19 into a severe and critical stage, including old age, male gender, underlying comorbidities such as hypertension, diabetes, obesity, chronic lung diseases, heart, liver and kidney diseases, tumors, clinically apparent immunodeficiencies, local immunodeficiencies, such as early type I interferon secretion capacity, and pregnancy. Possible complications include acute kidney injury, coagulation disorders, thoromboembolism. The development of lymphopenia and eosinopenia are laboratory indicators of COVID-19. Laboratory parameters to monitor disease progression include lactate dehydrogenase, procalcitonin, high-sensitivity C-reactive protein, proinflammatory cytokines such as interleukin (IL)-6, IL-1β, Krebs von den Lungen-6 (KL-6), and ferritin. The development of a cytokine storm and extensive chest computed tomography imaging patterns are indicators of a severe disease. In addition, socioeconomic status, diet, lifestyle, geographical differences, ethnicity, exposed viral load, day of initiation of treatment, and quality of health care have been reported to influence individual outcomes. In this review, we highlight the scientific evidence on the risk factors of severity of COVID-19.
Collapse
Affiliation(s)
- Ya‐dong Gao
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Mei Ding
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Xiang Dong
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Jin‐jin Zhang
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Ahmet Kursat Azkur
- Department of Virology Faculty of Veterinary Medicine University of Kirikkale Kirikkale Turkey
| | - Dilek Azkur
- Division of Pediatric Allergy and Immunology Department of Pediatrics Faculty of Medicine University of Kirikkale Kirikkale Turkey
| | - Hui Gan
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Yuan‐li Sun
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Wei Fu
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Wei Li
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Hui‐ling Liang
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Yi‐yuan Cao
- Department of Radiology Zhongnan Hospital of Wuhan University Wuhan China
| | - Qi Yan
- Department of Geriatrics Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Huazhong China
| | - Can Cao
- Department of Allergology Zhongnan Hospital of Wuhan University Wuhan China
| | - Hong‐yu Gao
- Department of Geriatrics Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Huazhong China
| | - Marie‐Charlotte Brüggen
- Department of Dermatology University Hospital Zurich Zurich Switzerland
- Faculty of Medicine University of Zurich Zurich Switzerland
- Hochgebirgsklinik Davos Christine Kühne‐Center for Allergy Research and Education Davos Switzerland
| | - Willem Veen
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| | - Cezmi A. Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF) University of Zurich Zurich Switzerland
| |
Collapse
|
1850
|
Ma Y, Zhang Q, Liu W, Chen Z, Zou C, Fu L, Wang Y, Liu Y. Preventive Effect of Depolymerized Sulfated Galactans from Eucheuma serra on Enterotoxigenic Escherichia coli-Caused Diarrhea via Modulating Intestinal Flora in Mice. Mar Drugs 2021; 19:80. [PMID: 33535475 PMCID: PMC7912752 DOI: 10.3390/md19020080] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
In this work, the preventive effect of depolymerized sulfated polysaccharides from Eucheuma serra (DESP) on bacterial diarrhea by regulating intestinal flora was investigated in vivo. Based on the enterotoxigenic Escherichia coli (ETEC)-infected mouse diarrhea model, DESP at doses ranging from 50 mg/kg to 200 mg/kg alleviated weight loss and decreased the diarrhea rate and diarrhea index. Serological tests showed that the levels of inflammation-related factors were effectively suppressed. Furthermore, the repaired intestinal mucosa was verified by morphology and pathological tissue section observations. Compared with the model group, the richness and diversity of the intestinal flora in the DESP group increased according to the 16S rRNA high-throughput sequencing of the gut microbiota. Specifically, Firmicutes and Actinobacteria increased, and Proteobacteria decreased after DESP administration. At the family level, DESP effectively improved the abundance of Lactobacillaceae, Bifidobacteriaceae, and Lachnospiraceae, while significantly inhibiting the growth of Enterobacteriaceae. Therefore, the antimicrobial diarrhea function of DESP may be related to the regulation of intestinal microbiota.
Collapse
Affiliation(s)
- Yu Ma
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
| | - Qian Zhang
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
| | - Wenqiang Liu
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
| | - Zhaohua Chen
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
| | - Chao Zou
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
| | - Linglin Fu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China;
| | - Yanbo Wang
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, China;
| | - Yixiang Liu
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China; (Y.M.); (Q.Z.); (W.L.); (Z.C.); (C.Z.); (Y.W.)
| |
Collapse
|