1
|
Zhu F, Sun K, Zhang H, Lu J, Guo P, Zhang J, Xu Y, Lyu B. Comparative Analyses of Lycodon rufozonatus and Lycodon rosozonatus Gut Microbiota in Different Regions. Ecol Evol 2024; 14:e70480. [PMID: 39440211 PMCID: PMC11495892 DOI: 10.1002/ece3.70480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/27/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024] Open
Abstract
The interactions between hosts and the gut microbiota are intricate and can significantly affect the ecology and evolution of both parties. Various host traits, including taxonomy, diet, social behaviour, and external factors such as prey availability and the local environment, all play an important role in shaping composition and diversity of the gut microbiogta. In this study, we explored the impact of intestinal microorganisms on the host in adapting to their respective ecological niches in two species of snakes. We collected feces from Lycodon rufozonatus and Lycodon rosozonatus from different geographical locations and used 16S rRNA gene sequencing technology to sequence the v3-v4 region. The results revealed that there was no significant difference in the alpha diversity of intestinal microorganisms between L. rufozonatus and L. rosozonatus. The gut microbiota of all individuals comprised four main phyla: Pseudomonadota, Bacteroidota, Bacillota, and Actinomycetota. At the genus level, the genus Salmonella dominated the enterobacterial microbiota in the samples from Hainan, while there was no obvious dominant genus in the enterobacterial microbiota of the samples from the other four localities. Comparative analysis of enzyme families annotated to the gut microbiota between L. rufozonatus and L. rosozonatus from the four sampling regions by CAZy carbohydrate annotation revealed that nine enzyme families differed significantly in terms of glycoside hydrolases (GHs). In addition, we compared the composition of gut microbial communities between L. rufozonatus and L. rosozonatus and investigated the impact of the differences on their functions. Our results will provide insights into the coevolution of host and gut microbes.
Collapse
Affiliation(s)
- Fei Zhu
- School of Life SciencesGuizhou Normal UniversityGuiyangGuizhouChina
| | - Ke Sun
- School of Life SciencesGuizhou Normal UniversityGuiyangGuizhouChina
| | - He Zhang
- Guizhou Academy of ForestryGuiyangGuizhouChina
| | - Jing Lu
- School of Life SciencesGuizhou Normal UniversityGuiyangGuizhouChina
| | - Peng Guo
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinSichuanChina
| | - Jiaqi Zhang
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinSichuanChina
| | - Yu Xu
- School of Life SciencesGuizhou Normal UniversityGuiyangGuizhouChina
| | - Bing Lyu
- Faculty of Agriculture, Forestry and Food EngineeringYibin UniversityYibinSichuanChina
| |
Collapse
|
2
|
Perdijk O, Azzoni R, Marsland BJ. The microbiome: an integral player in immune homeostasis and inflammation in the respiratory tract. Physiol Rev 2024; 104:835-879. [PMID: 38059886 DOI: 10.1152/physrev.00020.2023] [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: 05/02/2023] [Revised: 11/07/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023] Open
Abstract
The last decade of microbiome research has highlighted its fundamental role in systemic immune and metabolic homeostasis. The microbiome plays a prominent role during gestation and into early life, when maternal lifestyle factors shape immune development of the newborn. Breast milk further shapes gut colonization, supporting the development of tolerance to commensal bacteria and harmless antigens while preventing outgrowth of pathogens. Environmental microbial and lifestyle factors that disrupt this process can dysregulate immune homeostasis, predisposing infants to atopic disease and childhood asthma. In health, the low-biomass lung microbiome, together with inhaled environmental microbial constituents, establishes the immunological set point that is necessary to maintain pulmonary immune defense. However, in disease perturbations to immunological and physiological processes allow the upper respiratory tract to act as a reservoir of pathogenic bacteria, which can colonize the diseased lung and cause severe inflammation. Studying these host-microbe interactions in respiratory diseases holds great promise to stratify patients for suitable treatment regimens and biomarker discovery to predict disease progression. Preclinical studies show that commensal gut microbes are in a constant flux of cell division and death, releasing microbial constituents, metabolic by-products, and vesicles that shape the immune system and can protect against respiratory diseases. The next major advances may come from testing and utilizing these microbial factors for clinical benefit and exploiting the predictive power of the microbiome by employing multiomics analysis approaches.
Collapse
Affiliation(s)
- Olaf Perdijk
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Rossana Azzoni
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J Marsland
- Department of Immunology, School of Translational Science, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
3
|
Li L, Chen J, Sun H, Niu Q, Zhao Y, Yang X, Sun Q. Orm2 Deficiency Aggravates High-Fat Diet-Induced Obesity through Gut Microbial Dysbiosis and Intestinal Inflammation. Mol Nutr Food Res 2024; 68:e2300236. [PMID: 37853937 DOI: 10.1002/mnfr.202300236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/09/2023] [Indexed: 10/20/2023]
Abstract
SCOPE Orosomucoid 2 (Orm2) is a hepatocyte-secreted protein that plays a crucial role in regulating obesity-type metabolic disease and immunity. The imbalance of gut microbiota is one of the causes of obesity, but the mechanism of the relationship between Orm2 and gut microbiota in obesity remains unclear. METHODS AND RESULTS Orm2-/- (Orm2 knockout) mice on a normal diet developed spontaneous obesity and metabolic disturbances at the 20th week. Through 16S rRNA gene sequencing, the study finds that the gut microbiota of Orm2-/- mice has a different microbial composition compared to wild type (WT) mice. Furthermore, a high-fat diet (HFD) for 16 weeks exacerbates obesity in Orm2-/- mice. Lack of Orm2 promotes dysregulation of gut microbiota under the HFD, especially a reduction of Clostridium spp. Supplementation with Clostridium butyricum alleviates obesity and alters the gut microbial composition in WT mice, but has minimal effects on Orm2-/- mice. In contrast, co-housing of Orm2-/- mice with WT mice rescues Orm2-/- obesity by reducing pathogenic bacteria and mitigating intestinal inflammation. CONCLUSION These findings suggest Orm2 deficiency exacerbates HFD-induced gut microbiota disturbance and intestinal inflammation, providing a novel insight into the complex bacterial flora but not a single probiotic administration in the therapeutic strategy of obesity.
Collapse
Affiliation(s)
- Li Li
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jionghao Chen
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Haoming Sun
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qiang Niu
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yan Zhao
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xiaojun Yang
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Qingzhu Sun
- Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, China
| |
Collapse
|
4
|
Liu J, Chen B, Jiang M, Cui T, Lv B, Fu Z, Li X, Du Y, Guo J, Zhong X, Zou Y, Zhao X, Yang W, Gao X. Polygonatum odoratum polysaccharide attenuates lipopolysaccharide-induced lung injury in mice by regulating gut microbiota. Food Sci Nutr 2023; 11:6974-6986. [PMID: 37970373 PMCID: PMC10630852 DOI: 10.1002/fsn3.3622] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/26/2023] [Accepted: 07/29/2023] [Indexed: 11/17/2023] Open
Abstract
Polygonatum odoratum is appreciated for its edible and medicinal benefits especially for lung protection. However, the contained active components have been understudied, and further research is required to fully exploit its potential application. We aimed to probe into the beneficial effects of Polygonatum odoratum polysaccharide (POP) in lipopolysaccharide-induced lung inflammatory injury mice. POP treatment could ameliorate the survival rate, pulmonary function, lung pathological lesions, and immune inflammatory response. POP treatment could repair intestinal barrier, and modulate the composition of gut microbiota, especially reducing the abundance of Klebsiella, which were closely associated with the therapeutic effects of POP. Investigation of the underlying anti-inflammatory mechanism showed that POP suppressed the generation of pro-inflammatory molecules in lung by inhibiting iNOS+ M1 macrophages. Collectively, POP is a promising multi-target microecological regulator to prevent and treat the immuno-inflammation and lung injury by modulating gut microbiota.
Collapse
Affiliation(s)
- Jia‐rui Liu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Bo‐xue Chen
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Mei‐ting Jiang
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Tian‐yi Cui
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Bin Lv
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Zhi‐fei Fu
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xue Li
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yao‐dong Du
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Jin‐he Guo
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xin‐qin Zhong
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ya‐dan Zou
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xin Zhao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Wen‐zhi Yang
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xiu‐mei Gao
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- State Key Laboratory of Component‐based Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| |
Collapse
|
5
|
Shang Y, Zhong H, Liu G, Wang X, Wu X, Wei Q, Shi L, Zhang H. Characteristics of Microbiota in Different Segments of the Digestive Tract of Lycodon rufozonatus. Animals (Basel) 2023; 13:ani13040731. [PMID: 36830518 PMCID: PMC9952230 DOI: 10.3390/ani13040731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
The gastrointestinal tract of animals contains microbiota, forming a complex microecosystem. Gut microbes and their metabolites can regulate the development of host innate and adaptive immune systems. Animal immune systems maintain intestinal symbiotic microbiota homeostasis. However, relatively few studies have been published on reptiles, particularly snakes, and even fewer studies on different parts of the digestive tracts of these animals. Herein, we used 16S rRNA gene sequencing to investigate the microbial community composition and adaptability in the stomach and small and large intestines of Lycodon rufozonatus. Proteobacteria, Bacteroidetes, and Firmicutes were most abundant in the stomach; Fusobacteria in the small intestine; and Proteobacteria, Bacteroidetes, Fusobacteria, and Firmicutes in the large intestine. No dominant genus could be identified in the stomach; however, dominant genera were evident in the small and large intestines. The microbial diversity index was significantly higher in the stomach than in the small and large intestines. Moreover, the influence of the microbial community structure on function was clarified through function prediction. Collectively, the gut microbes in the different segments of the digestive tract revealed the unique features of the L. rufozonatus gut microbiome. Our results provide insights into the co-evolutionary relationship between reptile gut microbiota and their hosts.
Collapse
Affiliation(s)
- Yongquan Shang
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
| | - Huaming Zhong
- College of Biology and Food, Shangqiu Normal University, Shangqiu 476000, China
| | - Gang Liu
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
| | - Xibao Wang
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
| | - Xiaoyang Wu
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
| | - Qinguo Wei
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
| | - Lupeng Shi
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
| | - Honghai Zhang
- College of Life Sciences, Qufu Normal University, Qufu 273165, China
- Correspondence:
| |
Collapse
|
6
|
Traina G. The Connection between Gut and Lung Microbiota, Mast Cells, Platelets and SARS-CoV-2 in the Elderly Patient. Int J Mol Sci 2022; 23:ijms232314898. [PMID: 36499222 PMCID: PMC9740794 DOI: 10.3390/ijms232314898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 11/29/2022] Open
Abstract
The human coronavirus SARS-CoV-2 or COVID-19 that emerged in late 2019 causes a respiratory tract infection and has currently resulted in more than 627 million confirmed cases and over 6.58 million deaths worldwide up to October 2022. The highest death rate caused by COVID-19 is in older people, especially those with comorbidities. This evidence presents a challenge for biomedical research on aging and also identifies some key players in inflammation, including mast cells and platelets, which could represent important markers and, at the same time, unconventional therapeutic targets. Studies have shown a decrease in the diversity of gut microbiota composition in the elderly, particularly a reduced abundance of butyrate-producing species, and COVID-19 patients manifest faecal microbiome alterations, with an increase in opportunistic pathogens and a depletion of commensal beneficial microorganisms. The main purpose of this narrative review is to highlight how an altered condition of the gut microbiota, especially in the elderly, could be an important factor and have a strong impact in the lung homeostasis and COVID-19 phenomenon, jointly to the activation of mast cells and platelets, and also affect the outcomes of the pathology. Therefore, a targeted and careful control of the intestinal microbiota could represent a complementary intervention to be implemented for the management and the challenge against COVID-19.
Collapse
Affiliation(s)
- Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, Via Romana, 06126 Perugia, Italy
| |
Collapse
|
7
|
Sharma P, Silva C, Pfreundschuh S, Ye H, Sampath H. Metabolic protection by the dietary flavonoid 7,8-dihydroxyflavone requires an intact gut microbiome. Front Nutr 2022; 9:987956. [PMID: 36061902 PMCID: PMC9428675 DOI: 10.3389/fnut.2022.987956] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/29/2022] [Indexed: 11/18/2022] Open
Abstract
Background 7,8-dihydroxyflavone (DHF) is a naturally occurring flavonoid found in Godmania, Tridax, and Primula species that confers protection against high-fat diet (HFD) induced metabolic pathologies selectively in female mice. We have previously reported that this metabolic protection is associated with early and stable remodeling of the intestinal microbiome, evident in female but not male DHF-supplemented mice. Early changes in the gut microbiome in female DHF-fed mice were highly predictive of subsequent metabolic protection, suggesting a causative association between the gut microbiome and the metabolic effects of DHF. Objective To investigate a causal association between the gut microbiome and the metabolic effects of DHF using a model of antibiotic-induced gut microbiome ablation. Materials and methods Age-matched male and female C57Bl6/J mice were given ad libitum access to HFD and drinking water containing vehicle or DHF for 12 weeks. For antibiotic (Abx) treatment, female mice were given drinking water containing a cocktail of antibiotics for 2 weeks prior to HFD feeding and throughout the feeding period. Metabolic phenotyping consisted of longitudinal assessments of body weights, body composition, food, and water intake, as well as measurement of energy expenditure, glucose tolerance, and plasma and hepatic lipids. Protein markers mediating the cellular effects of DHF were assessed in brown adipose tissue (BAT) and skeletal muscle. Results Metabolic protection conferred by DHF in female HFD-fed mice was only apparent in the presence of an intact gut microbiome. Abx-treated mice were not protected from HFD-induced obesity by DHF administration. Further, tissue activation of the tropomyosin-related kinase receptor B (TrkB) receptor, which has been attributed to the biological activity of DHF, was lost upon gut microbiome ablation, indicating a requirement for microbial “activation” of DHF for its systemic effects. In addition, we report for the first time that DHF supplementation significantly activates TrkB in BAT of female, but not male, mice uncovering a novel target tissue of DHF. DHF supplementation also increased uncoupling protein 1 (UCP1) and AMP-activated protein kinase (AMPK) protein in BAT, consistent with protection from diet-induced obesity. Conclusion These results establish for the first time a requirement for the gut microbiome in mediating the metabolic effects of DHF in female mice and uncover a novel target tissue that may mediate these sexually-dimorphic protective effects.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
| | - Camila Silva
- Department of Biotechnology, Rutgers University, New Brunswick, NJ, United States
| | - Sarah Pfreundschuh
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Hong Ye
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
| | - Harini Sampath
- Department of Nutritional Sciences, Rutgers University, New Brunswick, NJ, United States
- Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
- Center for Microbiome, Nutrition, and Health, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, United States
- *Correspondence: Harini Sampath,
| |
Collapse
|
8
|
Danenberg AH. The etiology of gut dysbiosis and its role in chronic disease. MICROBIOME, IMMUNITY, DIGESTIVE HEALTH AND NUTRITION 2022:71-91. [DOI: 10.1016/b978-0-12-822238-6.00020-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
9
|
Mathieu E, Marquant Q, Descamps D, Riffault S, Saint-Criq V, Thomas M. Le poumon est sensible aux effets locaux et à distance des microbiotes. NUTR CLIN METAB 2021. [DOI: 10.1016/j.nupar.2021.04.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
10
|
Sinha R, Ngo MD, Bartlett S, Bielefeldt-Ohmann H, Keshvari S, Hasnain SZ, Donovan ML, Kling JC, Blumenthal A, Chen C, Short KR, Ronacher K. Pre-Diabetes Increases Tuberculosis Disease Severity, While High Body Fat Without Impaired Glucose Tolerance Is Protective. Front Cell Infect Microbiol 2021; 11:691823. [PMID: 34295838 PMCID: PMC8291147 DOI: 10.3389/fcimb.2021.691823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/17/2021] [Indexed: 12/15/2022] Open
Abstract
Type 2 diabetes (T2D) is a well-known risk factor for tuberculosis (TB), but little is known about pre-diabetes and the relative contribution of impaired glucose tolerance vs. obesity towards susceptibility to TB. Here, we developed a preclinical model of pre-diabetes and TB. Mice fed a high fat diet (HFD) for 12 weeks presented with impaired glucose tolerance and hyperinsulinemia compared to mice fed normal chow diet (NCD). Infection with M. tuberculosis (Mtb) H37Rv after the onset of dysglycemia was associated with significantly increased lung pathology, lower concentrations of TNF-α, IFN-γ, IFN-β and IL-10 and a trend towards higher bacterial burden at 3 weeks post infection. To determine whether the increased susceptibility of pre-diabetic mice to TB is reversible and is associated with dysglycemia or increased body fat mass, we performed a diet reversal experiment. Pre-diabetic mice were fed a NCD for 10 additional weeks (HFD/NCD) at which point glucose tolerance was restored, but body fat mass remained higher compared to control mice that consumed NCD throughout the entire experiment (NCD/NCD). Upon Mtb infection HFD/NCD mice had significantly lower bacterial burden compared to NCD/NCD mice and this was accompanied by restored IFN-γ responses. Our findings demonstrate that pre-diabetes increases susceptibility to TB, but a high body mass index without dysglycemia is protective. This murine model offers the opportunity to further study the underlying immunological, metabolic and endocrine mechanisms of this association.
Collapse
Affiliation(s)
- Roma Sinha
- Translational Research Institute, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Minh Dao Ngo
- Translational Research Institute, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Stacey Bartlett
- Translational Research Institute, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Helle Bielefeldt-Ohmann
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre - The University of Queensland, Brisbane, QLD, Australia
| | - Sahar Keshvari
- Translational Research Institute, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Sumaira Z Hasnain
- Translational Research Institute, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre - The University of Queensland, Brisbane, QLD, Australia
| | - Meg L Donovan
- The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Jessica C Kling
- The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Antje Blumenthal
- Australian Infectious Diseases Research Centre - The University of Queensland, Brisbane, QLD, Australia.,The University of Queensland Diamantina Institute, Brisbane, QLD, Australia
| | - Chen Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre - The University of Queensland, Brisbane, QLD, Australia
| | - Katharina Ronacher
- Translational Research Institute, Mater Research Institute, The University of Queensland, Brisbane, QLD, Australia.,Australian Infectious Diseases Research Centre - The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
11
|
Sun M, Ma K, Wen J, Wang G, Zhang C, Li Q, Bao X, Wang H. A Review of the Brain-Gut-Microbiome Axis and the Potential Role of Microbiota in Alzheimer's Disease. J Alzheimers Dis 2021; 73:849-865. [PMID: 31884474 DOI: 10.3233/jad-190872] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative process characterized by loss of neurons in the hippocampus and cerebral cortex, leading to progressive cognitive decline. Pathologically, the hallmark of AD is accumulation of "senile" plaques composed of amyloid-β (Aβ) protein surrounding neurons in affected regions. Despite extensive research into AD pathogenesis and therapeutic targets, there remains no breakthroughs in its management. In recent years, there has been a spark of interest in the connection between the brain and gastrointestinal tract, referred to as the brain-gut axis, and its potential implications for both metabolic and neurologic disease. Moreover, the gastrointestinal flora, referred to as the microbiome, appears to exert significant influence over the brain-gut axis. With the need for expanded horizons in understanding and treating AD, many have turned to the brain-gut-microbiome axis for answers. Here we provide a review of the brain-gut-microbiome axis and discuss the evidence supporting alterations of the axis in the pathogenesis of AD. Specifically, we highlight the role for the microbiome in disruption of Aβ metabolism/clearance, increased permeability of the blood-brain barrier and modulation of the neuroinflammatory response, and inhibition of hippocampal neurogenesis. The majority of the above described findings are the result of excellent, albeit basic and pre-clinical studies. Therefore, we conclude with a brief description of documented clinical support for brain-gut-microbiome axis alteration in AD, including potential microbiome-based therapeutics for AD. Collectively, these findings suggest that the brain-gut-microbiome axis may be a "lost link" in understanding and treating AD and call for future work.
Collapse
Affiliation(s)
- Miao Sun
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Kai Ma
- Probiotics Australia, Ormeau, QLD, Australia
| | - Jie Wen
- Beijing Allwegene Health, Beijing, China
| | | | | | - Qi Li
- Beijing Allwegene Health, Beijing, China
| | - Xiaofeng Bao
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China.,Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, Nantong University, Nantong, China
| | - Hui Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
12
|
Gheorghe CE, Ritz NL, Martin JA, Wardill HR, Cryan JF, Clarke G. Investigating causality with fecal microbiota transplantation in rodents: applications, recommendations and pitfalls. Gut Microbes 2021; 13:1941711. [PMID: 34328058 PMCID: PMC8331043 DOI: 10.1080/19490976.2021.1941711] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 02/04/2023] Open
Abstract
In recent years, studies investigating the role of the gut microbiota in health and diseases have increased enormously - making it essential to deepen and question the research methodology employed. Fecal microbiota transplantation (FMT) in rodent studies (either from human or animal donors) allows us to better understand the causal role of the intestinal microbiota across multiple fields. However, this technique lacks standardization and requires careful experimental design in order to obtain optimal results. By comparing several studies in which rodents are the final recipients of FMT, we summarize the common practices employed. In this review, we document the limitations of this method and highlight different parameters to be considered while designing FMT Studies. Standardizing this method is challenging, as it differs according to the research topic, but avoiding common pitfalls is feasible. Several methodological questions remain unanswered to this day and we offer a discussion on issues to be explored in future studies.
Collapse
Affiliation(s)
- Cassandra E. Gheorghe
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Jason A. Martin
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hannah R. Wardill
- Precision Medicine, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
- Adelaide Medical School, the University of Adelaide, Adelaide, Australia
| | - John F. Cryan
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
- APC Microbiome Ireland, University College Cork, Cork, Ireland
- INFANT Research Centre, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Shabat Y, Lichtenstein Y, Ilan Y. Short-Term Cohousing of Sick with Healthy or Treated Mice Alleviates the Inflammatory Response and Liver Damage. Inflammation 2020; 44:518-525. [PMID: 32978699 DOI: 10.1007/s10753-020-01348-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/19/2020] [Accepted: 09/22/2020] [Indexed: 01/08/2023]
Abstract
Cohousing of sick with healthy or treated animals is based on the concept of sharing an intestinal ecosystem and coprophagy, the consumption of feces, which includes sharing of the microbiome and of active drug metabolites secreted in the feces or urine. To develop a model for short-term cohousing, enabling the study of the effect of sharing an ecosystem on inflammatory states. To determine the impact of cohousing of sick and healthy mice on the immune-mediated disorders, mice injected with concanavalin A (ConA) were cohoused with healthy or sick mice or with steroid-treated or untreated mice. To determine the effect of cohousing on acetaminophen (APAP)-induced liver damage, APAP-injected mice were cohoused with N-acetyl-cysteine (NAC)-treated or untreated mice. In the ConA-induced immune-mediated hepatitis model, cohousing of sick with healthy mice was associated with the alleviation of liver damage in sick animals. Similarly, a significant decrease in serum ALT was noted in ConA-injected mice kept in the same cage as ConA-injected mice treated with steroids. A trend for reduction in liver enzymes in APAP-injected mice was observed upon cohousing with NAC-treated animals. Cohousing of sick mice with healthy or treated mice ameliorated the immune-mediated inflammatory state induced by ConA and APAP. These models for liver damage can serve as biological systems for determining the effects of alterations in the ecosystem on the immune system.
Collapse
Affiliation(s)
- Yehudit Shabat
- Gastroenterology and Liver Units, Department of Medicine, Hebrew University-Hadassah Medical Center, Ein-Kerem, POB 1200, IL91120, Jerusalem, Israel
| | - Yoav Lichtenstein
- Gastroenterology and Liver Units, Department of Medicine, Hebrew University-Hadassah Medical Center, Ein-Kerem, POB 1200, IL91120, Jerusalem, Israel
| | - Yaron Ilan
- Gastroenterology and Liver Units, Department of Medicine, Hebrew University-Hadassah Medical Center, Ein-Kerem, POB 1200, IL91120, Jerusalem, Israel.
| |
Collapse
|
14
|
Galan-Ros J, Ramos-Arenas V, Conesa-Zamora P. Predictive values of colon microbiota in the treatment response to colorectal cancer. Pharmacogenomics 2020; 21:1045-1059. [PMID: 32896201 DOI: 10.2217/pgs-2020-0044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The crosstalk between the colon mucosa and the microbiota represents a complex and delicate equilibrium. Gastrointestinal diseases such as inflammatory bowel disease and colorectal cancer (CRC) are associated with a state of altered microbiota composition known as dysbiosis, which seems to play a causative role in some of these illnesses. Recent reports have shown that the colorectal microbiome is responsible for the response and safety to treatments against CRC, especially immunotherapy, hence opening the possibility to use bacteria as a predictive marker and also as a therapeutic agent. The review objective is to summarize updated reports about the the implication of the colorectal microbiome in the development of CRC, in treatment response and its potential as a therapeutic approach.
Collapse
Affiliation(s)
- Jorge Galan-Ros
- Microbiology Department, Santa Lucia University Hospital (HGUSL), Cartagena, 30202, Spain
| | - Verónica Ramos-Arenas
- Clinical Analysis Department, Santa Lucia University Hospital (HGUSL), Cartagena, 30202, Spain
| | - Pablo Conesa-Zamora
- Clinical Analysis Department, Santa Lucia University Hospital (HGUSL), Cartagena, 30202, Spain.,Department of Histology & Pathology, Faculty of Life Sciences, Universidad Católica de Murcia (UCAM), Murcia, 30107, Spain.,Research Group on Molecular Pathology & Pharmacogenetics, Institute for Biomedical Research of Murcia (IMIB), Calle Mezquita sn, Cartagena, 30202, Spain
| |
Collapse
|
15
|
The variable oligomeric state of Amuc_1100 from Akkermansia muciniphila. J Struct Biol 2020; 212:107593. [PMID: 32736072 DOI: 10.1016/j.jsb.2020.107593] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 07/12/2020] [Accepted: 07/23/2020] [Indexed: 12/25/2022]
Abstract
Akkermansia muciniphila is a beneficial microorganism colonized in the human gut that can reverse many intestinal metabolic-related diseases. Amuc_1100 is an outer-membrane protein of A. muciniphila. Oral administration of Amuc_1100 can reduce fat mass development, insulin resistance, and dyslipidemia in mice and activated the toll-like receptor 2 (TLR2) to regulate the immune response of the host, but the molecular mechanism remains unclear. Here we report the crystal structure of the extramembranous domain of Amuc_1100, which consists of a four-stranded antiparallel β-sheet and four α-helices. Two C-terminal helices and the four-stranded antiparallel β-sheet formed two "αββ" motifs and constituted the core domain, which shared a similar fold with type IV pili and type II Secretion system protein. Although the full-length of the extramembranous domain of Amuc_1100 existed as a monomer in solution, they formed trimer in the crystal. Elimination of the N-terminal coiled-coil helix α1 led to dimerization of Amuc_1100 both in solution and in crystal, indicating that the oligomeric state of Amuc_1100 was variable and could be influenced by α1. In addition, we identified that Amuc_1100 could directly bind human TLR2 (hTRL2) in vitro, suggesting that Amuc_1100 may serve as a new ligand for hTLR2. Dimerization of Amuc_1100 improved its hTLR2-binding affinity, suggesting that the α1-truncated Amuc_1100 could be a beneficial candidate for the development of A. muciniphila related drugs.
Collapse
|
16
|
Salek Farrokhi A, Darabi N, Yousefi B, Askandar RH, Shariati M, Eslami M. Is it true that gut microbiota is considered as panacea in cancer therapy? J Cell Physiol 2019; 234:14941-14950. [PMID: 30786013 DOI: 10.1002/jcp.28333] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 01/26/2019] [Accepted: 01/30/2019] [Indexed: 02/06/2023]
Abstract
Recent studies demonstrated that a combination of the gut microbiome has the vital effect on the efficacy of anticancer immune therapies. Regulatory effects of microbiota have been shown in different types of cancer therapies such as chemotherapy and immunotherapy. Immune-checkpoint-blocked therapies are the recent efficient cancer immunotherapy strategies. The target of immune-checkpoint blocking is cytotoxic T lymphocyte protein-4 (CTLA-4) or blockade of programmed death-1 (PD-1) protein and its ligand programmed death ligand 1 (PD-L1) that they have been considered as cancer immunotherapy in recent years. In the latest studies, it have been demonstrated that several gut bacteria such as Akkermansia muciniphila, Bifidobacterium spp., Faecalibacterium spp., and Bacteroides fragilis have the regulatory effects on PD-1, PD-L1, and CTLA-4 blocked anticancer therapy outcome.
Collapse
Affiliation(s)
- Amir Salek Farrokhi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Narges Darabi
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Rafee Habib Askandar
- Nursing Department, Halabja Technical Institute, Sulaimani Polytechnic University, Sulaimani, Iraq
| | - Mansoreh Shariati
- Faculty of Basic Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
17
|
Xiong X, Kuang H, Ansari S, Liu T, Gong J, Wang S, Zhao XY, Ji Y, Li C, Guo L, Zhou L, Chen Z, Leon-Mimila P, Chung MT, Kurabayashi K, Opp J, Campos-Pérez F, Villamil-Ramírez H, Canizales-Quinteros S, Lyons R, Lumeng CN, Zhou B, Qi L, Huertas-Vazquez A, Lusis AJ, Xu XZS, Li S, Yu Y, Li JZ, Lin JD. Landscape of Intercellular Crosstalk in Healthy and NASH Liver Revealed by Single-Cell Secretome Gene Analysis. Mol Cell 2019; 75:644-660.e5. [PMID: 31398325 PMCID: PMC7262680 DOI: 10.1016/j.molcel.2019.07.028] [Citation(s) in RCA: 516] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/27/2022]
Abstract
Cell-cell communication via ligand-receptor signaling is a fundamental feature of complex organs. Despite this, the global landscape of intercellular signaling in mammalian liver has not been elucidated. Here we perform single-cell RNA sequencing on non-parenchymal cells isolated from healthy and NASH mouse livers. Secretome gene analysis revealed a highly connected network of intrahepatic signaling and disruption of vascular signaling in NASH. We uncovered the emergence of NASH-associated macrophages (NAMs), which are marked by high expression of triggering receptors expressed on myeloid cells 2 (Trem2), as a feature of mouse and human NASH that is linked to disease severity and highly responsive to pharmacological and dietary interventions. Finally, hepatic stellate cells (HSCs) serve as a hub of intrahepatic signaling via HSC-derived stellakines and their responsiveness to vasoactive hormones. These results provide unprecedented insights into the landscape of intercellular crosstalk and reprogramming of liver cells in health and disease.
Collapse
Affiliation(s)
- Xuelian Xiong
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Henry Kuang
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Sahar Ansari
- Department of Human Genetics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Tongyu Liu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jianke Gong
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA; International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, and College of Life Science and Technology, and Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuai Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xu-Yun Zhao
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Chuan Li
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Liang Guo
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Linkang Zhou
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Zhimin Chen
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Paola Leon-Mimila
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Meng Ting Chung
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48105, USA
| | - Judy Opp
- University of Michigan DNA Sequencing Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Francisco Campos-Pérez
- Clínica Integral de Cirugía para la Obesidad y Enfermedades Metabólicas, Hospital General Dr. Rubén Lénero, Mexico City, Mexico
| | - Hugo Villamil-Ramírez
- Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Unidad de Genómica de Poblaciones Aplicada a la Salud, Mexico City, Mexico
| | - Samuel Canizales-Quinteros
- Facultad de Química, UNAM/Instituto Nacional de Medicina Genómica (INMEGEN), Unidad de Genómica de Poblaciones Aplicada a la Salud, Mexico City, Mexico
| | - Robert Lyons
- University of Michigan DNA Sequencing Core, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Beiyan Zhou
- Department of Immunology, School of Medicine, University of Connecticut, Farmington, CT 06030, USA
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Adriana Huertas-Vazquez
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, Los Angeles, CA, USA
| | - X Z Shawn Xu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Molecular and Integrative Physiology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Siming Li
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Yonghao Yu
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Z Li
- Department of Human Genetics, University of Michigan Medical Center, Ann Arbor, MI 48109, USA
| | - Jiandie D Lin
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical Center, Ann Arbor, MI 48109, USA.
| |
Collapse
|
18
|
Ji Y, Sun S, Shrestha N, Darragh LB, Shirakawa J, Xing Y, He Y, Carboneau BA, Kim H, An D, Ma M, Oberholzer J, Soleimanpour SA, Gannon M, Liu C, Naji A, Kulkarni RN, Wang Y, Kersten S, Qi L. Toll-like receptors TLR2 and TLR4 block the replication of pancreatic β cells in diet-induced obesity. Nat Immunol 2019; 20:677-686. [PMID: 31110312 PMCID: PMC6531334 DOI: 10.1038/s41590-019-0396-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 04/04/2019] [Indexed: 12/21/2022]
Abstract
Consumption of a high-energy Western diet triggers mild adaptive β cell proliferation to compensate for peripheral insulin resistance; however, the underlying molecular mechanism remains unclear. In the present study we show that the toll-like receptors TLR2 and TLR4 inhibited the diet-induced replication of β cells in mice and humans. The combined, but not the individual, loss of TLR2 and TLR4 increased the replication of β cells, but not that of α cells, leading to enlarged β cell area and hyperinsulinemia in diet-induced obesity. Loss of TLR2 and TLR4 increased the nuclear abundance of the cell cycle regulators cyclin D2 and Cdk4 in a manner dependent on the signaling mediator Erk. These data reveal a regulatory mechanism controlling the proliferation of β cells in diet-induced obesity and suggest that selective targeting of the TLR2/TLR4 pathways may reverse β cell failure in patients with diabetes.
Collapse
Affiliation(s)
- Yewei Ji
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Shengyi Sun
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Center for Molecular Medicine and Genetics, Department of Microbiology, Immunology and Biochemistry, Wayne State University School of Medicine, Detroit, MI, USA
| | - Neha Shrestha
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Laurel B Darragh
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Department of Radiation Oncology, School of Medicine, University of Colorado, Aurora, CO, USA
| | - Jun Shirakawa
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Yuan Xing
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Yi He
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Bethany A Carboneau
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hana Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- XBiotech USA, Inc., Austin, TX, USA
| | - Duo An
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Jose Oberholzer
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Scott A Soleimanpour
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Maureen Gannon
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chengyang Liu
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Naji
- Department of Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Rohit N Kulkarni
- Section of Islet Cell and Regenerative Biology, Joslin Diabetes Center, Department of Medicine, Brigham and Women's Hospital, Harvard Stem Cell Institute, Harvard Medical School, Boston, MA, USA
| | - Yong Wang
- Department of Surgery, University of Virginia, Charlottesville, VA, USA
| | - Sander Kersten
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Nutrition Metabolism and Genomics group, Wageningen University, Wageningen, the Netherlands
| | - Ling Qi
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA.
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
19
|
Friberg IM, Taylor JD, Jackson JA. Diet in the Driving Seat: Natural Diet-Immunity-Microbiome Interactions in Wild Fish. Front Immunol 2019; 10:243. [PMID: 30837993 PMCID: PMC6389695 DOI: 10.3389/fimmu.2019.00243] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
Natural interactions between the diet, microbiome, and immunity are largely unstudied. Here we employ wild three-spined sticklebacks as a model, combining field observations with complementary experimental manipulations of diet designed to mimic seasonal variation in the wild. We clearly demonstrate that season-specific diets are a powerful causal driver of major systemic immunophenotypic variation. This effect occurred largely independently of the bulk composition of the bacterial microbiome (which was also driven by season and diet) and of host condition, demonstrating neither of these, per se, constrain immune allocation in healthy individuals. Nonetheless, through observations in multiple anatomical compartments, differentially exposed to the direct effects of food and immunity, we found evidence of immune-driven control of bacterial community composition in mucus layers. This points to the interactive nature of the host-microbiome relationship, and is the first time, to our knowledge, that this causal chain (diet → immunity → microbiome) has been demonstrated in wild vertebrates. Microbiome effects on immunity were not excluded and, importantly, we identified outgrowth of potentially pathogenic bacteria (especially mycolic-acid producing corynebacteria) as a consequence of the more animal-protein-rich summertime diet. This may provide part of the ultimate explanation (and possibly a proximal cue) for the dramatic immune re-adjustments that we saw in response to diet change.
Collapse
Affiliation(s)
- Ida M Friberg
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Joe D Taylor
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| | - Joseph A Jackson
- School of Environment and Life Sciences, University of Salford, Salford, United Kingdom
| |
Collapse
|
20
|
Jha AR, Davenport ER, Gautam Y, Bhandari D, Tandukar S, Ng KM, Fragiadakis GK, Holmes S, Gautam GP, Leach J, Sherchand JB, Bustamante CD, Sonnenburg JL. Gut microbiome transition across a lifestyle gradient in Himalaya. PLoS Biol 2018; 16:e2005396. [PMID: 30439937 PMCID: PMC6237292 DOI: 10.1371/journal.pbio.2005396] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 10/15/2018] [Indexed: 01/04/2023] Open
Abstract
The composition of the gut microbiome in industrialized populations differs from those living traditional lifestyles. However, it has been difficult to separate the contributions of human genetic and geographic factors from lifestyle. Whether shifts away from the foraging lifestyle that characterize much of humanity's past influence the gut microbiome, and to what degree, remains unclear. Here, we characterize the stool bacterial composition of four Himalayan populations to investigate how the gut community changes in response to shifts in traditional human lifestyles. These groups led seminomadic hunting-gathering lifestyles until transitioning to varying levels of agricultural dependence upon farming. The Tharu began farming 250-300 years ago, the Raute and Raji transitioned 30-40 years ago, and the Chepang retain many aspects of a foraging lifestyle. We assess the contributions of dietary and environmental factors on their gut-associated microbes and find that differences in the lifestyles of Himalayan foragers and farmers are strongly correlated with microbial community variation. Furthermore, the gut microbiomes of all four traditional Himalayan populations are distinct from that of the Americans, indicating that industrialization may further exacerbate differences in the gut community. The Chepang foragers harbor an elevated abundance of taxa associated with foragers around the world. Conversely, the gut microbiomes of the populations that have transitioned to farming are more similar to those of Americans, with agricultural dependence and several associated lifestyle and environmental factors correlating with the extent of microbiome divergence from the foraging population. The gut microbiomes of Raute and Raji reveal an intermediate state between the Chepang and Tharu, indicating that divergence from a stereotypical foraging microbiome can occur within a single generation. Our results also show that environmental factors such as drinking water source and solid cooking fuel are significantly associated with the gut microbiome. Despite the pronounced differences in gut bacterial composition across populations, we found little differences in alpha diversity across lifestyles. These findings in genetically similar populations living in the same geographical region establish the key role of lifestyle in determining human gut microbiome composition and point to the next challenging steps of determining how large-scale gut microbiome reconfiguration impacts human biology.
Collapse
Affiliation(s)
- Aashish R. Jha
- Department of Biomedical Data Science, Stanford University, Stanford, California, United States of America
- Center for Computational, Evolutionary, and Human Genetics, Stanford University, Stanford, California, United States of America
| | - Emily R. Davenport
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Yoshina Gautam
- Department of Biomedical Data Science, Stanford University, Stanford, California, United States of America
| | - Dinesh Bhandari
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
| | - Sarmila Tandukar
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
| | - Katharine M. Ng
- Department of Microbiology and Immunology, Stanford University, Stanford, California, United States of America
| | - Gabriela K. Fragiadakis
- Department of Microbiology and Immunology, Stanford University, Stanford, California, United States of America
| | - Susan Holmes
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | | | - Jeff Leach
- Human Food Project, Terlingua, Texas, United States of America
- Department of Twin Research and Genetic Epidemiology, King’s College London, St. Thomas’ Hospital, London, United Kingdom
| | | | - Carlos D. Bustamante
- Department of Biomedical Data Science, Stanford University, Stanford, California, United States of America
- Center for Computational, Evolutionary, and Human Genetics, Stanford University, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| | - Justin L. Sonnenburg
- Department of Microbiology and Immunology, Stanford University, Stanford, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
- Center for Human Microbiome Studies, Stanford University, Stanford, California, United States of America
| |
Collapse
|
21
|
Mathieu E, Escribano-Vazquez U, Descamps D, Cherbuy C, Langella P, Riffault S, Remot A, Thomas M. Paradigms of Lung Microbiota Functions in Health and Disease, Particularly, in Asthma. Front Physiol 2018; 9:1168. [PMID: 30246806 PMCID: PMC6110890 DOI: 10.3389/fphys.2018.01168] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 08/03/2018] [Indexed: 12/22/2022] Open
Abstract
Improvements in our knowledge of the gut microbiota have broadened our vision of the microbes associated with the intestine. These microbes are essential actors and protectors of digestive and extra-digestive health and, by extension, crucial for human physiology. Similar reconsiderations are currently underway concerning the endogenous microbes of the lungs, with a shift in focus away from their involvement in infections toward a role in physiology. The discovery of the lung microbiota was delayed by the long-held view that the lungs of healthy individuals were sterile and by sampling difficulties. The lung microbiota has a low density, and the maintenance of small numbers of bacteria seems to be a critical determinant of good health. This review aims to highlight how knowledge about the lung microbiota can change our conception of lung physiology and respiratory health. We provide support for this point of view with knowledge acquired about the gut microbiota and intestinal physiology. We describe the main characteristics of the lung microbiota and its functional impact on lung physiology, particularly in healthy individuals, after birth, but also in asthma. We describe some of the physiological features of the respiratory tract potentially favoring the installation of a dysbiotic microbiota. The gut microbiota feeds and matures the intestinal epithelium and is involved in immunity, when the principal role of the lung microbiota seems to be the orientation and balance of aspects of immune and epithelial responsiveness. This implies that the local and remote effects of bacterial communities are likely to be determinant in many respiratory diseases caused by viruses, allergens or genetic deficiency. Finally, we discuss the reciprocal connections between the gut and lungs that render these two compartments inseparable.
Collapse
Affiliation(s)
- Elliot Mathieu
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Unai Escribano-Vazquez
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Delphyne Descamps
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, Jouy-en-Josas, France
| | - Claire Cherbuy
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Sabine Riffault
- Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique, Université Paris-Saclay, Jouy-en-Josas, France
| | - Aude Remot
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Thomas
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| |
Collapse
|
22
|
Lazar V, Ditu LM, Pircalabioru GG, Gheorghe I, Curutiu C, Holban AM, Picu A, Petcu L, Chifiriuc MC. Aspects of Gut Microbiota and Immune System Interactions in Infectious Diseases, Immunopathology, and Cancer. Front Immunol 2018; 9:1830. [PMID: 30158926 PMCID: PMC6104162 DOI: 10.3389/fimmu.2018.01830] [Citation(s) in RCA: 326] [Impact Index Per Article: 46.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 07/24/2018] [Indexed: 12/12/2022] Open
Abstract
The microbiota consists of a dynamic multispecies community of bacteria, fungi, archaea, and protozoans, bringing to the host organism a dowry of cells and genes more numerous than its own. Among the different non-sterile cavities, the human gut harbors the most complex microbiota, with a strong impact on host homeostasis and immunostasis, being thus essential for maintaining the health condition. In this review, we outline the roles of gut microbiota in immunity, starting with the background information supporting the further presentation of the implications of gut microbiota dysbiosis in host susceptibility to infections, hypersensitivity reactions, autoimmunity, chronic inflammation, and cancer. The role of diet and antibiotics in the occurrence of dysbiosis and its pathological consequences, as well as the potential of probiotics to restore eubiosis is also discussed.
Collapse
Affiliation(s)
- Veronica Lazar
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Lia-Mara Ditu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Gratiela Gradisteanu Pircalabioru
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Irina Gheorghe
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Carmen Curutiu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Alina Maria Holban
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| | - Ariana Picu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- National Institute for Diabetes, Nutrition and Metabolic Diseases Prof. Dr. N. Paulescu, Bucharest, Romania
| | - Laura Petcu
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- National Institute for Diabetes, Nutrition and Metabolic Diseases Prof. Dr. N. Paulescu, Bucharest, Romania
| | - Mariana Carmen Chifiriuc
- Department of Microbiology and Immunology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Earth, Environmental and Life Sciences Section, Research Institute of the University of Bucharest, Bucharest, Romania
| |
Collapse
|
23
|
Rea D, Coppola G, Palma G, Barbieri A, Luciano A, Del Prete P, Rossetti S, Berretta M, Facchini G, Perdonà S, Turco MC, Arra C. Microbiota effects on cancer: from risks to therapies. Oncotarget 2018; 9:17915-17927. [PMID: 29707157 PMCID: PMC5915165 DOI: 10.18632/oncotarget.24681] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 02/27/2018] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota, a group of 1014 bacteria, eukaryotes and virus living in gastrointestinal tract, is crucial for many physiological processes in particular plays an important role in inflammatory and immune reactions. Several internal and external factors can influence this population, and shifts in their composition, have been demonstrated to contribute and affect different diseases. During dysbiosis several bacteria related to inflammation, one of the most necessary factors in carcinogenesis; it has been shown that some bacterial strains through deregulation of different signals/pathways may affect tumor development through the production of many factors. Gut microbiota might be considered as a holistic hub point for cancer development: direct and indirect involvements have been studying in several neoplasms such as colon rectal cancer, hepatocellular carcinoma and breast cancer. This review discuss over the evidence of crosstalk between gut microbiota and cancer, its ability to modulate chemotherapy, radiotherapy and immunotherapy, and the possibility that the intestinal microbial is a new target for therapeutic approaches to improve the prognosis and quality of life of cancer patients.
Collapse
Affiliation(s)
- Domenica Rea
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Giovanni Coppola
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Giuseppe Palma
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Antonio Barbieri
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Antonio Luciano
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Paola Del Prete
- Direzione Scientifica, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Sabrina Rossetti
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO- Aviano, National Cancer Institute, Aviano, Italy
| | - Gaetano Facchini
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Maria Caterina Turco
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy
| | - Claudio Arra
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| |
Collapse
|
24
|
Zhang N, Ju Z, Zuo T. Time for food: The impact of diet on gut microbiota and human health. Nutrition 2018; 51-52:80-85. [PMID: 29621737 DOI: 10.1016/j.nut.2017.12.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 01/05/2023]
Abstract
There is growing recognition of the role of diet on modulating the composition and metabolic activity of the human gut microbiota, which in turn influence health. Dietary ingredients and food additives have a substantial impact on the gut microbiota and hence affect human health. Updates on current understanding of the gut microbiota in diseases and metabolic disorders are addressed in this review, providing insights into how this can be transferred from bench to bench side as gut microbes are integrated with food. The potency of microbiota-targeted biomarkers as a state-of-art tool for diagnosis of diseases was also discussed, and it would instruct individuals with healthy dietary consumption. Herein, recent advances in understanding the effect of diet on gut microbiota from an ecological perspective, and how these insights might promote health by guiding development of prebiotic and probiotic strategies and functional foods, were explored.
Collapse
Affiliation(s)
- Na Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Zhongjie Ju
- Yantai Center for Food and Drug Control, Yantai, Shandong, China
| | - Tao Zuo
- State Key Laboratory of Digestive Disease, Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; Institute of Digestive Disease, Prince of Wales Hospital, Shatin, Hong Kong SAR, China.
| |
Collapse
|
25
|
Sica A, Massarotti M. Myeloid suppressor cells in cancer and autoimmunity. J Autoimmun 2017; 85:117-125. [PMID: 28728794 DOI: 10.1016/j.jaut.2017.07.010] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 12/18/2022]
Abstract
A bottleneck for immunotherapy of cancer is the immunosuppressive microenvironment in which the tumor cells proliferate. Cancers harness the immune regulatory mechanism that prevents autoimmunity from evading immunosurveillance and promoting immune destruction. Regulatory T cells, myeloid suppressor cells, inhibitory cytokines and immune checkpoint receptors are the major components of the immune system acting in concert with cancer cells and causing the subversion of anti-tumor immunity. This redundant immunosuppressive network poses an impediment to efficacious immunotherapy by facilitating tumor progression. Tumor-associated myeloid cells comprise heterogeneous populations acting systemically (myeloid-derived suppressor cells/MDSCs) and/or locally in the tumor microenvironment (MDSCs and tumor-associated macrophages/TAMs). Both populations promote cancer cell proliferation and survival, angiogenesis and lymphangiogenesis and elicit immunosuppression through different pathways, including the expression of immunosuppressive cytokines and checkpoint inhibitors. Several evidences have demonstrated that myeloid cells can express different functional programs in response to different microenvironmental signals, a property defined as functional plasticity. The opposed extremes of this functional flexibility are generally represented by the classical macrophage activation, which identifies inflammatory and cytotoxic M1 polarized macrophages, and the alternative state of macrophage activation, which identifies M2 polarized anti-inflammatory and immunosuppressive macrophages. Functional skewing of myeloid cells occurs in vivo under physiological and pathological conditions, including cancer and autoimmunity. Here we discuss how myeloid suppressor cells can on one hand support tumor growth and, on the other, limit autoimmune responses, indicating that their therapeutic reprogramming can generate opportunities in relieving immunosuppression in the tumor microenvironment or reinstating tolerance in autoimmune conditions.
Collapse
Affiliation(s)
- Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy; Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Center for Translational Research on Autoimmune and Allergic Diseases, CAAD, Novara, Italy.
| | - Marco Massarotti
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089 Rozzano, Milan, Italy; Department of Rheumatology, University Hospitals of Morecambe Bay NHS Foundation Trust, Royal Lancaster Infirmary, Ashton Road, LA1 4RP Lancaster, United Kingdom
| |
Collapse
|
26
|
Zhang N, Hou E, Song J, Li J, Tang Q, Mao X. Neoagarotetraose-modulated gut microbiota and alleviated gut inflammation in antibiotic treatment mice. FOOD AGR IMMUNOL 2017. [DOI: 10.1080/09540105.2017.1346063] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Affiliation(s)
- Na Zhang
- College of Food Science and Engineering, Ocean University of China, Qingdao, People’s Republic of China
| | - Enling Hou
- College of Food Science and Engineering, Ocean University of China, Qingdao, People’s Republic of China
| | - Jia Song
- College of Food Science and Engineering, Ocean University of China, Qingdao, People’s Republic of China
| | - Jing Li
- College of Food Science and Engineering, Ocean University of China, Qingdao, People’s Republic of China
| | - Qingjuan Tang
- College of Food Science and Engineering, Ocean University of China, Qingdao, People’s Republic of China
| | - Xiangzhao Mao
- College of Food Science and Engineering, Ocean University of China, Qingdao, People’s Republic of China
| |
Collapse
|
27
|
Age-Associated Microbial Dysbiosis Promotes Intestinal Permeability, Systemic Inflammation, and Macrophage Dysfunction. Cell Host Microbe 2017; 21:455-466.e4. [PMID: 28407483 PMCID: PMC5392495 DOI: 10.1016/j.chom.2017.03.002] [Citation(s) in RCA: 773] [Impact Index Per Article: 96.6] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 02/01/2017] [Accepted: 03/02/2017] [Indexed: 12/20/2022]
Abstract
Levels of inflammatory mediators in circulation are known to increase with age, but the underlying cause of this age-associated inflammation is debated. We find that, when maintained under germ-free conditions, mice do not display an age-related increase in circulating pro-inflammatory cytokine levels. A higher proportion of germ-free mice live to 600 days than their conventional counterparts, and macrophages derived from aged germ-free mice maintain anti-microbial activity. Co-housing germ-free mice with old, but not young, conventionally raised mice increases pro-inflammatory cytokines in the blood. In tumor necrosis factor (TNF)-deficient mice, which are protected from age-associated inflammation, age-related microbiota changes are not observed. Furthermore, age-associated microbiota changes can be reversed by reducing TNF using anti-TNF therapy. These data suggest that aging-associated microbiota promote inflammation and that reversing these age-related microbiota changes represents a potential strategy for reducing age-associated inflammation and the accompanying morbidity. Age-associated inflammation drives macrophage dysfunction and tissue damage Mice under germ-free conditions are protected from age-associated inflammation Co-housing germ-free mice with old, but not young, mice increases age-related inflammation Age-related microbiota changes can be reversed by reducing TNF levels
Collapse
|
28
|
Ji Y, Kim H, Yang L, Sha H, Roman CA, Long Q, Qi L. The Sel1L-Hrd1 Endoplasmic Reticulum-Associated Degradation Complex Manages a Key Checkpoint in B Cell Development. Cell Rep 2016; 16:2630-2640. [PMID: 27568564 DOI: 10.1016/j.celrep.2016.08.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 07/06/2016] [Accepted: 07/31/2016] [Indexed: 01/09/2023] Open
Abstract
Endoplasmic reticulum (ER)-associated degradation (ERAD) is a principal mechanism that targets ER-associated proteins for cytosolic proteasomal degradation. Here, our data demonstrate a critical role for the Sel1L-Hrd1 complex, the most conserved branch of ERAD, in early B cell development. Loss of Sel1L-Hrd1 ERAD in B cell precursors leads to a severe developmental block at the transition from large to small pre-B cells. Mechanistically, we show that Sel1L-Hrd1 ERAD selectively recognizes and targets the pre-B cell receptor (pre-BCR) for proteasomal degradation in a BiP-dependent manner. The pre-BCR complex accumulates both intracellularly and at the cell surface in Sel1L-deficient pre-B cells, leading to persistent pre-BCR signaling and pre-B cell proliferation. This study thus implicates ERAD mediated by Sel1L-Hrd1 as a key regulator of B cell development and reveals the molecular mechanism underpinning the transient nature of pre-BCR signaling.
Collapse
Affiliation(s)
- Yewei Ji
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Hana Kim
- Graduate Field of Immunology and Infectious Disease, Cornell University, Ithaca, NY 14853, USA
| | - Liu Yang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Haibo Sha
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA
| | - Christopher A Roman
- Department of Cell Biology, College of Medicine and Program in Molecular and Cellular Biology, The School of Graduate Studies, State University of New York, Downstate Medical Center at Brooklyn, New York, NY 11203, USA
| | - Qiaoming Long
- Laboratory Animal Research Center, Medical College of Soochow University, Suzhou 215006, Jiangsu, China
| | - Ling Qi
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853, USA; Graduate Field of Immunology and Infectious Disease, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
29
|
Abstract
The intestinal microbiome is a signalling hub that integrates environmental inputs, such as diet, with genetic and immune signals to affect the host's metabolism, immunity and response to infection. The haematopoietic and non-haematopoietic cells of the innate immune system are located strategically at the host-microbiome interface. These cells have the ability to sense microorganisms or their metabolic products and to translate the signals into host physiological responses and the regulation of microbial ecology. Aberrations in the communication between the innate immune system and the gut microbiota might contribute to complex diseases.
Collapse
|
30
|
Polycarpou A, Holland MJ, Karageorgiou I, Eddaoudi A, Walker SL, Willcocks S, Lockwood DNJ. Mycobacterium leprae Activates Toll-Like Receptor-4 Signaling and Expression on Macrophages Depending on Previous Bacillus Calmette-Guerin Vaccination. Front Cell Infect Microbiol 2016; 6:72. [PMID: 27458573 PMCID: PMC4937034 DOI: 10.3389/fcimb.2016.00072] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/24/2016] [Indexed: 01/09/2023] Open
Abstract
Toll-like receptor (TLR)-1 and TLR2 have been shown to be receptors for Mycobacterium leprae (M. leprae), yet it is unclear whether M. leprae can signal through alternative TLRs. Other mycobacterial species possess ligands for TLR4 and genetic association studies in human populations suggest that people with TLR4 polymorphisms may be protected against leprosy. Using human embryonic kidney (HEK)-293 cells co-transfected with TLR4, we demonstrate that M. leprae activates TLR4. We used human macrophages to show that M. leprae stimulation of cytokine production is diminished if pre-treated with TLR4 neutralizing antibody. TLR4 protein expression was up-regulated on macrophages derived from non-bacillus Calmette-Guerin (BCG) vaccinated healthy volunteers after incubation with M. leprae, whereas it was down-regulated in macrophages derived from BCG-vaccinated donors. Finally, pre-treatment of macrophages derived from BCG-naive donors with BCG reversed the effect of M. leprae on TLR4 expression. This may be a newly described phenomenon by which BCG vaccination stimulates “non-specific” protection to the human immune system.
Collapse
Affiliation(s)
- Anastasia Polycarpou
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Martin J Holland
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Ioannis Karageorgiou
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Ayad Eddaoudi
- Molecular and Cellular Immunology Unit, Institute of Child Health, University College London London, UK
| | - Stephen L Walker
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Sam Willcocks
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| | - Diana N J Lockwood
- Department of Clinical Research, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine London, UK
| |
Collapse
|
31
|
TLR2/4 deficiency prevents oxygen-induced vascular degeneration and promotes revascularization by downregulating IL-17 in the retina. Sci Rep 2016; 6:27739. [PMID: 27297042 PMCID: PMC4906284 DOI: 10.1038/srep27739] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 05/23/2016] [Indexed: 12/25/2022] Open
Abstract
Vascular degeneration is a critical pathological process in many human degenerative diseases, which need efficient ways to revascularization. However, little is known about cellular and molecular mechanisms that are used during vascular degeneration and revascularization. Here, we show that Toll-like receptor 2 and 4 (TLR2/4) double deficiency suppressed hyperoxia induced retinal vessel regression in an oxygen-induced retinopathy (OIR) model. Notably, the TLR2/4−/− mice experienced more revascularization after reduced vessel regression compared with wild-type mice, accompanied with less activation of glial cells. Mechanistically, TLR2/4 activation can tip the balance between Th17 cells and regulatory T cells towards Th17 cells, a critical source of the IL-17A. Less migration and infiltration of IL-17A-expressing proinflammatory cells but elevated regulatory T cells were observed in OIR-retinae from TLR2/4−/− mice. Coincidentally, TLR2/4 deficiency suppressed IL-17A production and increased expressions of anti-inflammatory genes. Furthermore, IL-17A promoted activation of glial cells. IL-17A blockade using a neutralizing antibody alleviated retinal cell apoptosis and glial activation in C57/B6-OIR mice, demonstrating the important role of IL-17A pathway in glial function during revascularization. Thus TLR2/4-mediated IL-17A inflammatory signaling is involved in vessel degeneration and revascularization, indicating that modulation of the TLR2/4-IL-17A pathway may be a novel therapeutic strategy for degenerative diseases.
Collapse
|
32
|
Abrupt suspension of probiotics administration may increase host pathogen susceptibility by inducing gut dysbiosis. Sci Rep 2016; 6:23214. [PMID: 26983596 PMCID: PMC4794715 DOI: 10.1038/srep23214] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 02/29/2016] [Indexed: 01/14/2023] Open
Abstract
In this study, we investigated the risk associated with suspension of probiotics administration in tilapia, an animal model that may mimic immune-compromised conditions in humans. Tilapias were fed for 14 days using a probiotics-supplemented diet, followed by a three-day suspension of probiotics treatment and a subsequent challenge by Aeromonas hydrophila. Unexpectedly, the suspension of a probiotic strain Lactobacillus plantarum JCM1149 significantly triggered susceptibility of the host to A. hydrophila. We further observed that suspension of JCM1149 resulted in host gut microbiota dysbiosis and the subsequent disorder in the intestinal metabolites (bile acids, amino acids, and glucose) and damage in the intestinal epithelium, giving rise to a condition similar to antibiotics-induced gut dysbiosis, which collectively impaired tilapia’s gut health and resistance to pathogenic challenges. Additionally, we determined that JCM1149 adhered relatively poorly to tilapia intestinal mucosa and was rapidly released from the gastrointestinal tract (GIT) after suspension, with the rapid loss of probiotic strain probably being the direct cause of gut dysbiosis. Finally, three other probiotic Lactobacillus strains with low intestinal mucosa binding activity showed similar rapid loss phenotype following administration suspension, and induced higher host susceptibility to infection, indicating that the risk is a generic phenomenon in Lactobacillus.
Collapse
|
33
|
Sun S, Lourie R, Cohen SB, Ji Y, Goodrich JK, Poole AC, Ley RE, Denkers EY, McGuckin MA, Long Q, Duhamel GE, Simpson KW, Qi L. Epithelial Sel1L is required for the maintenance of intestinal homeostasis. Mol Biol Cell 2015; 27:483-90. [PMID: 26631554 PMCID: PMC4751599 DOI: 10.1091/mbc.e15-10-0724] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/23/2015] [Indexed: 01/05/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an incurable chronic idiopathic disease that drastically decreases quality of life. Endoplasmic reticulum (ER)-associated degradation (ERAD) is responsible for the clearance of misfolded proteins; however, its role in disease pathogenesis remains largely unexplored. Here we show that the expression of SEL1L and HRD1, the most conserved branch of mammalian ERAD, is significantly reduced in ileal Crohn's disease (CD). Consistent with this observation, laboratory mice with enterocyte-specific Sel1L deficiency (Sel1L(ΔIEC)) develop spontaneous enteritis and have increased susceptibility to Toxoplasma gondii-induced ileitis. This is associated with profound defects in Paneth cells and a disproportionate increase of Ruminococcus gnavus, a mucolytic bacterium with known association with CD. Surprisingly, whereas both ER stress sensor IRE1α and effector CHOP are activated in the small intestine of Sel1L(ΔIEC) mice, they are not solely responsible for ERAD deficiency-associated lesions seen in the small intestine. Thus our study points to a constitutive role of Sel1L-Hrd1 ERAD in epithelial cell biology and the pathogenesis of intestinal inflammation in CD.
Collapse
Affiliation(s)
- Shengyi Sun
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853 Graduate Program in Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853
| | - Rohan Lourie
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Sara B Cohen
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Yewei Ji
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853
| | - Julia K Goodrich
- Graduate Program in Genetics, Genomics and Development, Cornell University, Ithaca, NY 14853
| | - Angela C Poole
- Department of Microbiology, Cornell University, Ithaca, NY 14853 Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Ruth E Ley
- Graduate Program in Genetics, Genomics and Development, Cornell University, Ithaca, NY 14853 Department of Microbiology, Cornell University, Ithaca, NY 14853 Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853
| | - Eric Y Denkers
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853
| | - Michael A McGuckin
- Immunity, Infection and Inflammation Program, Mater Medical Research Institute, Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Qiaoming Long
- Laboratory Animal Research Center, Medical College of Soochow University, Suzhou 215006, Jiangsu, China
| | - Gerald E Duhamel
- Department of Biomedical Sciences, Cornell University, Ithaca, NY 14853
| | - Kenneth W Simpson
- Department of Clinical Science, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Ling Qi
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14853 Graduate Program in Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, NY 14853
| |
Collapse
|
34
|
Abstract
The innate immune system includes several classes of pattern recognition receptors (PRRs), including membrane-bound Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). These receptors detect pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) in the extracellular and intracellular space. Intracellular NLRs constitute inflammasomes, which activate and release caspase-1, IL-1β, and IL-18 thereby initiating an inflammatory response. Systemic and local low-grade inflammation and release of proinflammatory cytokines are implicated in the development and progression of diabetes mellitus and diabetic nephropathy. TLR2, TLR4, and the NLRP3 inflammasome can induce the production of various proinflammatory cytokines and are critically involved in inflammatory responses in pancreatic islets, and in adipose, liver and kidney tissues. This Review describes how innate immune system-driven inflammatory processes can lead to apoptosis, tissue fibrosis, and organ dysfunction resulting in insulin resistance, impaired insulin secretion, and renal failure. We propose that careful targeting of TLR2, TLR4, and NLRP3 signalling pathways could be beneficial for the treatment of diabetes mellitus and diabetic nephropathy.
Collapse
|
35
|
Sun S, Shi G, Sha H, Ji Y, Han X, Shu X, Ma H, Inoue T, Gao B, Kim H, Bu P, Guber RD, Shen X, Lee AH, Iwawaki T, Paton AW, Paton JC, Fang D, Tsai B, Yates JR, Wu H, Kersten S, Long Q, Duhamel GE, Simpson KW, Qi L. IRE1α is an endogenous substrate of endoplasmic-reticulum-associated degradation. Nat Cell Biol 2015; 17:1546-55. [PMID: 26551274 PMCID: PMC4670240 DOI: 10.1038/ncb3266] [Citation(s) in RCA: 180] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 10/05/2015] [Indexed: 12/13/2022]
Abstract
Endoplasmic reticulum (ER)-associated degradation (ERAD) represents a principle quality control mechanism to clear misfolded proteins in the ER; however its physiological significance and the nature of endogenous ERAD substrates remain largely unexplored. Here we discover that IRE1α, the sensor of unfolded protein response (UPR), is a bona fide substrate of the Sel1L-Hrd1 ERAD complex. ERAD-mediated IRE1α degradation occurs under basal conditions in a BiP-dependent manner, requires both intramembrane hydrophilic residues of IRE1α and lectin protein OS9, and is attenuated by ER stress. ERAD deficiency causes IRE1α protein stabilization, accumulation and mild activation both in vitro and in vivo. Although enterocyte-specific Sel1L-knockout mice (Sel1LΔIEC) are viable and appear normal, they are highly susceptible to experimental colitis and inflammation-associated dysbiosis, in an IRE1α-dependent but CHOP-independent manner. Hence, Sel1L-Hrd1 ERAD serves a distinct, essential function in restraint of IRE1α signaling in vivo by managing its protein turnover.
Collapse
Affiliation(s)
- Shengyi Sun
- Graduate Program in Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, USA
| | - Guojun Shi
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Haibo Sha
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Yewei Ji
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Xuemei Han
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Xin Shu
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Hongming Ma
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905, USA
| | - Takamasa Inoue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - Beixue Gao
- Department of Pathology, Northwestern University, Chicago, Illinois 60611, USA
| | - Hana Kim
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Pengcheng Bu
- Department of Electrical and Computer Engineering, Cornell University, Ithaca, New York 14853, USA.,Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Robert D Guber
- Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Xiling Shen
- Department of Electrical and Computer Engineering, Cornell University, Ithaca, New York 14853, USA.,Department of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA.,Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Ann-Hwee Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York 10021, USA
| | - Takao Iwawaki
- Education and Research Support Center, Gunma University Graduate School of Medicine, 3-39-22 Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - James C Paton
- Research Centre for Infectious Diseases, School of Molecular and Biomedical Science, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Deyu Fang
- Department of Pathology, Northwestern University, Chicago, Illinois 60611, USA
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | - John R Yates
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Haoquan Wu
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, Texas 79905, USA
| | - Sander Kersten
- Nutrition Metabolism and Genomics group, Wageningen University, Bomenweg 2, 6703HD Wageningen, The Netherlands
| | - Qiaoming Long
- Laboratory Animal Research Center, Medical College of Soochow University, Suzhou 215006, Jiangsu, China
| | - Gerald E Duhamel
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
| | - Kenneth W Simpson
- Department of Clinical Science, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
| | - Ling Qi
- Graduate Program in Biochemistry, Molecular and Cell Biology, Cornell University, Ithaca, New York 14853, USA.,Division of Nutritional Sciences, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
36
|
Stearic acid induces proinflammatory cytokine production partly through activation of lactate-HIF1α pathway in chondrocytes. Sci Rep 2015; 5:13092. [PMID: 26271607 PMCID: PMC4536527 DOI: 10.1038/srep13092] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/17/2015] [Indexed: 12/22/2022] Open
Abstract
The biomechanics stress and chronic inflammation in obesity are causally linked to osteoarthritis. However, the metabolic factors mediating obesity-related osteoarthritis are still obscure. Here we scanned and identified at least two elevated metabolites (stearic acid and lactate) from the plasma of diet-induced obese mice. We found that stearic acid potentiated LDH-a-dependent production of lactate, which further stabilized HIF1α protein and increased VEGF and proinflammatory cytokine expression in primary mouse chondrocytes. Treatment with LDH-a and HIF1α inhibitors notably attenuated stearic acid-or high fat diet-stimulated proinflammatory cytokine production in vitro and in vivo. Furthermore, positive correlation of plasma lactate, cartilage HIF1α and cytokine levels with the body mass index was observed in subjects with osteoarthritis. In conclusion, saturated free fatty acid induced proinflammatory cytokine production partly through activation of a novel lactate-HIF1α pathway in chondrocytes. Our findings hold promise of developing novel clinical strategies for the management of obesity-related diseases such as osteoarthritis.
Collapse
|
37
|
Tilg H, Moschen AR. Food, immunity, and the microbiome. Gastroenterology 2015; 148:1107-19. [PMID: 25575570 DOI: 10.1053/j.gastro.2014.12.036] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/22/2014] [Accepted: 12/23/2014] [Indexed: 12/12/2022]
Abstract
There is increasing evidence that ingested diet-borne components are involved in the pathogenesis of disorders such as inflammatory bowel diseases, atherosclerosis, and type 2 diabetes. Nutrients can have short- and long-term effects in shaping the composition of the microbiota. Western diets (enriched in fat, phosphatidylcholine, and L-carnitine) promote inflammation and atherosclerosis through specific fatty acids and degradation products such as trimethylamine N-oxide. Other dietary factors such as carbazoles or tryptophan-enriched proteins have anti-inflammatory properties-partly via activation of aryl hydrocarbon receptors. The microbiota and its metabolic machinery produce a myriad of metabolites that serve as important messengers between the diet, microbiota, and host. Short-chain fatty acids affect immune responses and epithelial integrity via G-protein-coupled receptors and epigenetic mechanisms. By increasing our understanding of interactions between diet, immunity, and the microbiota, we might develop food-based approaches to prevent or treat many diseases. There now is scientific evidence to support the adage "we are what we eat," and this process begins in early life.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Endocrinology, Gastroenterology and Metabolism, Medical University Innsbruck, Innsbruck, Austria.
| | - Alexander R Moschen
- Department of Internal Medicine I, Endocrinology, Gastroenterology and Metabolism, Medical University Innsbruck, Innsbruck, Austria
| |
Collapse
|
38
|
Hörmann N, Brandão I, Jäckel S, Ens N, Lillich M, Walter U, Reinhardt C. Gut microbial colonization orchestrates TLR2 expression, signaling and epithelial proliferation in the small intestinal mucosa. PLoS One 2014; 9:e113080. [PMID: 25396415 PMCID: PMC4232598 DOI: 10.1371/journal.pone.0113080] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 10/19/2014] [Indexed: 01/19/2023] Open
Abstract
The gut microbiota is an environmental factor that determines renewal of the intestinal epithelium and remodeling of the intestinal mucosa. At present, it is not resolved if components of the gut microbiota can augment innate immune sensing in the intestinal epithelium via the up-regulation of Toll-like receptors (TLRs). Here, we report that colonization of germ-free (GF) Swiss Webster mice with a complex gut microbiota augments expression of TLR2. The microbiota-dependent up-regulation of components of the TLR2 signaling complex could be reversed by a 7 day broad-spectrum antibiotic treatment. TLR2 downstream signaling via the mitogen-activated protein kinase (ERK1/2) and protein-kinase B (AKT) induced by bacterial TLR2 agonists resulted in increased proliferation of the small intestinal epithelial cell line MODE-K. Mice that were colonized from birth with a normal gut microbiota (conventionally-raised; CONV-R) showed signs of increased small intestinal renewal and apoptosis compared with GF controls as indicated by elevated mRNA levels of the proliferation markers Ki67 and Cyclin D1, elevated transcripts of the apoptosis marker Caspase-3 and increased numbers of TUNEL-positive cells per intestinal villus structure. In accordance, TLR2-deficient mice showed reduced proliferation and reduced apoptosis. Our findings suggest that a tuned proliferation response of epithelial cells following microbial colonization could aid to protect the host from its microbial colonizers and increase intestinal surface area.
Collapse
Affiliation(s)
- Nives Hörmann
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
| | - Inês Brandão
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
| | - Sven Jäckel
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
| | - Nelli Ens
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
| | - Maren Lillich
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
| | - Ulrich Walter
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis (CTH), University Medical Center Mainz, Junior Group Translational Research in Thrombosis and Hemostasis, Mainz, Germany
- * E-mail:
| |
Collapse
|