scholarly journals Gut Microbiota Dysbiosis Aggravates Mycoplasma gallisepticum Colonization in the Chicken Lung

2021 ◽  
Vol 8 ◽  
Author(s):  
Jian Wang ◽  
Xueping Chen ◽  
Jichang Li ◽  
Muhammad Ishfaq

Mycoplasma gallisepticum (MG) is the pathogen that causes chronic respiratory diseases in chickens. Gut microbiota plays an important role in maintaining body health and resisting respiratory infection, but the correlation between gut microbiota and MG infection is poorly defined. Therefore, in this study, the correlation between gut microbiota and MG infection was explored by disturbing gut microbiota in chickens with antibiotic cocktail. The results showed that the gut microbiota dysbiosis impairs pulmonary immune response against MG infection. It has been noted that MG colonization in the lung was significantly increased following gut microbiota dysbiosis, and this could be reversed by intranasally administrated toll-like receptor 2 (TLR2) ligand, recombinant chicken IL-17 protein or recombinant chicken granulocyte-macrophage colony-stimulating factor (GM-CSF) protein. In addition, the levels of short-chain fatty acids (SCFAs) and vitamin A were significantly reduced in gut microbiota dysbiosis group, however, butyric acid or vitamin A as feed additives promoted MG clearance in the lung of gut microbiota dysbiosis group via increasing TLR2/IL17/GM-CSF and host defense peptides genes expression. The present study revealed an important role of gut microbiota in the defense against MG colonization in the lung of chicken.

2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Xiuli Lin ◽  
Yingying Liu ◽  
Lili Ma ◽  
Xiaomeng Ma ◽  
Liping Shen ◽  
...  

Abstract Background Constipation is a common gastrointestinal dysfunction which has a potential impact on people's immune state and their quality of life. Here we investigated the effects of constipation on experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Methods Constipation was induced by loperamide in female C57BL/6 mice. The alternations of gut microbiota, permeability of intestinal barrier and blood–brain barrier, and histopathology of colon were assessed after constipation induction. EAE was induced in the constipation mice. Fecal microbiota transplantation (FMT) was performed from constipation mice into microbiota-depleted mice. Clinical scores, histopathology of inflammation and demyelination, Treg/Th17 and Treg17/Teff17 imbalance both in the peripheral lymphatic organs and central nervous system, cytokines include TGF-β, GM-CSF, IL-10, IL-17A, IL-17F, IL-21, IL-22, and IL-23 in serum were assessed in different groups. Results Compared with the vehicle group, the constipation mice showed gut microbiota dysbiosis, colon inflammation and injury, and increased permeability of intestinal barrier and blood–brain barrier. We found that the clinical and pathological scores of the constipation EAE mice were severer than that of the EAE mice. Compared with the EAE mice, the constipation EAE mice showed reduced percentage of Treg and Treg17 cells, increased percentage of Th17 and Teff17 cells, and decreased ratio of Treg/Th17 and Treg17/Teff17 in the spleen, inguinal lymph nodes, brain, and spinal cord. Moreover, the serum levels of TGF-β, IL-10, and IL-21 were decreased while the GM-CSF, IL-17A, IL-17F, IL-22, and IL-23 were increased in the constipation EAE mice. In addition, these pathological processes could be transferred via their gut microbiota. Conclusions Our results verified that constipation induced gut microbiota dysbiosis exacerbated EAE via aggravating Treg/Th17 and Treg17/Teff17 imbalance and cytokines disturbance in C57BL/6 mice.


Author(s):  
Katarina Bojović ◽  
Ður -d ica Ignjatović ◽  
Svetlana Soković Bajić ◽  
Danijela Vojnović Milutinović ◽  
Mirko Tomić ◽  
...  

2021 ◽  
Author(s):  
Leonardo Lupori ◽  
Sara Cornuti ◽  
Raffaele M Mazziotti ◽  
Elisa Borghi ◽  
Emerenziana Ottaviano ◽  
...  

Exposing animals to an enriched environment (EE) has dramatic effects on brain structure, function and plasticity. The poorly known "EE derived signals" mediating the EE effects are thought to be generated within the central nervous system. Here, we shift the focus to the body periphery, revealing that gut microbiota signals are crucial for EE-driven plasticity. Developmental analysis of intestinal bacteria composition in EE mice revealed striking differences from standard condition (ST) animals and enhanced levels of short-chain fatty acids (SCFA). Depleting the EE mice gut microbiota with an antibiotic cocktail decreased SCFA and prevented EE induction of adult ocular dominance (OD) plasticity, spine dynamics and microglia rearrangement. SCFA treatment in ST mice mimicked the EE induction of adult OD plasticity and morphological microglial rearrangement. Remarkably, transferring the microbiota of EE mice to ST recipients activated adult OD plasticity. Thus, taken together our data suggest that experience-dependent changes in gut microbiota regulate brain plasticity.


2021 ◽  
Vol 8 ◽  
Author(s):  
Yang Guo ◽  
Xiaosu Li ◽  
Zhijian Wang ◽  
Bo Yu

Introduction: Hypertension is one of the major risk factors to human health and human studies on association between gut microbiota and hypertension or blood pressure have received increased attention. In the present study, we aim to evaluate gut microbiota dysbiosis in human hypertension using a method of systematic review.Methods: PubMed, EMBASE, and Web of Science databases were searched until March 2021 to identify eligible articles. Additional articles were also identified by searching specific authors in this field. Inclusion criteria were observational studies based on stool samples with hypertension group and control group. Newcastle-Ottawa quality assessment scale (NOS) was used to assess the quality of the included studies. PROSPERO registration number: CRD42020212219.Results: A total of 17 studies enrolling 9,085 participants were included. Fifteen of the enrolled studies showed good quality and two studies showed fair quality based on NOS. We found alpha diversity in hypertension decreased significantly and microbial structure can be separated compared with control groups. Gut microbiota of hypertension showed depletion of short chain fatty acids (SCFAs) producers and over-growth of some Proteobacteria and Bacteroidetes members. Up-regulation of lipopolysaccharide biosynthesis, phosphotransferase system, ABC transporters, etc. and down-regulation of some amino acid metabolism, etc. in hypertension were reported. Fecal SCFAs levels increased and plasma SCFAs levels decreased in hypertension. Stronger microbial interactions in hypertension were seen.Conclusion: In conclusion, gut microbiota dysbiosis was observed in hypertension, including decreased diversity, altered microbial structure, compositional change of taxa, alterations of microbial function, nutritional and immunological factors, and microbial interactions. Poor absorption and high excretion of SCFAs may play an important role in the pathogenesis of hypertension. These findings may provide insights into etiology study and new microbial-based therapies of hypertension.Systematic Review Registration: PROSPERO database, identifier CRD42020212219.


2021 ◽  
Vol 15 (2) ◽  
pp. 137-148
Author(s):  
Alina M Rüb ◽  
Anastasia Tsakmaklis ◽  
Stefanie K Gräfe ◽  
Marie-Christine Simon ◽  
Maria JGT Vehreschild ◽  
...  

The association of gut microbiota dysbiosis with various human diseases is being substantiated with increasing evidence. Metabolites derived from both, microbiota and the human host play a central role in disease susceptibility and disease progression by extensively modulating host physiology and metabolism. Several of these metabolites have the potential to serve as diagnostic biomarkers for monitoring disease states in conjunction with intestinal microbiota dysbiosis. In this narrative review we evaluate the potential of trimethylamine-N-oxide, short-chain fatty acids, 3-indoxyl sulfate, p-cresyl sulfate, secondary bile acids, hippurate, human β-defensin-2, chromogranin A, secreted immunoglobulins and zonulin to serve as biomarkers for metabolite profiling and diagnostic suitability for dysbiosis and disease.


2021 ◽  
Author(s):  
Pratikshya Ray ◽  
Subhayan Chakraborty ◽  
Arindam Ghosh ◽  
Palok Aich

AbstractHigher organisms, especially mammals, harbor diverse microbiota in the gut that plays a major role in maintaining health and physiological homeostasis. Perturbation of gut flora helps identifying their roles. Antibiotics are potent perturbing agents of microbiome. Select antibiotics like vancomycin, neomycin, and AVNM (an antibiotic cocktail containing ampicillin, vancomycin, neomycin, and metronidazole) were used to perturb the gut microbiota of C57BL/6 male mice to understand their roles in host immunity and metabolism. The current study revealed that the resulting gut microbial composition was different, and diversity (at the phylum and genus level) was reduced differentially following each antibiotic treatment. Vancomycin treatment caused a significant increase in Verrucomicrobia and Proteobacteria phyla. The treatment with neomycin yielded an increase in the Bacteroidetes phylum, while the treatment with AVNM led to an increase in Proteobacteria phylum with lowest diversity of microbiome in the gut. The current results also revealed that the different antibiotics treatment caused variation in the cecal index, expression of immune genes (TNF-α, IL-10, IFN-γ) in the colon, and short-chain fatty acids (SCFA) level in the blood of mice. A strong correlation was observed for antibiotic-induced differential dysbiosis patterns of gut microbiota and the altered immune and SCFA profile of the host. The outcome of the present study could be clinically important.


Author(s):  
An Zhou ◽  
Yuanyuan Lei ◽  
Li Tang ◽  
Shiping Hu ◽  
Min Yang ◽  
...  

The gut microbiota plays a crucial role in the development of the immune system and confers benefits or disease susceptibility to the host. Emerging studies have indicated that the gut microbiota could affect pulmonary health and disease through cross-talk between the gut microbiota and the lungs. Gut microbiota dysbiosis could lead to acute or chronic lung disease, such as asthma, tuberculosis and lung cancer. In addition, the composition of the gut microbiota may be associated with different lung diseases, the prevalence of which also vary by age. Modulation of the gut microbiota through short-chain fatty acids, probiotics and micronutrients may present potential therapeutic strategies to protect against lung diseases. In this review, we will provide an overview of the cross-talk between the gut microbiota and the lungs as well as elucidate the underlying pathogenesis or potential therapeutic strategies of some lung diseases from the point of view of the gut microbiota.


2021 ◽  
Vol 12 ◽  
Author(s):  
Xiefang Yuan ◽  
Hongmei Tang ◽  
Renlan Wu ◽  
Xingjie Li ◽  
Hongyu Jiang ◽  
...  

Gut-microbiota dysbiosis links to allergic diseases. The mechanism of the exacerbation of food allergy caused by gut-microbiota dysbiosis remains unknown. Regulation of retinoic acid receptor alpha (RARα) signaling is critical for gut immune homeostasis. Here we clarified that RARα in dendritic cells (DCs) promotes Th2 cell differentiation. Antibiotics treatment stimulates retinoic acid signaling in mucosal DCs. We found microbiota metabolites short-chain fatty acids (SCFAs) maintain IGF-1 levels in serum and mesenteric lymph nodes. The IGF-1/Akt pathway is essential for regulating the transcription of genes targeted by RARα. And RARα in DCs affects type I interferon (IFN-I) responses through regulating transcription of IFN-α. Our study identifies SCFAs crosstalk with RARα in dendritic cells as a critical modulator that plays a core role in promoting Th2 cells differentiation at a state of modified/disturbed microbiome.


2021 ◽  
Vol 13 ◽  
Author(s):  
Ting Shen ◽  
Yumei Yue ◽  
Tingting He ◽  
Cong Huang ◽  
Boyi Qu ◽  
...  

Patients with Parkinson's disease (PD) were often observed with gastrointestinal symptoms, which preceded the onset of motor symptoms. Neuropathology of PD has also been found in the enteric nervous system (ENS). Many studies have reported significant PD-related alterations of gut microbiota. This meta-analysis was performed to evaluate the differences of gut microbiota between patients with PD and healthy controls (HCs) across different geographical regions. We conducted a systematic online search for case-control studies detecting gut microbiota in patients with PD and HCs. Mean difference (MD) and 95% confidence interval (CI) were calculated to access alterations in the abundance of certain microbiota families in PD. Fifteen case-control studies were included in this meta-analysis study. Our results showed significant lower abundance levels of Prevotellaceae (MD = −0.37, 95% CI = −0.62 to −0.11), Faecalibacterium (MD = −0.41, 95% CI: −0.57 to −0.24), and Lachnospiraceae (MD = −0.34, 95% CI = −0.59 to −0.09) in patients with PD compared to HCs. Significant higher abundance level of Bifidobacteriaceae (MD = 0.38, 95%; CI = 0.12 to 0.63), Ruminococcaceae (MD = 0.58, 95% CI = 0.07 to 1.10), Verrucomicrobiaceae (MD = 0.45, 95% CI = 0.21 to 0.69), and Christensenellaceae (MD = 0.20, 95% CI = 0.07 to 0.34) was also found in patients with PD. Thus, shared alterations of certain gut microbiota were detected in patients with PD across different geographical regions. These PD-related gut microbiota dysbiosis might lead to the impairment of short-chain fatty acids (SCFAs) producing process, lipid metabolism, immunoregulatory function, and intestinal permeability, which contribute to the pathogenesis of PD.


Proceedings ◽  
2020 ◽  
Vol 61 (1) ◽  
pp. 28
Author(s):  
Omorogieva Ojo ◽  
Qianqian Feng ◽  
Osarhumwese Osaretin Ojo ◽  
Xiaohua Wang

Background: Diabetes prevalence is on the increase globally and its impact on those with the condition in terms of acute and chronic complications can be profound. People with type 2 diabetes constitute the majority of those with the condition and the risk factors include obesity, lifestyle and gut microbiota dysbiosis. Poor dietary intake has been reported to influence the community of the gut microbiome. Therefore, a higher intake of dietary fibre may alter the environment in the gut and promote microbial growth and proliferation. Aim: This is a systematic review and meta-analysis which examined the effect of dietary fibre on gut microbiota in patients with type 2 diabetes. Method: This review was conducted in line with the PRISMA framework. Databases were searched for relevant articles which were screened based on inclusion and exclusion criteria. Results: Nine articles which met the inclusion criteria were selected for the systematic review and meta-analysis. High dietary fibre intake significantly improved (p < 0.05) the abundance of Bifidobacterium, total short-chain fatty acids (SCFAs) and HbA1c. Discussion: The promotion of SCFA producers in terms of greater diversity and abundance by dietary fibre may have resulted in improvement in glycated haemoglobin, partly due to increased GLP–1 production. Conclusion: High consumption of dietary fibre has a significant (p < 0.05) effect on Bifidobacterium, total SCFAs and HbA1c, but not (p > 0.05) on propionic, butyric and acetic acid, fasting blood glucose and the homeostatic model assessment of insulin resistance HOMAR–IR.


Sign in / Sign up

Export Citation Format

Share Document