Next Article in Journal
Knowledge, Attitudes, and Practices of Antibiotic Use among Small-, Medium-, and Large-Scale Fish Farmers of the Stratum II of the Volta Lake of Ghana
Previous Article in Journal
Effect of Antimicrobial Peptide BiF2_5K7K on Contaminated Bacteria Isolated from Boar Semen and Semen Qualities during Preservation and Subsequent Fertility Test on Pig Farm
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Impact of Surgical Bowel Preparation on the Microbiome in Colon and Rectal Surgery

by
Lauren Weaver
1,†,
Alexander Troester
1,† and
Cyrus Jahansouz
2,*
1
Department of Surgery, University of Minnesota, Minneapolis, MN 55455, USA
2
Division of Colon & Rectal Surgery, Department of Surgery, University of Minnesota, 420 Delaware St. SE, MMC 450, Minneapolis, MN 55455, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Antibiotics 2024, 13(7), 580; https://doi.org/10.3390/antibiotics13070580
Submission received: 15 May 2024 / Revised: 13 June 2024 / Accepted: 21 June 2024 / Published: 23 June 2024

Abstract

:
Preoperative bowel preparation, through iterations over time, has evolved with the goal of optimizing surgical outcomes after colon and rectal surgery. Although bowel preparation is commonplace in current practice, its precise mechanism of action, particularly its effect on the human gut microbiome, has yet to be fully elucidated. Absent intervention, the gut microbiota is largely stable, yet reacts to dietary influences, tissue injury, and microbiota-specific byproducts of metabolism. The routine use of oral antibiotics and mechanical bowel preparation prior to intestinal surgical procedures may have detrimental effects previously thought to be negligible. Recent evidence highlights the sensitivity of gut microbiota to antibiotics, bowel preparation, and surgery; however, there is a lack of knowledge regarding specific causal pathways that could lead to therapeutic interventions. As our understanding of the complex interactions between the human host and gut microbiota grows, we can explore the role of bowel preparation in specific microbiome alterations to refine perioperative care and improve outcomes. In this review, we outline the current fund of information regarding the impact of surgical bowel preparation and its components on the adult gut microbiome. We also emphasize key questions pertinent to future microbiome research and their implications for patients undergoing colorectal surgery.

1. Introduction

Colorectal surgery is associated with the highest rates of surgical site infection (SSI) among elective surgeries [1]. These infectious complications impose a significant financial burden on the healthcare system and have a profound impact on patient morbidity and mortality [1]. Despite rigorous asepsis surgical measures, SSI rates in elective colorectal surgery range from 5.4% to 23.2%, with a weighted mean of 11.4% [1,2]. Since 2016, the World Health Organization has endorsed the use of preoperative bowel preparation with both oral antibiotics (OAs) and mechanical bowel preparation (MBP) to reduce SSIs in elective colorectal surgery [1,3]. Similarly, the American College of Surgeons and the American Society of Colon and Rectal Surgeons (ASCRS) recommend OAs and MBP to lower SSIs, anastomotic leaks, Clostridium difficile infections, and other surgical complications [4,5]. Although infectious complications are currently at a historic low, it is still unclear why certain patients develop SSIs and suffer anastomotic leaks despite uniform empiric antimicrobial treatment for patients [6].
Some answers to these questions may lie within the colon itself. Recent recognition of the crucial role the gut microbiome plays in maintaining human health and survival has created a new research frontier aimed at further understanding and preventing surgical complications [7]. Although often used interchangeably with microbiota, the term “microbiome” refers to the collection of genomes from all gut microorganisms, including microbial structural elements, metabolites, and the overall environmental condition. The microbiome encompasses the gut microbiota, the assemblage of microbes present in a defined environment [8,9]. Perturbations in the gut microbiota, catalyzed by various factors, including dietary changes, parenteral or enteral antibiotic use, and high-volume lavage from mechanical bowel preparation, result in a phenomenon known as dysbiosis [10]. Although bowel preparation has been around for over one hundred years, its precise mechanism of action and its effects on the human gut microbiome remain elusive [6].
Over the past decade, emerging data have shed light on the impact of bowel preparation on the colonic microbiome. Several studies have demonstrated significant differences in stool and mucosal microbial profiles following MBP, including a decrease in Lactobacillaceae, a microbial family typically associated with positive gut health [11,12,13]. Surgical bowel preparation (SBP), consisting of OAs and MBP, has been shown to alter microbiome composition more than MBP alone in colonoscopy patients, although the implications of this are unknown [14]. There has been growing interest and an increase in research published on the effects of SBP on the microbiome, yet a comprehensive understanding of its mechanism of action is lacking. Currently, there is no expert consensus on the microbiota that should be preserved or neutralized to prevent infectious complications [6]. These studies are often limited by small sample sizes, and their conflicting conclusions have left researchers and clinicians with more questions than answers [11,12,13,14].
This review aims to describe the current fund of knowledge regarding the impact of bowel preparation on the human gut microbiome in colorectal surgery. Additionally, it will underscore key questions pertinent to future microbiome research and their implications for surgical outcomes after colorectal surgery.

2. Bowel Preparation

2.1. Evolution of Bowel Preparation

In the early 20th century, bowel preparation, also referred to as “intestinal asepsis”, emerged in response to elevated rates of infectious complications among patients undergoing colorectal surgery [15]. During this period, postoperative mortality rates ranged from 10 to 12%, with infectious complications being the primary cause. Meanwhile, over 20% of patients experienced an anastomotic leak, and of the patients who survived the surgery, 80–90% suffered from a wound infection [16]. To mitigate these alarming infection rates, MBP was introduced, which included starting patients on a special diet and/or laxatives, in hopes that reducing fecal bacterial load would decrease surgical field contamination and minimize subsequent infectious complications [15]. An additional proposed benefit was that eliminating hard feces would prevent undue pressure and ischemia on the new colonic anastomosis and consequently reduce anastomotic leak rates [17]. Despite conflicting evidence for the efficacy of MBP in reducing bacterial counts, its routine adoption became entrenched in colorectal surgery practice by the 1970s [15,18]. This practice persisted into the modern surgical era, with various combinations of dietary restrictions, enemas, and large-volume saline irrigation via a nasogastric tube. The introduction of polyethylene glycol made MBP more tolerable for the patient by reducing bowel preparation time and electrolyte derangements seen with saline irrigation [15,19]. Although strong evidence behind the use of MBP was sparse, a 1996 national survey of 471 North American colorectal surgeons found that 100% of them endorsed the use of MBP [20,21].
The concurrent advancements in OA regimens further substantiated the use of MBP, as patients who participated in clinical trials evaluating different OA regimens also underwent MBP [22]. Cohn et al. emphasized the three best qualities of an OA regimen: strong bactericidal activity against colonic bacteria, prevention of pathogenic microorganism overgrowth, and a satisfactory patient tolerance profile with low GI tract absorption [23]. Since the most commonly isolated bacteria from SSIs after large bowel surgery were Escherichia coli and Bacteroides fragilis, early clinical trials evaluated OA efficacy by its ability to eliminate all aerobic and anerobic gut microflora [24]. Some of the first studied OAs were sulfonamide derivatives. However, sulfonamides were deemed an “unsatisfactory agent,” since they failed to eliminate the “streptococci, enterococci, coliform organisms, and bacteriodes” in bacteriologic stool studies [23,25]. Sellwood et al. found that 48 h of preoperative treatment with neomycin and bacitracin was superior to phthalylsulphathiazole, showing a significant reduction in streptococci and coliform bacteria. Interestingly, both antibiotics also reduced quantities of Bacteroides fragilis, which these agents are not known to be bactericidal against [26]. One review noted that these results emphasize “our lack of knowledge regarding the interaction of bacterial species and their possible dependence on each other for survival in nature” [24]. Researchers are still unraveling the complexities of gut bacterial interactions, and our understanding of the gut microbiome’s dynamics continues to evolve.
In the 1970s, several clinical trials were conducted on various combinations of OAs in surgical bowel preparation, aiming to further reduce colonic bacterial counts and SSIs [25,27,28]. A notable clinical trial by Nichols et al. compared surgical outcomes and differences in colonic flora amongst elective colorectal surgery patients who underwent MBP followed by preoperative erythromycin–neomycin or no OAs [27]. The results were impressive. Despite the high prevalence of SSIs after colorectal surgery during this era, none of the 20 randomized patients contracted a wound infection, nor did any of the 69 retrospectively analyzed patients who underwent the same preoperative regimen [27]. Furthermore, stool studies revealed the suppression of both anerobic and aerobic bacteria in the erythromycin–neomycin cohort, which the authors attributed as a likely explanation for their lack of SSIs [27]. In addition, Clarke et al. conducted a randomized, double-blind clinical trial on 116 elective colorectal surgery patients who underwent MBP followed by either preoperative neomycin–erythromycin or placebo. The septic complication rate was 9% in the neomycin–erythromycin group compared to 43% in the placebo group [28]. In addition to several other studies, the consensus was that elective colon resection should include both preoperative OA and MBP [27,28].

2.2. Mechanical Bowel Preparation Controversy

New evidence emerged regarding the relatively low infection rates following emergency colon surgery, in which patients do not undergo preoperative MBP [29]. This spurred numerous clinical trials in the 1990s and early 2000s, the results of which challenged the prevailing “surgical dogma” regarding the efficacy of MBP in improving surgical outcomes [29,30]. When comparing a mechanically prepped vs. unprepped colon, several studies failed to demonstrate significant differences in surgical outcomes, leading to their conclusion that MBP is not necessary [21,30,31,32]. In one randomized trial, Santos et al. suggested that MBP may even be detrimental, as they observed higher complication rates in the MBP cohort (MBP 21% vs. no MBP 11%, p < 0.05), particularly an increase in wound infections (MBP 17% vs. No MBP 9%, p < 0.05) [33]. Interestingly, they also analyzed the bacterial composition of the bowel content and peritoneal fluid and found no significant differences in bacterial quantity or composition between patients who underwent MBP and those who did not, which challenged the pre-existing notion that MBP reduces colonic bacterial counts [33]. In a meta-analysis encompassing seven randomized clinical trials conducted between 1992 and 2005, Bucher at al. found that elective colorectal surgery patients who received MBP were more likely to suffer an anastomotic leak (MBP 36/642 vs. no MBP 18/655; OR 1.84; p = 0.03) [34]. Infection rates were also higher in the MBP group but failed to reach statistical significance [34]. Lastly, a Cochrane review by Guenaga et al. pooled 5805 clinical trial participants who did or did not receive MBP prior to elective colorectal surgery. Based on their measured outcomes of anastomotic leak and wound infection, they concluded that there was no significant evidence to support the benefit of MBP for patients undergoing colorectal surgery, and it can be “safely omitted” [35].
The controversial evidence related to MBP has caused contention amongst international guidelines pertaining to the optimal bowel preparation prior to elective colorectal surgery. The Enhanced Recovery After Surgery (ERAS) Society and the United Kingdom’s National Institute for Health and Care Excellence recommend against the routine use of MBP [36,37]. Meanwhile, the 2018 WHO, 2019 ASCRS, and 2021 Japan Society for Surgical Infection guidelines support the combined use of OAs along with MBP prior to elective colorectal surgery [3,5,38].
A major criticism of past clinical trials assessing SBP is the lack of granular data between colon versus rectal surgery. Rectal surgery patients, who have often received neoadjuvant chemoradiation, incur a higher SSI rate, particularly in the setting of low colorectal or coloanal anastomosis [39]. Past clinical trials also did not account for the use of minimally invasive surgical (MIS) techniques, which have further decreased SSIs and are now the gold standard for many elective colon and rectal surgeries [40]. One retrospective study using the National Surgical Quality Improvement Program (NSQIP) database compared SSI rates between MIS colon and rectal surgeries [41]. Amongst 12,417 rectal cancer patients, superficial and organ space infection rates were similar; however, the combined OA and MBP group had a significantly lower rate of deep surgical infections (0.1% vs. 0.9% p = 0.004). For colon cancer resection, the OA and MBP group incurred lower rates of superficial SSIs (1.1% vs. 1.9% p = 0.043), but deep and organ space infections had similar rates [41]. The recently published multicenter, double-blind MOBILE-2 trial randomized 565 patients who underwent MIS or open rectal surgery to receive MBP followed by either oral neomycin and metronidazole or placebo. The OA and MBP cohort experienced a lower rate of postoperative complications, fewer SSIs, and less anastomotic dehiscence [42]. Although compelling, more research is needed to determine the efficacy of SBP for rectal surgery.
Preoperative bowel preparation, in its various iterations, has been around for almost a century. Yet, the evolving and conflicting evidence surrounding SBP showcases our limited understanding of its true mechanism, the impact on the gut microbiome, and potential association with surgical outcomes. If researchers and clinicians aim to further reduce SSIs, more investigation into the gut microbiome may be a promising area to explore.

3. Microbiome

The individual configuration and function of the adult gut microbiota are governed by diet and host factors that regulate and direct microbial growth. Disruptions of this homeostasis, known as dysbiosis, have been increasingly linked to a number of human illnesses [43]. The National Institute of Health’s Human Microbiome Project, which aims to further characterize and comprehend how the microbiome affects human health and disease, has established three important factual properties about the human microbiome: (1) the microbiome is exceedingly complex, (2) the spatial distribution of microbes within the body is nonrandom, and (3) microbes associated with the human body can be beneficial, neutral, or pathogenic [44,45]. Understanding the factors that influence microbiota composition, function, and their roles in particular disease processes or pharmacological interventions requires the utilization of metabolomics, proteomics, and metatranscriptomics [8]. These tools have allowed researchers, particularly within colorectal surgical specialties, to analyze the impact of SBP on the gut microbiota.
In the adult human gastrointestinal tract, the total number of commensal enteric bacteria varies greatly from ~102,3 cells/gram in the jejunum and proximal ileum to ~107,8 in the distal ileum, and ~1011,12 within the ascending colon [46]. Human studies have identified 2100 species classified into 12 different phyla with 16S rRNA gene sequencing analyses, demonstrating that more than 90% of bacterial species found within the gut belong to 4 phyla: Proteobacteria, Actinobacteria, Firmicutes, and Bacteroidetes [46]. Obligately anaerobic primary fermenters, belonging to the classes Bacteroidia (phylum Bacteroidetes) and Clostridia (phylum Firmicutes), dominate the high-density microbial community in the large intestine due to the limited availability of oxygen and nitrates [47,48]. In contrast, Enterobacteriaceae (phylum Proteobacteria) are found in the small intestine, due to shorter transit time and high bile concentrations, but not the colon [49]. Besides nonrandom spatial distribution, gut microbiota also differs by age. Microbiota diversity in children is dominated by Akkermansia muciniphila, Bacteroides, Clostridium coccoides spp., Clostridium botulinum spp., and Veillonella [50]. However, by age 3, an individual’s gut microbiota becomes comparable to that of adults, largely persisting until people become elderly [51]. At that point, due to poorly understood mechanisms, elderly individuals typically exhibit decreased Bifidobacterium and increased Clostridium and Proteobacteria [52]. In addition to generalized compositional changes observed over time, the Firmicutes to Bacteroidetes ratio has been previously used to characterize dysbiosis in diseased states [53].
The intestinal microbiota serves a variety of functions critical to wound healing, including energy extraction in the form of short-chain fatty acid (SCFA) production and the modulation of pro- and anti-inflammatory immune responses [54]. Butyric acid is associated with the maintenance of gut barrier function and integrity via the regulation of tight junction proteins [55,56]. The depletion of SCFA, including butyric acid, results in gut and systemic inflammation, delayed wound healing, and gut barrier disruption [57,58]. Our group has previously demonstrated that butyric acid levels decreased by 80% after bowel preparation and surgical intervention [14].
Furthermore, biomarkers reflecting gut permeability have been studied, including zonulin and tight junction proteins zonula occludens-1 (ZO-1) and occludin. Zonulin is a primary regulator of intestinal permeability and is implicated in IBD, autoimmune diseases, obesity, and diabetes [59,60,61]. Enteric pathogens are a main trigger for zonulin release, which causes the displacement of ZO-1 and occludin from the tight junction complex [62]. If disrupted, tight junction proteins are released from the barrier and are detectable in blood, reflecting increased gut permeability [60,63,64].
Finally, the effects of antibiotic-induced disruption of the microbiome in association with alterations to the immune system have been studied in Rhesus macaques [65]. Changes in colonic and systemic immunity were seen in the colon with the recruitment of neutrophils, CD4+ T helper 17 (Th17), which produces IL-17, and T helper 22 (Th22) cells, which are major sources of IL-22. IL-17 promotes neutrophil recruitment and synergizes with IL-22 to activate epithelial defense through the expression of anti-bacterial proteins and peptides. These cytokines are crucial in maintaining mucosal immunity against pathogenic bacteria and promoting epithelial regeneration and repair [66,67,68].
Despite all of the characterized changes, absent intervention, the human gut microbiota is largely stable, with one study reporting that >60% of intestinal microbial strains are retained in their host over the course of 5 years [69]. While dynamic alterations of each individual’s microbiota occur in response to a myriad of inputs, such as dietary influences, tissue injury, host and microbiota-specific byproducts of metabolism, we aim to highlight the compositional gut microbiota changes in response to SBP and its components commonly used by colorectal surgery patients.

4. Impact of Bowel Preparation on the Microbiome

Despite being commonly employed and highly debated over decades, SBP, the combination of preoperative OA, intravenous antibiotics, and/or MBP, has a poorly understood mechanism. Designed with the goal to broadly decontaminate the intestine, a recent critique of this extensive microbial depletion is that perturbations in the diverse and commensal microbial community can lead to compositional shifts favoring the proliferation of pathologic genera that can negatively impact wound healing and recovery [70].
Previous interventional studies have demonstrated gut microbial sensitivity to antibiotics and bowel surgery [71,72,73,74]. Dethlefsen et al. demonstrated that broad-spectrum antibiotic therapy resulted in long-lasting, although not permanent, alterations in intestinal colonization profiles [72]. In addition, Croswell et al. observed that antibiotic administration led to an increased susceptibility to pathogenic organisms, such as Salmonella enterica [75]. Fang et al. compared inflammatory bowel disease (IBD) patients undergoing ileocolonic resection or colectomy to no surgery and observed decreased alpha diversity in the microbiome and metabolome, as well as an elevated relative abundance of E. coli in surgery patients [76]. In a systematic literature review, Ferrie et al. reported detailed outcomes from eight studies that examined the effect of surgery on microbiota alterations in patients with IBD, and they observed several compositional changes with beneficial, harmful, and unclear effects of differing magnitudes [77]. A similar variability of outcomes was observed in four studies looking at the impact of colorectal surgery on the microbiota from the same systematic review [77]. While a link between antibiotic administration as well as surgical intervention and their respective changes on the gut microbiota undoubtedly exists, the current state of the literature highlights an abundance of raw data and a paucity of causal pathways.
Despite studies calling into question the utility of MBP on clinical outcomes, few studies have examined the effects of MBP on microbiota compositional shifts with somewhat equivocal results [12,78,79,80]. O’Brien et al. compared stool samples from colonoscopy patients who completed MBP to controls not undergoing a procedure, finding that a small number of subjects had short-term changes but no variance in composition between the two groups, leading to their conclusion that bowel preparation does not have a lasting effect [80]. Despite this controversial negative result, the bulk of evidence supports that MBP can cause widespread and potentially lasting compositional changes [12,81,82,83]. Drago et al. conducted a diet-controlled study of 10 patients undergoing colonoscopy who received 4 L of polyethylene glycol solution and provided three stool samples: 1 week preoperatively, immediately before colonoscopy, and 1 month post-procedure. In these patients, MBP resulted in significant compositional shifts within the microbiota at phylum, class, and family level between pre- and immediately post-bowel lavage. While most of these changes reverted to baseline at the 1-month mark, a persistent reduction in Lactobacillaceae and Enterobacteriaceae occurred while an abundance of Rikenellaceae, Eubacteriaceae, and Streptococcaceae was observed (p < 0.05) [12]. From a functional standpoint, the impact of these changes is difficult to interpret. Lactobacillaceae are known to foster the development of the immune system, provide barrier function, and act to salvage energy as short-chain fatty acids [84,85]. Meanwhile, Enterobacteriaceae include many nosocomial pathogens with substantial antibiotic resistance, and Streptococcaceae is associated with increases in fecal proteases, which are known to cause inflammation, disrupt mucosal barrier function, and potentially provide a metabolic advantage for certain bacteria [86,87,88].
While individual components of SBP have been shown to cause compositional changes within the gut microbiota, even fewer studies have examined the impact of SBP as a whole. Our group previously published a longitudinal pilot study comparing MBP alone in patients undergoing colonoscopy versus SBP (OA + MBP) in patients undergoing colon surgery [14]. In line with the previous literature, patients in the MBP alone arm had minimal microbiota alterations with rapid return to baseline. However, the SBP group demonstrated larger shifts that persisted for an average of 1 month, a decrease in α diversity, and an increased abundance of Lactobacillus, Enterococcus, and Streptococcus. These results in patients undergoing colon surgery differ from the published data by Drago et al., [12] potentially suggesting that oral antibiotics and surgical stress, the two additional components of the colon surgery arm, as described by Nalluri-Butz et al., play an important role in long-term microbiome compositional alterations (Figure 1). Taken together, although relative abundances of bacteria pre- and post-intervention provide insight into possible downstream functional impacts, the gut microbiota remains a partially understood and exceedingly complex entity.

5. Role of Probiotics in Colorectal Surgery

Paralleling the growing fund of knowledge regarding the human gut microbiome and its impact on colorectal patients, the use of probiotics has been an area of research aimed at reducing infectious complications after colorectal surgery [89,90]. Probiotics may help maintain homeostasis within the gut microbiome, which, as discussed, can be disrupted by SBP. Several clinical trials have demonstrated that probiotics aid in maintaining intestinal integrity, reducing bacterial translocation, and promoting a greater number of beneficial versus pathogenic microorganisms [91,92,93]. A systematic review and meta-analysis of 21 clinical trials comparing perioperative probiotics or synbiotics, which are combinations of probiotics with indigestible food ingredients that stimulate bacterial activity to placebo or standard of care in elective colorectal surgery, found significantly fewer pulmonary and urinary tract infections [90]. Interestingly, there was no difference in anastomotic leaks or wound infections [90]. The use of perioperative probiotics shows promise in reducing postoperative complications. However, there is currently a wide variety of probiotic products available, and the effect of specific probiotic strains on the gut microbiome, as well as surgical outcomes, is unknown.

6. Clinical Implications and Areas of Future Study

The current body of clinical evidence regarding the effect of bowel preparation on the microbiome in colorectal surgery is expanding. However, conflicting results have hindered the formation of a clear consensus on targeted interventions for patient care [94]. Another challenge in detecting the effect of microbiome changes on patient outcomes arises from the overall low event rates of complications such as anastomotic leaks and surgical site infections in colorectal surgery. At present, interventions directed at beneficial microbiota alteration should not supersede current recommendations for asepsis practices, as the published literature remains largely exploratory. The translation of microbiome data into impactful clinical changes will require the discovery of causal and modifiable pathways that implicate specific microbiota for targeted intervention. With the advancement in technological capabilities, researchers will be better equipped to study the interactions between the human host and microbiome, paving the way for new therapies geared towards microbiome-precision medicine [95].

7. Limitations

Studies of the gut microbiome, by their essence, unintentionally incorporate many confounders that impact the quality of study results, including lifestyle or environmental factors, as well as diet. The impact of pre-surgical fasting, preoperative administration of prebiotics or probiotics, and controlling for the use of bowel preparation or delivery of different antibiotics can all significantly impact study results when examining the microbiome. Additionally, the method of sample preservation and analysis has the potential to alter the results. The gold standard for long-term fecal sample preservation is storage in a −80 °C freezer, and standardization of this process could lead to more generalizable results.
As technology improves, detailed characterization of the microbiota and their components will also be enhanced. At present, comparisons between studies examining similar concepts yet utilizing different analytic techniques can generate unique results that can obscure the narrative [96]. This dilemma, ubiquitous in fields at the forefront of scientific discovery, remains an ever-present issue when attempting to link the human microbiome to an ever-increasing number of disease states.
Future research incorporating methodological rigor to control these confounding variables and standardized analytic techniques is essential as we attempt to define causal pathways linking gut microbiota changes with perioperative surgical care.

8. Conclusions

Preoperative bowel preparation has successfully reduced the rates of infectious complications in patients undergoing colorectal surgery, yet, until recently, its impact on the gut microbiome has been overlooked. As our collective knowledge of the gut microbiome continues to evolve, we will face many challenges in translating data into impactful clinical interventions. However, harnessing the power of the gut microbiome holds great promise in helping researchers and clinicians achieve their overarching goal of improving patient outcomes in colorectal surgery.

Author Contributions

L.W.: project conception and design, drafting and editing manuscript. A.T.: project conception and design, drafting and editing manuscript. C.J.: editing manuscript, final approval. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Minnesota Colorectal Cancer Research Foundation, a 501(c)(3) nonprofit foundation.

Conflicts of Interest

None of the authors have any conflicts of interest to declare.

Abbreviations

ASCRSAmerican Society of Colon and Rectal Surgeons
ERASEnhanced Recovery After Surgery
IBDInflammatory bowel disease
MBPMechanical bowel preparation
NSQIPNational Surgical Quality Improvement Program
OAOral antibiotic
SBPSurgical bowel preparation
SSISurgical site infection

References

  1. Petrou, N.A.; Kontovounisios, C. The Use of Mechanical Bowel Preparation and Oral Antibiotic Prophylaxis in Elective Colorectal Surgery: A Call for Change in Practice. Cancers 2022, 14, 5990. [Google Scholar] [CrossRef] [PubMed]
  2. Young, H.; Knepper, B.; Moore, E.E.; Johnson, J.L.; Mehler, P.; Price, C.S. Surgical Site Infection after Colon Surgery: National Healthcare Safety Network Risk Factors and Modeled Rates Compared with Published Risk Factors and Rates. J. Am. Coll. Surg. 2012, 214, 852–859. [Google Scholar] [CrossRef] [PubMed]
  3. World Health Organization. Global Guidelines for the Prevention of Surgical Site Infection, 2nd ed.; World Health Organization: Geneva, Switzerland, 2018; Available online: https://iris.who.int/handle/10665/277399 (accessed on 14 May 2024).
  4. Ban, K.A.; Minei, J.P.; Laronga, C.; Harbrecht, B.G.; Jensen, E.H.; Fry, D.E.; Itani, K.M.; E Dellinger, P.; Ko, C.Y.; Duane, T.M. American College of Surgeons and Surgical Infection Society: Surgical Site Infection Guidelines, 2016 Update. J. Am. Coll. Surg. 2017, 224, 59–74. [Google Scholar] [CrossRef]
  5. Migaly, J.; Bafford, A.C.; Francone, T.D.; Gaertner, W.B.; Eskicioglu, C.; Bordeianou, L.; Feingold, D.L.; Steele, S.R. The American Society of Colon and Rectal Surgeons Clinical Practice Guidelines for the Use of Bowel Preparation in Elective Colon and Rectal Surgery. Dis Colon Rectum. 2019, 62, 3–8, Correction in Dis. Colon. Rectum. 2019, 62, e436. [Google Scholar] [CrossRef]
  6. Alverdy, J.C.; Hyman, N.; Gilbert, J.; Luo, J.N.; Krezalek, M. Preparing the Bowel for Surgery: Learning from the Past and Planning for the Future. J. Am. Coll. Surg. 2017, 225, 324–332. [Google Scholar] [CrossRef] [PubMed]
  7. Russ, A.J.; Casillas, M.A. Gut Microbiota and Colorectal Surgery: Impact on Postoperative Complications. Clin. Colon. Rectal Surg. 2016, 29, 253–257. [Google Scholar] [CrossRef]
  8. Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Vergès, M.-C.C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G.H.; et al. Microbiome definition re-visited: Old concepts and new challenges. Microbiome 2020, 8, 103, Correction in Microbiome 2020, 8, 119. [Google Scholar] [CrossRef]
  9. Hou, K.; Wu, Z.-X.; Chen, X.-Y.; Wang, J.-Q.; Zhang, D.; Xiao, C.; Zhu, D.; Koya, J.B.; Wei, L.; Li, J.; et al. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar] [CrossRef] [PubMed]
  10. Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef]
  11. Shobar, R.M.; Velineni, S.; Keshavarzian, A.; Swanson, G.; DeMeo, M.T.; E Melson, J.; Losurdo, J.; Engen, P.A.; Sun, Y.; Koenig, L.; et al. The Effects of Bowel Preparation on Microbiota-Related Metrics Differ in Health and in Inflammatory Bowel Disease and for the Mucosal and Luminal Microbiota Compartments. Clin. Transl. Gastroenterol. 2016, 7, e143. [Google Scholar] [CrossRef]
  12. Drago, L.; Toscano, M.; De Grandi, R.; Casini, V.; Pace, F. Persisting changes of intestinal microbiota after bowel lavage and colonoscopy. Eur. J. Gastroenterol. Hepatol. 2016, 28, 532–537. [Google Scholar] [CrossRef]
  13. Harrell, L.; Wang, Y.; Antonopoulos, D.; Young, V.; Lichtenstein, L.; Huang, Y.; Hanauer, S.; Chang, E. Standard Colonic Lavage Alters the Natural State of Mucosal-Associated Microbiota in the Human Colon. PLoS ONE 2012, 7, e32545. [Google Scholar] [CrossRef] [PubMed]
  14. Nalluri-Butz, H.; Bobel, M.C.; Nugent, J.; Boatman, S.; Emanuelson, R.; Melton-Meaux, G.; Madoff, R.D.; Jahansouz, C.; Staley, C.; Gaertner, W.B. A pilot study demonstrating the impact of surgical bowel preparation on intestinal microbiota composition following colon and rectal surgery. Sci. Rep. 2022, 12, 10559. [Google Scholar] [CrossRef] [PubMed]
  15. Duncan, J.E.; Quietmeyer, C.M. Bowel preparation: Current status. Clin. Colon. Rectal Surg. 2009, 22, 14–20. [Google Scholar] [CrossRef]
  16. Poth, E.J. Historical development of intestinal antisepsis. World J. Surg. 1982, 6, 153–159. [Google Scholar] [CrossRef]
  17. Kumar, A.S.; Kelleher, D.C.; Sigle, G.W. Bowel Preparation before Elective Surgery. Clin. Colon. Rectal. Surg. 2013, 26, 146–152. [Google Scholar] [CrossRef] [PubMed]
  18. Keighley, M.R. A clinical and physiological evaluation of bowel preparation for elective colorectal surgery. World J. Surg. 1982, 6, 464–470. [Google Scholar] [CrossRef] [PubMed]
  19. Davis, G.R.; Santa Ana, C.A.; Morawski, S.G.; Fordtran, J.S. Development of a lavage solution associated with minimal water and electrolyte absorption or secretion. Gastroenterology 1980, 78, 991–995. [Google Scholar] [CrossRef] [PubMed]
  20. Nichols, R.L.; Smith, J.W.; Garcia, R.Y.; Waterman, R.S.; Holmes, J.W. Current practices of preoperative bowel preparation among North American colorectal surgeons. Clin. Infect. Dis. 1997, 24, 609–619. [Google Scholar] [CrossRef]
  21. Zmora, O.; Wexner, S.D.; Hajjar, L.; Park, T.; Efron, J.E.; Nogueras, J.J.; Weiss, E.G. Trends in Preparation for Colorectal Surgery: Survey of the Members of the American Society of Colon and Rectal Surgeons. Am. Surg. 2003, 69, 150–154. [Google Scholar] [CrossRef]
  22. Rosenberg, I.L.; Graham, N.G.; De Dombal, F.T.; Goligher, J.C. Preparation of the intestine in patients undergoing major large-bowel surgery, mainly for neoplasms of the colon and rectum. Br. J. Surg. 1971, 58, 266–269. [Google Scholar] [CrossRef]
  23. Cohn, I., Jr. Kanamycin for bowel sterilization. Ann. N. Y. Acad. Sci. 1958, 76, 212–223. [Google Scholar] [CrossRef] [PubMed]
  24. Bowel preparation for surgery. Lancet 1978, 2, 1132–1133.
  25. Nichols, R.L.; Condon, R.E. Antibiotic preparation of the colon: Failure of commonly used regimens. Surg. Clin. N. Am. 1971, 51, 223–231. [Google Scholar] [CrossRef] [PubMed]
  26. Sellwood, R.A.; Burn, J.I.; Waterworth, P.M.; Welbourn, R.B. A second clinical trial to compare two methods for preoperative preparation of the large bowel. Br. J. Surg. 1969, 56, 610–612. [Google Scholar] [CrossRef] [PubMed]
  27. Nichols, R.L.; Broido, P.; Condon, R.E.; Gorbach, S.L.; Nyhus, L.M. Effect of preoperative neomycin-erythromycin intestinal preparation on the incidence of infectious complications following colon surgery. Ann. Surg. 1973, 178, 453–462. [Google Scholar] [CrossRef]
  28. Clarke, J.S.; Condon, R.E.; Bartlett, J.G.; Gorbach, S.L.; Nichols, R.L.; Ochi, S. Preoperative oral antibiotics reduce septic complications of colon operations: Results of prospective, randomized, double-blind clinical study. Ann. Surg. 1977, 186, 251–259. [Google Scholar] [CrossRef]
  29. Ju, Y.U.; Min, B.W. A Review of Bowel Preparation Before Colorectal Surgery. Ann. Coloproctol. 2021, 37, 75–84. [Google Scholar] [CrossRef] [PubMed]
  30. Platell, C.; Hall, J. What is the role of mechanical bowel preparation in patients undergoing colorectal surgery? Dis. Colon. Rectum. 1998, 41, 875–883. [Google Scholar] [CrossRef]
  31. Young Tabusso, F.; Celis Zapata, J.; Berrospi Espinoza, F.; Payet Meza, E.; Ruiz Figueroa, E. Preparación mecánica en cirugía electiva colo-rectal. Costumbre o necesidad? [Mechanical preparation in elective colorectal surgery, a usual practice or a necessity?]. Rev. Gastroenterol. Peru 2002, 22, 152–158. [Google Scholar]
  32. Burke, P.; Mealy, K.; Gillen, P.; Joyce, W.; Traynor, O.; Hyland, J. Requirement for bowel preparation in colorectal surgery. Br. J. Surg. 1994, 81, 907–910. [Google Scholar] [CrossRef] [PubMed]
  33. Santos, J.C., Jr.; Batista, J.; Sirimarco, M.T.; Guimarães, A.S.; Levy, C.E. Prospective randomized trial of mechanical bowel preparation in patients undergoing elective colorectal surgery. Br. J. Surg. 1994, 81, 1673–1676. [Google Scholar] [CrossRef]
  34. Bucher, P.; Mermillod, B.; Gervaz, P.; Morel, P. Mechanical bowel preparation for elective colorectal surgery: A meta-analysis. Arch. Surg. 2004, 139, 1359–1365. [Google Scholar] [CrossRef] [PubMed]
  35. Güenaga, K.F.; Matos, D.; Wille-Jørgensen, P. Mechanical bowel preparation for elective colorectal surgery. Cochrane Database Syst. Rev. 2011, 2011, CD001544. [Google Scholar] [CrossRef] [PubMed]
  36. Gustafsson, U.O.; Scott, M.J.; Schwenk, W.; Demartines, N.; Roulin, D.; Francis, N.; McNaught, C.E.; MacFie, J.; Liberman, A.S.; Soop, M.; et al. Guidelines for perioperative care in elective colonic surgery: Enhanced Recovery After Surgery (ERAS(®)) Society recommendations. World J. Surg. 2013, 37, 259–284. [Google Scholar] [CrossRef] [PubMed]
  37. NICE. Surgical Site Infections: Prevention and Treatment; National Institute for Health and Care Excellence (NICE): London, UK, 2019; Available online: https://www.nice.org.uk/guidance/ng125/resources/surgical-site-infections-prevention-and-treatment-pdf-66141660564421 (accessed on 14 May 2024).
  38. Ohge, H.; Mayumi, T.; Haji, S.; Kitagawa, Y.; Kobayashi, M.; Kobayashi, M.; Mizuguchi, T.; Mohri, Y.; Sakamoto, F.; Shimizu, J.; et al. The Japan Society for Surgical Infection: Guidelines for the prevention, detection, and management of gastroenterological surgical site infection, 2018. Surg. Today 2021, 51, 1–31. [Google Scholar] [CrossRef] [PubMed]
  39. Konishi, T.; Watanabe, T.; Kishimoto, J.; Nagawa, H. Elective colon and rectal surgery differ in risk factors for wound infection: Results of prospective surveillance. Ann. Surg. 2006, 244, 758–763. [Google Scholar] [CrossRef]
  40. Singh, S.S.; Shinde, R.K. Minimally Invasive Gastrointestinal Surgery: A Review. Cureus 2023, 15, e48864. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  41. Abd El Aziz, M.A.; Grass, F.; Calini, G.; Behm, K.T.; D’angelo, A.-L.; Kelley, S.R.; Mathis, K.L.; Larson, D.W.M. Oral Antibiotics Bowel Preparation Without Mechanical Preparation for Minimally Invasive Colorectal Surgeries: Current Practice and Future Prospects. Dis Colon. Rectum. 2022, 65, e897–e906. [Google Scholar] [CrossRef]
  42. Koskenvuo, L.; Lunkka, P.; Varpe, P.; Hyöty, M.; Satokari, R.; Haapamäki, C.; Lepistö, A.; Sallinen, V. Mechanical bowel preparation and oral antibiotics versus mechanical bowel preparation only prior rectal surgery (MOBILE2): A multicentre, double-blinded, randomised controlled trial—Study protocol. BMJ Open 2021, 11, e051269. [Google Scholar] [CrossRef]
  43. Lee, J.Y.; Tsolis, R.M.; Bäumler, A.J. The microbiome and gut homeostasis. Science 2022, 377, eabp9960. [Google Scholar] [CrossRef] [PubMed]
  44. Peterson, J.; Garges, S.; Giovanni, M.; McInnes, P.; Wang, L.; Schloss, J.A.; Bonazzi, V.; McEwen, J.E.; Wetterstrand, K.A.; Deal, C.; et al. The NIH human microbiome project. Genome Res. 2009, 19, 317–2323. [Google Scholar]
  45. Morowitz, M.J.; Babrowski, T.; Carlisle, E.M.; Olivas, A.; Romanowski, K.S.; Seal, J.B.; Liu, D.C.; Alverdy, J.C. The Human Microbiome and Surgical Disease. Ann. Surg. 2011, 253, 1094–1101. [Google Scholar] [CrossRef]
  46. Lloyd-Price, J.; Mahurkar, A.; Rahnavard, G.; Crabtree, J.; Orvis, J.; Hall, A.B.; Brady, A.; Creasy, H.H.; McCracken, C.; Giglio, M.G.; et al. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature 2017, 550, 61–66, Correction in Nature 2017, 551, 256. [Google Scholar] [CrossRef] [PubMed]
  47. Tropini, C.; Earle, K.A.; Huang, K.C.; Sonnenburg, J.L. The Gut Microbiome: Connecting Spatial Organization to Function. Cell Host Microbe 2017, 21, 433–442. [Google Scholar] [CrossRef]
  48. Sundin, O.H.; Mendoza-Ladd, A.; Zeng, M.; Diaz-Arévalo, D.; Morales, E.; Fagan, B.M.; Ordoñez, J.; Velez, P.; Antony, N.; McCallum, R.W. The human jejunum has an endogenous microbiota that differs from those in the oral cavity and colon. BMC Microbiol. 2017, 17, 160. [Google Scholar] [CrossRef]
  49. Flint, H.J.; Scott, K.P.; Louis, P.; Duncan, S.H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 577–589. [Google Scholar] [CrossRef] [PubMed]
  50. Amabebe, E.; Robert, F.O.; Agbalalah, T.; Orubu, E.S.F. Microbial dysbiosis-induced obesity: Role of gut microbiota in homoeostasis of energy metabolism. Br. J. Nutr. 2020, 123, 1127–1137. [Google Scholar] [CrossRef]
  51. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  52. Guigoz, Y.; Doré, J.; Schiffrin, E.J. The inflammatory status of old age can be nurtured from the intestinal environment. Curr. Opin. Clin. Nutr. Metab. Care 2008, 11, 13–20. [Google Scholar] [CrossRef]
  53. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef] [PubMed]
  54. Mulders, R.J.; de Git, K.C.G.; Schéle, E.; Dickson, S.L.; Sanz, Y.; Adan, R.A.H. Microbiota in obesity: Interactions with enteroendocrine, immune and central nervous systems. Obes. Rev. 2018, 19, 435–451. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, G.; Ran, X.; Li, B.; Li, Y.; He, D.; Huang, B.; Fu, S.; Liu, J.; Wang, W. Sodium Butyrate Inhibits Inflammation and Maintains Epithelium Barrier Integrity in a TNBS-induced Inflammatory Bowel Disease Mice Model. EBioMedicine 2018, 30, 317–325. [Google Scholar] [CrossRef] [PubMed]
  56. Zheng, L.; Kelly, C.J.; Battista, K.D.; Schaefer, R.; Lanis, J.M.; Alexeev, E.E.; Wang, R.X.; Onyiah, J.C.; Kominsky, D.J.; Colgan, S.P. Microbial-Derived Butyrate Promotes Epithelial Barrier Function through IL-10 Receptor-Dependent Repression of Claudin-2. J. Immunol. 2017, 199, 2976–2984. [Google Scholar] [CrossRef] [PubMed]
  57. Alam, A.; Leoni, G.; Quiros, M.; Wu, H.; Desai, C.; Nishio, H.; Jones, R.M.; Nusrat, A.; Neish, A.S. The microenvironment of injured murine gut elicits a local pro-restitutive microbiota. Nat. Microbiol. 2016, 1, 15021. [Google Scholar] [CrossRef] [PubMed]
  58. Ilhan, Z.E.; DiBaise, J.K.; Isern, N.G.; Hoyt, D.W.; Marcus, A.K.; Kang, D.-W.; Crowell, M.D.; E Rittmann, B.; Krajmalnik-Brown, R. Distinctive microbiomes and metabolites linked with weight loss after gastric bypass, but not gastric banding. ISME J. 2017, 11, 2047–2058. [Google Scholar] [CrossRef]
  59. Żak-Gołąb, A.; Kocełak, P.; Aptekorz, M.; Zientara, M.; Juszczyk, Ł.; Martirosian, G.; Chudek, J.; Olszanecka-Glinianowicz, M. Gut Microbiota, Microinflammation, Metabolic Profile, and Zonulin Concentration in Obese and Normal Weight Subjects. Int. J. Endocrinol. 2013, 2013, 674106. [Google Scholar] [CrossRef]
  60. Sapone, A.; de Magistris, L.; Pietzak, M.; Clemente, M.G.; Tripathi, A.; Cucca, F.; Lampis, R.; Kryszak, D.; Cartenì, M.; Generoso, M.; et al. Zonulin Upregulation Is Associated with Increased Gut Permeability in Subjects with Type 1 Diabetes and Their Relatives. Diabetes 2006, 55, 1443–1449. [Google Scholar] [CrossRef]
  61. Sturgeon, C.; Fasano, A. Zonulin, a regulator of epithelial and endothelial barrier functions, and its involvement in chronic inflammatory diseases. Tissue Barriers 2016, 4, e1251384. [Google Scholar] [CrossRef]
  62. Goldblum, S.E.; Rai, U.; Tripathi, A.; Thakar, M.; De Leo, L.; Di Toro, N.; Not, T.; Ramachandran, R.; Puche, A.C.; Hollenberg, M.D.; et al. The active Zot domain (aa 288-293) increases ZO-1 and myosin 1C serine/threonine phosphorylation, alters interaction between ZO-1 and its binding partners, and induces tight junction disassembly through proteinase activated receptor 2 activation. FASEB J. 2011, 25, 144–158. [Google Scholar] [CrossRef]
  63. Ott, B.; Skurk, T.; Hastreiter, L.; Lagkouvardos, I.; Fischer, S.; Büttner, J.; Kellerer, T.; Clavel, T.; Rychlik, M.; Haller, D.; et al. Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women. Sci. Rep. 2017, 7, 11955. [Google Scholar] [CrossRef] [PubMed]
  64. Olsson, A.; Gustavsen, S.; Langkilde, A.R.; Hansen, T.; Sellebjerg, F.; Søndergaard, H.B.; Oturai, A. Circulating levels of tight junction proteins in multiple sclerosis: Association with inflammation and disease activity before and after disease modifying therapy. Mult. Scler. Relat. Disord. 2021, 54, 103136. [Google Scholar] [CrossRef] [PubMed]
  65. Manuzak, J.A.; Zevin, A.S.; Cheu, R.; Richardson, B.; Modesitt, J.; Hensley-McBain, T.; Miller, C.; Gustin, A.T.; Coronado, E.; Gott, T.; et al. Antibiotic-induced microbiome perturbations are associated with significant alterations to colonic mucosal immunity in rhesus macaques. Mucosal Immunol. 2020, 13, 471–480, Correction in Mucosal Immunol. 2020, 13, 558. [Google Scholar] [CrossRef] [PubMed]
  66. Basu, R.; O’Quinn, D.B.; Silberger, D.J.; Schoeb, T.R.; Fouser, L.; Ouyang, W.; Hatton, R.D.; Weaver, C.T. Th22 cells are an important source of IL-22 for host protection against enteropathogenic bacteria. Immunity 2012, 37, 1061–1075. [Google Scholar] [CrossRef] [PubMed]
  67. Wang, Z.; Friedrich, C.; Hagemann, S.C.; Korte, W.H.; Goharani, N.; Cording, S.; Eberl, G.; Sparwasser, T.; Lochner, M. Regulatory T cells promote a protective Th17-associated immune response to intestinal bacterial infection with C. rodentium. Mucosal Immunol. 2014, 7, 1290–1301. [Google Scholar] [CrossRef] [PubMed]
  68. Liang, S.C.; Tan, X.-Y.; Luxenberg, D.P.; Karim, R.; Dunussi-Joannopoulos, K.; Collins, M.; Fouser, L.A. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J. Exp. Med. 2006, 203, 2271–2279. [Google Scholar] [CrossRef] [PubMed]
  69. Faith, J.J.; Guruge, J.L.; Charbonneau, M.; Subramanian, S.; Seedorf, H.; Goodman, A.L.; Clemente, J.C.; Knight, R.; Heath, A.C.; Leibel, R.L.; et al. The Long-Term Stability of the Human Gut Microbiota. Science 2013, 341, 1237439. [Google Scholar] [CrossRef] [PubMed]
  70. Gaines, S.; Shao, C.; Hyman, N.; Alverdy, J.C. Gut microbiome influences on anastomotic leak and recurrence rates following colorectal cancer surgery. Br. J. Surg. 2018, 105, e131–e141. [Google Scholar] [CrossRef] [PubMed]
  71. Dethlefsen, L.; Relman, D.A. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4554–4561. [Google Scholar] [CrossRef]
  72. Dethlefsen, L.; Huse, S.; Sogin, M.L.; Relman, D.A. The pervasive effects of an antibiotic on the human gut microbiota, as revealed by deep 16S rRNA sequencing. PLoS Biol. 2008, 6, e280. [Google Scholar] [CrossRef]
  73. Jakobsson, H.E.; Jernberg, C.; Andersson, A.F.; Sjölund-Karlsson, M.; Jansson, J.K.; Engstrand, L. Short-term antibiotic treatment has differing long-term impacts on the human throat and gut microbiome. PLoS ONE 2010, 5, e9836. [Google Scholar] [CrossRef] [PubMed]
  74. Hartman, A.L.; Lough, D.M.; Barupal, D.K.; Fiehn, O.; Fishbein, T.; Zasloff, M.; Eisen, J.A. Human gut microbiome adopts an alternative state following small bowel transplantation. Proc. Natl. Acad. Sci. USA 2009, 106, 17187–17192. [Google Scholar] [CrossRef]
  75. Croswell, A.; Amir, E.; Teggatz, P.; Barman, M.; Salzman, N.H. Prolonged Impact of Antibiotics on Intestinal Microbial Ecology and Susceptibility to Enteric Salmonella Infection. Infect. Immun. 2009, 77, 2741–2753. [Google Scholar] [CrossRef]
  76. Fang, X.; Vázquez-Baeza, Y.; Elijah, E.; Vargas, F.; Ackermann, G.; Humphrey, G.; Lau, R.; Weldon, K.C.; Sanders, J.G.; Panitchpakdi, M.; et al. Gastrointestinal Surgery for Inflammatory Bowel Disease Persistently Lowers Microbiome and Metabolome Diversity. Inflamm Bowel Dis. 2021, 27, 603–616, Correction in Inflamm. Bowel Dis. 2021, 27, 1368. [Google Scholar] [CrossRef] [PubMed]
  77. Ferrie, S.; Webster, A.; Wu, B.; Tan, C.; Carey, S. Gastrointestinal surgery and the gut microbiome: A systematic literature review. Eur. J. Clin. Nutr. 2021, 75, 12–25. [Google Scholar] [CrossRef]
  78. Pineda, C.E.; Shelton, A.A.; Hernandez-Boussard, T.; Morton, J.M.; Welton, M.L. Mechanical bowel preparation in intestinal surgery: A meta-analysis and review of the literature. J. Gastrointest. Surg. 2008, 12, 2037–2044. [Google Scholar] [CrossRef]
  79. Dahabreh, I.J.; Steele, D.W.; Shah, N.; Trikalinos, T.A. Oral mechanical bowel preparation for colorectal surgery: Systematic review and meta-analysis. Dis. Colon. Rectum. 2015, 58, 698–707. [Google Scholar] [CrossRef] [PubMed]
  80. O’Brien, C.L.; Allison, G.E.; Grimpen, F.; Pavli, P. Impact of colonoscopy bowel preparation on intestinal microbiota. PLoS ONE 2013, 8, e62815. [Google Scholar] [CrossRef]
  81. Gorkiewicz, G.; Thallinger, G.G.; Trajanoski, S.; Lackner, S.; Stocker, G.; Hinterleitner, T.; Gülly, C.; Högenauer, C. Alterations in the Colonic Microbiota in Response to Osmotic Diarrhea. PLoS ONE 2013, 8, e55817. [Google Scholar] [CrossRef]
  82. Jalanka, J.; Salonen, A.; Salojärvi, J.; Ritari, J.; Immonen, O.; Marciani, L.; Gowland, P.; Hoad, C.; Garsed, K.; Lam, C.; et al. Effects of bowel cleansing on the intestinal microbiota. Gut 2015, 64, 1562–1568. [Google Scholar] [CrossRef]
  83. Drago, L.; Valentina, C.; Fabio, P. Gut microbiota, dysbiosis and colon lavage. Dig. Liver. Dis. 2019, 51, 1209–1213. [Google Scholar] [CrossRef]
  84. McHeyzer-Williams, M. Local sentries for class switching. Nat. Immunol. 2007, 8, 230–232. [Google Scholar] [CrossRef] [PubMed]
  85. Di Cagno, R.; De Angelis, M.; De Pasquale, I.; Ndagijimana, M.; Vernocchi, P.; Ricciuti, P.; Gagliardi, F.; Laghi, L.; Crecchio, C.; Guerzoni, M.E.; et al. Duodenal and faecal microbiota of celiac children: Molecular, phenotype and metabolome characterization. BMC Microbiol. 2011, 11, 219. [Google Scholar] [CrossRef] [PubMed]
  86. Pace, N.R.; Olsen, G.J.; Woese, C.R. Ribosomal RNA phylogeny and the primary lines of evolutionary descent. Cell 1986, 45, 325–326. [Google Scholar] [CrossRef] [PubMed]
  87. Carroll, I.M.; Ringel-Kulka, T.; Ferrier, L.; Wu, M.C.; Siddle, J.P.; Bueno, L.; Ringel, Y. Fecal protease activity is associated with compositional alterations in the intestinal microbiota. PLoS ONE 2013, 8, e78017. [Google Scholar] [CrossRef]
  88. Carroll, I.M.; Maharshak, N. Enteric bacterial proteases in inflammatory bowel disease-pathophysiology and clinical implications. World J. Gastroenterol. 2013, 19, 7531–7543. [Google Scholar] [CrossRef]
  89. Persson, J.E.; Viana, P.; Persson, M.; Relvas, J.H.; Danielski, L.G. Perioperative or Postoperative Probiotics Reduce Treatment-Related Complications in Adult Colorectal Cancer Patients Undergoing Surgery: A Systematic Review and Meta-analysis. J. Gastrointest. Cancer 2024, 55, 740–748. [Google Scholar] [CrossRef] [PubMed]
  90. Veziant, J.; Bonnet, M.; Occean, B.V.; Dziri, C.; Pereira, B.; Slim, K. Probiotics/Synbiotics to Reduce Infectious Complications after Colorectal Surgery: A Systematic Review and Meta-Analysis of Randomised Controlled Trials. Nutrients 2022, 14, 3066. [Google Scholar] [CrossRef]
  91. Liu, Z.H.; Huang, M.-J.; Zhang, X.-W.; Wang, L.; Huang, N.-Q.; Peng, H.; Lan, P.; Peng, J.S.; Yang, Z.; Xia, Y.; et al. The effects of perioperative probiotic treatment on serum zonulin concentration and subsequent postoperative infectious complications after colorectal cancer surgery: A double-center and double-blind randomized clinical trial. Am. J. Clin. Nutr. 2013, 97, 117–126. [Google Scholar] [CrossRef] [PubMed]
  92. Zhang, J.W.; Du, P.; Gao, J.; Yang, B.R.; Fang, W.J.; Ying, C.M. Preoperative probiotics decrease postoperative infectious complications of colorectal cancer. Am. J. Med. Sci. 2012, 343, 199–205. [Google Scholar] [CrossRef]
  93. Pitsillides, L.; Pellino, G.; Tekkis, P.; Kontovounisios, C. The Effect of Perioperative Administration of Probiotics on Colorectal Cancer Surgery Outcomes. Nutrients 2021, 13, 1451. [Google Scholar] [CrossRef] [PubMed]
  94. Paine, H.; Jones, F.; Kinross, J. Preparing the Bowel (Microbiome) for Surgery: Surgical Bioresilience. Clin. Colon. Rectal. Surg. 2023, 36, 138–145. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  95. Kwa, W.T.; Sundarajoo, S.; Toh, K.Y.; Lee, J. Application of emerging technologies for gut microbiome research. Singap. Med. J. 2023, 64, 45–53. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  96. Roume, H.; Mondot, S.; Saliou, A.; Le Fresne-Languille, S.; Doré, J. Multicenter evaluation of gut microbiome profiling by next-generation sequencing reveals major biases in partial-length metabarcoding approach. Sci. Rep. 2023, 13, 22593. [Google Scholar] [CrossRef] [PubMed]
Figure 1. During homeostasis, the colon demonstrates a wide variety of microbial diversity. After preoperative surgical bowel preparation consisting of mechanical bowel preparation, oral antibiotics, and preoperative intravenous antibiotics along with colorectal surgery, postoperative microbial analyses show a reduction in α diversity and microbial-derived metabolites. There is a relative increase in Lactobacillus, Enterococcus, and Streptococcus species, as well as a reduction in Bacteroides, Roseburia, Faecalibacterium, and short-chain fatty acids.
Figure 1. During homeostasis, the colon demonstrates a wide variety of microbial diversity. After preoperative surgical bowel preparation consisting of mechanical bowel preparation, oral antibiotics, and preoperative intravenous antibiotics along with colorectal surgery, postoperative microbial analyses show a reduction in α diversity and microbial-derived metabolites. There is a relative increase in Lactobacillus, Enterococcus, and Streptococcus species, as well as a reduction in Bacteroides, Roseburia, Faecalibacterium, and short-chain fatty acids.
Antibiotics 13 00580 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Weaver, L.; Troester, A.; Jahansouz, C. The Impact of Surgical Bowel Preparation on the Microbiome in Colon and Rectal Surgery. Antibiotics 2024, 13, 580. https://doi.org/10.3390/antibiotics13070580

AMA Style

Weaver L, Troester A, Jahansouz C. The Impact of Surgical Bowel Preparation on the Microbiome in Colon and Rectal Surgery. Antibiotics. 2024; 13(7):580. https://doi.org/10.3390/antibiotics13070580

Chicago/Turabian Style

Weaver, Lauren, Alexander Troester, and Cyrus Jahansouz. 2024. "The Impact of Surgical Bowel Preparation on the Microbiome in Colon and Rectal Surgery" Antibiotics 13, no. 7: 580. https://doi.org/10.3390/antibiotics13070580

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop