Contribution of preoperative gut microbiota in postoperative neurocognitive dysfunction in elderly patients undergoing orthopedic surgery

Jiangjiang Bi, Yifan Xu, Shiyong Li, Gaofeng Zhan, Dongyu Hua, Juan Tan, Xiaohui Chi, Hongbing Xiang, Fengjing Guo, Ailin Luo, Jiangjiang Bi, Yifan Xu, Shiyong Li, Gaofeng Zhan, Dongyu Hua, Juan Tan, Xiaohui Chi, Hongbing Xiang, Fengjing Guo, Ailin Luo

Abstract

Objective: To investigate the role of gut microbiota and metabolites in POCD in elderly orthopedic patients, and screen the preoperative diagnostic indicators of gut microbiota in elderly POCD.

Method: 40 elderly patients undergoing orthopedic surgery were enrolled and divided into Control group and POCD group following neuropsychological assessments. Gut microbiota was determined by 16S rRNA MiSeq sequencing, and metabolomics of GC-MS and LC-MS was used to screen the differential metabolites. We then analyzed the pathways enriched by metabolites.

Result: There was no difference in alpha or beta diversity between Control group and POCD group. There were significant differences in 39 ASV and 20 genera bacterium in the relative abundance. Significant diagnostic efficiency analyzed by the ROC curves were found in 6 genera bacterium. Differential metabolites in the two groups including acetic acid, arachidic acid, pyrophosphate etc. were screened out and enriched to certain metabolic pathways which impacted the cognition function profoundly.

Conclusion: Gut microbiota disorders exist preoperatively in the elderly POCD patients, by which there could be a chance to predict the susceptible population.

Clinical trial registration: [http://www.chictr.org.cn/edit.aspx?pid=133843&htm=4], identifier [ChiCTR2100051162].

Keywords: elderly patients; gut microbiota; metabolites; orthopedic surgery; postoperative neurocognitive dysfunction.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2023 Bi, Xu, Li, Zhan, Hua, Tan, Chi, Xiang, Guo and Luo.

Figures

Figure1
Figure1
Comparisons of relative abundance of gut microbiota. (A) Histogram results for the top 20 in order of relative abundance at phylum level, which shows Bacteroidota, Firmicutes, Proteobacteria etc. contribute the main components of gut microbiota, (B) Comparisons between the two groups using Wilcoxon rank sum test, significant differences in relative abundance were observed in 20 genus (*p < 0.05), and (CL) Relative abundance of differential genera bacterium, analyzed by two-tailed Wilcoxon rank-sum test, data represent the means ± SEM, n = 16 in each group, *p < 0.05 compared with Control group.
Figure 2
Figure 2
Alpha diversity and beta diversity in gut microbiota between the two groups. (A–C) There was no significant difference of alpha diversity as measured by the Chao1 diversity index (p = 0.098). The Shannon index (p = 0.065) and ACE index (p = 0.110) indicated species diversity and evenness, did not show significant differences. (D,E) There was no difference between the two groups measured by PCA and PCoA.
Figure 3
Figure 3
ROC curves of the gut genus bacterium relative abundance for the diagnosis of POCD. Vertical coordinate indicated the sensitivity of diagnosis, horizontal coordinate indicated the 1-specificity of diagnosis, AUC > 0.5 indicated a diagnosis efficiency of the gut bacterium.
Figure 4
Figure 4
(A,B) It showed significant differences between two groups at the metabolic profiling level both in PCA and OPLS-DA (R2 = 0.961, Q2 = −0.268), (C,D) Differential metabolites between the two groups performed by T-test, and visualization by their value of ps and Fold change values using volcano plots. The red dots represent metabolites that were significantly upregulated in the POCD group, the blue dots represent metabolites that were significantly downregulated and the grey dots represent metabolites that were not significant.
Figure 5
Figure 5
Heat map of differential metabolites. We performed hierarchical clustering of all significantly differential metabolites expressions, including 27 metabolites in GC–MS (A), and 125 metabolites in LC–MS (B). The horizontal coordinates indicate sample names and the vertical coordinates indicate differential metabolites. The colour ranges from blue to red, a redder colour indicates a higher abundance of expression of the differential metabolite.
Figure 6
Figure 6
Metabolic pathway enrichment bubble chart. The value of p of the metabolic pathway is the significance of the enrichment of the metabolic pathway, and the significant enrichment pathway was selected for bubble plotting. The vertical coordinate is the name of the metabolic pathway; the horizontal coordinate is the enrichment factor (Rich factor = number of significantly differential metabolites/total number of metabolites in the pathway), the larger the Rich factor, the greater the enrichment; the colour from green to red indicates that the value of p decreases; the larger the bubble, the greater the number of metabolites enriched to that pathway. (A) Metabolic pathway enrichment of GC-MS platform. (B) Metabolic pathway enrichment of LC-MS platform.
Figure 7
Figure 7
Heat map of correlation between microbiome and metabolites. Each column is for a different species and each row corresponds to a metabolite. The graph shows positive correlations in orange and negative correlations in blue, with darker colours representing greater correlations and colours closer to white representing correlations closer to zero. A *** in the graph represents a correlation p value less than 0.001, a ** in the graph represents a correlation p value less than 0.01 and a * in the graph represents a correlation p value less than 0.05.

References

    1. Berrett A. N., Gale S. D., Erickson L. D., Brown B. L., Hedges D. W. (2018). Helicobacter pylori moderates the association between 5-MTHF concentration and cognitive function in older adults. PLoS One 13:e0190475. doi: 10.1371/journal.pone.0190475, PMID:
    1. Bolyen E., Rideout J. R., Dillon M. R., Bokulich N. A., Abnet C. C., al-Ghalith G. A., et al. . (2019). Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 37, 852–857. doi: 10.1038/s41587-019-0209-9, PMID:
    1. Callahan B. J., McMurdie P. J., Rosen M. J., Han A. W., Amy Jo A., Johnson S. P., et al. . (2016). DADA2: high-resolution sample inference from Illumina amplicon data. Nat. Methods 13, 581–583. doi: 10.1038/nmeth.3869, PMID:
    1. Cárdenas V. M., Boller F., Román G. C. (2019). Helicobacter pylori, vascular risk factors and cognition in U.S. older adults. Brain Sci. 9:370. doi: 10.3390/brainsci9120370, PMID:
    1. Chen C., Liao J., Xia Y., Liu X., Jones R., Haran J., et al. . (2022). Gut microbiota regulate Alzheimer's disease pathologies and cognitive disorders via PUFA-associated neuroinflammation. Gut 71, 2233–2252. doi: 10.1136/gutjnl-2021-326269, PMID:
    1. Evered L., Silbert B., Scott D. A., Ames D., Maruff P., Blennow K. (2016). Cerebrospinal fluid biomarker for Alzheimer disease predicts postoperative cognitive dysfunction. Anesthesiology 124, 353–361. doi: 10.1097/ALN.0000000000000953, PMID:
    1. Fang E. F., Hou Y., Palikaras K., Adriaanse B. A., Kerr J. S., Yang B., et al. . (2019). Mitophagy inhibits amyloid-β and tau pathology and reverses cognitive deficits in models of Alzheimer's disease. Nat. Neurosci. 22, 401–412. doi: 10.1038/s41593-018-0332-9, PMID:
    1. Han M.-L., Chen J.-H., Tsai M.-K., Liou J.-M., Chiou J.-M., Chiu M.-J., et al. . (2018). Association between helicobacter pylori infection and cognitive impairment in the elderly. J. Formos. Med. Assoc. 117, 994–1002. doi: 10.1016/j.jfma.2017.11.005, PMID:
    1. Le Y., Liu S., Peng M., Tan C., Liao Q., Duan K., et al. . (2014). Aging differentially affects the loss of neuronal dendritic spine, neuroinflammation and memory impairment at rats after surgery. PLoS One 9:e106837. doi: 10.1371/journal.pone.0106837, PMID:
    1. Liu P., Wu L., Peng G., Han Y., Tang R., Ge J., et al. . (2019). Altered microbiomes distinguish Alzheimer's disease from amnestic mild cognitive impairment and health in a Chinese cohort. Brain Behav. Immun. 80, 633–643. doi: 10.1016/j.bbi.2019.05.008, PMID:
    1. Martin S. A., Brash A. R., Murphy R. C. (2016). The discovery and early structural studies of arachidonic acid. J. Lipid Res. 57, 1126–1132. doi: 10.1194/jlr.R068072, PMID:
    1. Moller J. T., Cluitmans P., Rasmussen L. S., Houx P., Rasmussen H., Canet J., et al. . (1998). Long-term postoperative cognitive dysfunction in the elderly ISPOCD1 study. ISPOCD investigators. International study of post-operative cognitive dysfunction. Lancet 351, 857–861. doi: 10.1016/S0140-6736(97)07382-0, PMID:
    1. Mulak A., Bonaz B. (2015). Brain-gut-microbiota axis in Parkinson's disease. World J. Gastroenterol. 21, 10609–10620. doi: 10.3748/wjg.v21.i37.10609, PMID:
    1. Naseer M. I., Bibi F., Alqahtani M. H., Chaudhary A., Azhar E., Kamal M., et al. . (2014). Role of gut microbiota in obesity, type 2 diabetes and Alzheimer's disease. CNS Neurol. Disord. Drug Targets 13, 305–311. doi: 10.2174/18715273113126660147, PMID:
    1. Ren T., Gao Y., Qiu Y., Jiang S., Zhang Q., Zhang J., et al. . (2020). Gut microbiota altered in mild cognitive impairment compared with Normal cognition in sporadic Parkinson's disease. Front. Neurol. 11:2020. doi: 10.3389/fneur.2020.00137.eCollection
    1. Skvarc D. R., Berk M., Byrne L. K., Dean O. M., Dodd S., Lewis M., et al. . (2018). Post-operative cognitive dysfunction: an exploration of the inflammatory hypothesis and novel therapies. Neurosci. Biobehav. Rev. 84, 116–133. doi: 10.1016/j.neubiorev.2017.11.011
    1. Tian Y., Guo S., Zhang Y., Ying X., Zhao P., Zhao X. (2017). Effects of hydrogen-rich saline on hepatectomy-induced postoperative cognitive dysfunction in old mice. Mol. Neurobiol. 54, 2579–2584. doi: 10.1007/s12035-016-9825-2, PMID:
    1. Tran T. T. T., Corsini S., Kellingray L., Hegarty C., Le Gall G., Narbad A., et al. . (2019). APOE genotype influences the gut microbiome structure and function in humans and mice: relevance for Alzheimer's disease pathophysiology. FASEB J. 33, 8221–8231. doi: 10.1096/fj.201900071R, PMID:
    1. Zameer S., Najmi A. K., Vohora D., Akhtar M. (2018). Bisphosphonates: future perspective for neurological disorders. Bisphosphonates: future perspective for neurological disorders. Pharmacol. Rep. 70, 900–907. doi: 10.1016/j.pharep.2018.03.011, PMID:
    1. Zhan G., Yang N., Li S., Huang N., Fang X., Zhang J., et al. . (2018). Abnormal gut microbiota composition contributes to cognitive dysfunction in SAMP8 mice. Aging (Albany NY) 10, 1257–1267. doi: 10.18632/aging.101464, PMID:
    1. Zheng H., Pengtao X., Jiang Q., Qingqing X., Zheng Y., Yan J., et al. . (2021). Depletion of acetate-producing bacteria from the gut microbiota facilitates cognitive impairment through the gut-brain neural mechanism in diabetic mice. Microbiome 9:145. doi: 10.1186/s40168-021-01088-9, PMID:

Source: PubMed

3
구독하다