intestinal dysbacteriosis
Recently Published Documents


TOTAL DOCUMENTS

33
(FIVE YEARS 22)

H-INDEX

5
(FIVE YEARS 4)

2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Yuwei Liu ◽  
Wanbin He ◽  
Jie Yang ◽  
Yuhua He ◽  
Ziqiang Wang ◽  
...  

Abstract Background Accumulating evidence suggests a critical role of intestinal dysbacteriosis in surgical site infections and anastomotic leakage after abdominal surgery. However, a direct correlation between pre-existing dysbacteriosis and postoperative infectious complications has not yet been established clinically. Methods A total of 353 consecutive patients who underwent colorectal cancer (CRC) surgery were enrolled. Gram-stained faecal smears were tested at admission and the first defecation after surgery. Intestinal dysbacteriosis was graded into three groups: normal or slightly decreased intestinal microflora (grade 1), moderate dysbacteriosis (grade 2), and severe dysbacteriosis (grade 3). Clinical outcomes were postoperative infections and anastomotic leakage within 30 days after surgery. Results At the preoperative assessment, 268 (75.9%) patients had normal or slightly decreased intestinal microflora, 58 (16.4%) patients had moderate dysbacteriosis, and 27 (7.6%) patients had severe dysbacteriosis. The patients with preoperative dysbacteriosis had a higher rate of early postoperative diarrhoea (grade 2: OR = 4.53, 95% CI 2.28–9.00, grade 3: OR = 4.52, 95% CI 1.81–11.31), total complications (grade 3 40.7% vs. grade 2 25.9% vs. grade 1 12.7%, P < 0.001), and anastomotic leakage (grade 3 11.1% vs. grade 2 5.2% vs. grade 1 1.5%, P = 0.002). An interaction effect among preoperative dysbacteriosis and early postoperative diarrhoea on total complications was observed in rectal cancer patients (P for interaction = 0.007). Conclusions An imbalance of the intestinal microbiome exists in a considerable proportion of CRC patients before surgery. Preoperative dysbacteriosis is associated with higher rates of early postoperative diarrhoea, which further correlates with infectious complications and anastomotic leakage. However, the contribution of preoperative dysbacteriosis to the occurrence of anastomotic leakage needs to be clarified in further studies. Trial registration ChiCTR, ChiCTR1800018755. Registered 8 October 2018—Retrospectively registered, http://www.chictr.org.cn/ChiCTR1800018755.


2021 ◽  
Author(s):  
Jie Sun ◽  
Y.Z. Wen ◽  
S.W. Feng ◽  
H.Y. Dai ◽  
M. Mao ◽  
...  

Abstract Background: Perioperative neurocognitive disorders (PND) occur frequently in elderly patients after surgery, but the mechanism of PND is not very clear at present. It is reported that anesthesia/surgery could cause intestinal flora imbalance and induce neurocognitive impairment. However, the effect of intestinal flora on PND is poorly understood. We previously found that peripheral interleukin-17A (IL17A) destroyed the blood-brain barrier (BBB), leading to central inflammation and neurocognitive impairment. The small intestine is the main place where Th17 cells are produced. Therefore, we hypothesized that Th17 cells and IL-17 may be an important bridge for intestinal microbes to cause neuroinflammation. Methods: Exploratory laparotomy was performed to establish PND model under sevoflurane anesthesia. 16S rRNA high-throughput sequencing was used to detect the changes of intestinal flora. To explore the relationship between intestinal flora and PND, compound antibiotics were used to eliminate intestinal flora before anesthesia/surgery, and behavior tests, such as open field, Y maze, and fear conditioning tests were applied to detect the changes of memory ability and which was compared with the rats that did not receive compound antibiotics. The number of Th17 cells and Foxp3 cells was detected by flow cytometry in the Peyer's patches (PP), mesenteric lymph nodes (MLN), blood and brain. Hippocampus IL17, IL17RA, IL6 and IL10 were detected by Western blot. Hippocampus IL17, IL17R and IBA1 (ionized calcium binding adaptor molecule1) were detected by immunofluorescence. Results: Anesthesia/surgery caused intestinal flora imbalance and induced neurocognitive impairment, increased the number of Th17 cells in the PP, MLN, blood and brain, up-regulated the lever of IL17, IL17R and inflammatory factor production in the hippocampus. The administration of compound antibiotics before anesthesia/surgery evidently inhibited this effect, including decreased the number of Th17 cells, down-regulated the lever of IL17, IL17R and inflammatory factor production, and improved the memory function. In addition, we found that IL17R was co-labeled with IBA1 in a large amount in the hippocampus through immunofluorescence double-staining. Conclusion: Our study suggested that intestinal dysbacteriosis-propelled T helper 17 cells activation might play an important role in the pathogenesis of PND.


2021 ◽  
Vol 80 (Suppl 1) ◽  
pp. 646.2-646
Author(s):  
Y. Bao ◽  
J. Ji ◽  
Z. Xue ◽  
Z. Gu

Background:Intestinal flora disorder and immune abnormalities have been reported in systemic lupus erythematosus (SLE) patients1,2. Berberine (BBR) showed significant effects in regulating the intestinal flora, repairing gut barriers and regulating immune cells3,4. While few reports mentioned the abnormal gut microbiota and metabolites in Chinese SLE patients.Objectives:Our investigation tried to illustrate the relationship between gut microbiota, intestinal metabolites and disease activity in Chinese SLE patients. And the effect of BBR to intestinal dysbacteriosis, multiple organ damages and over-activated immune system in MRL/Lpr mice.Methods:16S high-throughput (16S rRNA) sequence, qRT-PCR and gas chromatography technology were used to determine the gut microbiota and metabolites in 104 SLE patients from Affiliated Hospital of Nantong University, China. BBR was orally treated to the MRL/Lpr mice in low, medium and high doses. After 6 weeks treatment, mice were sacrificed. Serum, faeces and organs were collected for further studies.Results:Chinese SLE patients showed higher abundance of Bacteroidetes and lower abundance of Firmcutes. The results of qRT-PCR showed high Firmcutes/Bacteroidetes (F/B) ratio of SLE patients. The F/B ratio was negative correlated with SLE disease activity index (SLEDA) score. Almost all the tested short-chain fatty acids (SCFAs) found statistically significant results in SLE and LN (lupus nephritis) patients, especially the propanoic acid and butyric. BBR altered the relative abundance of Bacteroides and Verrucomicrobia and the butyric acid content in colon of MRL/Lpr mice. The increase of tight junction protein also indicated the gut barrier was repaired by BBR. Treg and Tfr cells in spleen and mesenteric lymph node (MLN) were increased. These results revealed a positive therapeutic effect of berberine on SLE from gut microbiota to immune status.Conclusion:Our study highlights current status of intestinal dysbacteriosis in Chinese patients with SLE and differences in intestinal metabolites among patients with different disease states. The regulation of intestinal flora and the repairment of gut barrier by intestinal metabolites in BBR treated mice seemed to be the factor that directed the immune responses and disease outcomes. The ultimate goal of our study was to determine the beneficial effects of regulating the gut microbiota on the treatment of SLE. The application of berberine is a relatively safe and convenient way. In the coming investigations, we plan to focus on the study of berberine and its metabolites on intestinal function and systemic immunity.References:[1]Guo, M. et al. Alteration in gut microbiota is associated with dysregulation of cytokines and glucocorticoid therapy in systemic lupus erythematosus. Gut microbes11, 1758-1773, doi:10.1080/19490976.2020.1768644 (2020).[2]Mu, Q. et al. Control of lupus nephritis by changes of gut microbiota. Microbiome5, 73, doi:10.1186/s40168-017-0300-8 (2017).[3]Habtemariam, S. Berberine pharmacology and the gut microbiota: A hidden therapeutic link. Pharmacological research155, 104722, doi:10.1016/j.phrs.2020.104722 (2020).[4]Cui, H. et al. Berberine Regulates Treg/Th17 Balance to Treat Ulcerative Colitis Through Modulating the Gut Microbiota in the Colon. Frontiers in pharmacology9, 571, doi:10.3389/fphar.2018.00571 (2018).Figure 1.Disclosure of Interests:None declared


2021 ◽  
Vol 19 (1) ◽  
Author(s):  
Xiao Ran ◽  
Yu He ◽  
Qing Ai ◽  
Yuan Shi

Abstract Background Modification of the gut microbiota by antibiotics may influence the disease susceptibility and immunological responses. Infants in the neonatal intensive care unit (NICU) subjected to frequent antibiotics and oxygen therapies, which may give rise to local and systemic inflammatory reactions and progression of bronchopulmonary dysplasia (BPD). This study aimed to investigate the role of intestinal dysbacteriosis by antibiotic therapy before hyperoxia exposure in the progression of BPD. Methods Mice had been exposed to hyperoxia (85% O2) since postnatal day 3 until day 16 for the BPD model establishment, treated with antibiotics from postnatal day 2 until day 8. Treated mice and appropriate controls were harvested on postnatal day 2 or 10 for 16S rRNA gene sequencing, or postnatal day 17 for assessment of alveolar morphometry and macrophages differentiation. Results Antibiotic-induced intestinal dysbacteriosis before hyperoxia exposure gave rise to deterioration of BPD evidenced by reduced survival rates and alveolarization. Moreover, antibiotic-induced intestinal dysbacteriosis resulted in increased M1 macrophage maker (iNOS) and decreased M2 macrophage maker (Arg-1) levels in lung homogenates. Conclusion Broad-spectrum antibiotic-induced intestinal dysbacteriosis may participate in BPD pathogenesis via alteration of the macrophage polarization status. Manipulating the gut microbiota may potentially intervene the therapy of BPD.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Pan Wang ◽  
Ke Tu ◽  
Peng Cao ◽  
Yuefan Yang ◽  
Hao Zhang ◽  
...  

An amendment to this paper has been published and can be accessed via the original article.


2021 ◽  
Vol 14 (1) ◽  
Author(s):  
Pan Wang ◽  
Ke Tu ◽  
Peng Cao ◽  
Yuefan Yang ◽  
Hao Zhang ◽  
...  

AbstractAntibiotics affect gut microbial composition, leading to Gut–Brain-Axis imbalance and neurobehavioral changes. However, the intestinal dysbacteriosis associated behavior changes are not consistently reported. It is not clear whether these changes are transient or permanent. The neuroprotective effect of probiotics against intestinal dysbacteriosis induced alternations needs to be determined either. In the present study, oral antibiotic mixture including Ampicillin, Streptomycin, and Clindamycin was utilized to induce intestinal dysbacteriosis in mice. Antibiotics application triggered mechanical allodynia in von frey test and spontaneous pain in open field test. It also resulted in increased anxiety and depressive-like behaviors and damaged spatial memory performance. After application of probiotics, the mechanical allodynia and spontaneous pain were alleviated significantly. The anxiety behaviors, depressive-like behaviors and recognitive performance were ameliorative as well. By using Fos protein as a marker, it is found that the sensory, emotion and memory related brain regions were activated in mice with intestinal dysbacteriosis. Our study is not only helpful for enriching our basic knowledge for understanding the changed pain responses and related brain disorders in antibiotics-induced dysbacteriosis mice, but also beneficial for providing a more comprehensive mechanistic explanation for the regulation of antibiotics and probiotics on gut microbiota and relevant alternations in animal neurological behaviors.


2021 ◽  
Vol 11 ◽  
Author(s):  
Fang Yang ◽  
Yi Yang ◽  
Yiwei Chen ◽  
Guobao Li ◽  
Guoliang Zhang ◽  
...  

Tuberculosis (TB), which is a frequent and important infectious disease caused by Mycobacterium tuberculosis, has resulted in an extremely high burden of morbidity and mortality. The importance of intestinal dysbacteriosis in regulating host immunity has been implicated in TB, and accumulating evidence suggests that microRNAs (miRNAs) might act as a key mediator in maintaining intestinal homeostasis through signaling networks. However, the involvement of miRNA in gut microbiota, TB and the host immune system remains unknown. Here we showed that intestinal dysbacteriosis increases the susceptibility to TB and remotely increased the expression of miR-21 in lung. Systemic antagonism of miR-21 enhanced IFN-γ production and further conferred immune protection against TB. Molecular experiments further indicated that miR-21a-3p could specifically target IFN-γ mRNA. These findings revealed regulatory pathways implicating intestinal dysbacteriosis induced-susceptibility to TB: intestinal dysbiosis→lung miRNA→targeting IFN-γ→impaired anti-TB immunity. This study also suggested that deregulated miRNAs by commensal bacteria could become promising targets as TB therapeutics.


2021 ◽  
Author(s):  
Pan Wang ◽  
Ke Tu ◽  
Peng Cao ◽  
Yue-Fan Yang ◽  
Hao Zhang ◽  
...  

Abstract Antibiotics affect gut microbial composition, leading to Gut-Brain-Axis imbalance and neurobehavioral changes. However, the intestinal dysbacteriosis associated behavior changes are not consistently reported. It is not clear whether these changes are transient or permanent. The neuroprotective effect of probiotics against intestinal dysbacteriosis induced alternations needs to be determined either. In the present study, oral antibiotic mixture including Ampicillin, Streptomycin, and Clindamycin was utilized to induce intestinal dysbacteriosis in mice. Antibiotics application triggered mechanical allodynia in von frey test and spontaneous pain in open field test. It also resulted in increased anxiety and depressive-like behaviors and damaged spatial memory performance. After application of probiotics, the mechanical allodynia and spontaneous pain were alleviated significantly. The anxiety behaviors, depressive-like behaviors and recognitive performance were ameliorative as well. By using Fos protein as a marker, it is found that the sensory, emotion and memory related brain regions were activated in mice with intestinal dysbacteriosis. Our study is not only helpful for enriching our basic knowledge for understanding the changed pain responses and related brain disorders in antibiotics-induced dysbacteriosis mice, but also beneficial for providing a more comprehensive mechanistic explanation for the regulation of antibiotics and probiotics on gut microbiota and relevant alternations in animal neurological behaviors.


2021 ◽  
Author(s):  
Xiao Ran ◽  
Yu He ◽  
Qing Ai ◽  
Yuan Shi

Abstract BackgroundThe modification of the gut microbiota by antibiotics may influence the disease susceptibility and immunological responses. Infants in the neonatal intensive care unit (NICU) subjected to frequent antibiotics and oxygen therapies, which may give rise to the local and systemic inflammatory reactions and progression of bronchopulmonary dysplasia (BPD). This study aimed to investigate the role of intestinal dysbacteriosis by antibiotic therapy before hyperoxia exposure in the progression of BPD.MethodsMice had been exposed to hyperoxia (85% O2) since postnatal day 3 until day 16 for the BPD model establishment, treated with antibiotics from postnatal day 2 until day 8. Treated mice and appropriate controls were harvested on postnatal day 10 for 16S rRNA gene sequencing, or postnatal day 17 for assessment of alveolar morphometry and macrophages differentiation.ResultsAntibiotic-induced intestinal dysbacteriosis before hyperoxia exposure gave rise to deterioration of BPD evidenced by reduced survival rates and alveolarization, moreover, antibiotic-induced intestinal dysbacteriosis resulted in increased iNOS and decreased Arg-1 levels in lung homogenates.ConclusionBroad-spectrum antibiotic-induced intestinal dysbacteriosis may participate in BPD pathogenesis via alteration of the macrophage polarization status. Manipulating the gut microbiota may potentially intervene the therapy of BPD.


2021 ◽  
Author(s):  
Yafeng Zhou ◽  
Shanshan Tian ◽  
Li Qian ◽  
Su Jiang ◽  
Yunping Tang ◽  
...  

Docosahexaenoic acid-enriched phosphatidylserine (DHA-PS) has attracted increasing attention because of its unique health benefits. In this study, DHA-PS was biosynthesized from DHA-enriched phosphatidylcholine (DHA-PC), which was extracted from the herring...


Sign in / Sign up

Export Citation Format

Share Document