Next Article in Journal
Adenocarcinomas of the Lung and Neurotrophin System: A Review
Next Article in Special Issue
Myxovirus resistance (Mx) Gene Diversity in Avian Influenza Virus Infections
Previous Article in Journal
Alcohol-Related Liver Disease: An Overview on Pathophysiology, Diagnosis and Therapeutic Perspectives
Previous Article in Special Issue
Retinol Binding Protein, Sunlight Hours, and the Influenza Virus-Specific Immune Response
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dysbiosis and Gastrointestinal Surgery: Current Insights and Future Research

1
Gastroenterology and Digestive Endoscopy Unit, Forlì-Cesena Hospitals, Ausl Romagna, 47121 Forlì-Cesena, Italy
2
Department of Medical and Surgical Sciences—DIMEC, Alma Mater Studiorum—University of Bologna, 40138 Bologna, Italy
3
General and Oncologic Surgery, Morgagni—Pierantoni Hospital, AUSL Romagna, 47121 Forlì-Cesena, Italy
4
Microbiology Unit, Hub Laboratory, AUSL della Romagna, 47121 Forlì-Cesena, Italy
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(10), 2532; https://doi.org/10.3390/biomedicines10102532
Submission received: 3 September 2022 / Revised: 2 October 2022 / Accepted: 3 October 2022 / Published: 10 October 2022
(This article belongs to the Special Issue Molecular Research in Infectious Diseases)

Abstract

:
Surgery of the gastrointestinal tract can result in deep changes among the gut commensals in terms of abundance, function and health consequences. Elective colorectal surgery can occur for neoplastic or inflammatory bowel disease; in these settings, microbiota imbalance is described as a preoperative condition, and it is linked to post-operative complications, as well. The study of bariatric patients led to several insights into the role of gut microbiota in obesity and after major surgical injuries. Preoperative dysbiosis and post-surgical microbiota reassessment are still poorly understood, and they could become a key part of preventing post-surgical complications. In the current review, we outline the most recent literature regarding agents and molecular pathways involved in pre- and post-operative dysbiosis in patients undergoing gastrointestinal surgery. Defining the standard method for microbiota assessment in these patients could set up the future approach and clinical practice.

1. Introduction

The gut microbiota (GM) consists of a complex ecosystem that is responsible for our health and characterizes us in a unique way, like a fingerprint [1]. Through metabolites and complex molecular pathways, GM interacts with multiple organs and diseases within a multidirectional network [2]. It is therefore to be expected that surgery on the digestive system may somehow disrupt the balance of the intestinal barrier with short- and long-term consequences. On the other hand, many surgically treated diseases are originally characterized by dysbiosis. It thus remains to be understood which is the cause and which the effect [3].
Certainly, advanced techniques for studying the microbiota, which have begun to spread in recent years, will open up new perspectives on dysbiosis factors predictive of surgical complications and poorer prognosis. Even in the surgical world, a role for the microbiome as an organ is now accepted, and its potential to be saved pre- and post-operatively is understood. The intent of this descriptive review is to summarize the latest evidence and research trends in the relationship between microbiota and digestive surgery.

2. Methods

We selected articles discussing the association of gastrointestinal surgery and microbiota. In particular, we chose articles published in the last twenty years, focusing on the latest scientific evidence. The studies were mostly European and American, with a minority performed on the eastern population. We developed a non-systematic review article using the following electronic sources: PubMed, EMBASE, Google Scholar, Ovid, MEDLINE, Scopus, Cochrane controlled trials register, and Web of Science. We used the following single combined search terms: “Gastrointestinal Surgery AND Gut microbiota”, “GI Surgery AND microbiota”, “Post-surgery AND Gut microbiome”, “Post-surgical infections AND microbiota”, “Bariatric Surgery AND microbiota”. We examined all the articles reporting humans’ related data (inclusion criteria) excluding works with not available full text, not in English language, book chapters, abstracts, and articles published before 1990 (exclusion criteria). Finally, we evaluated supplementary references among articles evaluated in the first search round.

3. Preoperative Management for Elective Colorectal Surgery: The Importance of Preserving the “Good” Resident Microbiota

Evidence from recent years shows that preparing the gut for surgery can affect the health of the microbiota, and thus the long-term prognosis. There has been a shift from the idea of sterile surgery to the realization that the “good” microbiota must be preserved. The issue is to avoid all the potential pathogens as much as possible.
Adoption of purgative cleansing, oral and intravenous antibiotics and application of topical antiseptic solutions is part of the routine practice usually adopted nowadays. The scientific community is now understanding the possible effects on gut microbiota. The administration of osmotic agents, such as polyethylene glycol (PEG) which is a metabolically inert isotonic laxative, is the most diffuse way to obtain adequate cleansing in the absence of presumed relevant consequences and side effects. However, there is still debate on the effective advantages of this practice, and moreover, preserving gut fecal contents seems to lead to better outcomes [4]. Mechanical bowel preparation impacts the complex intestinal barrier by interacting with endoluminal commensals and epithelial components. A recent meta-analysis conducted on eight studies and 1065 patients showed that bowel preparation does not lower the risk of colorectal anastomotic leakages [5]. Beyond the evidence of microbiota variation during diagnostic and operative endoscopy cleaning, important data are emerging about the consequences for those undergoing elective colon-rectal surgery. A recent Cochrane metanalysis showed that there is no statistically significant evidence that patients benefit from mechanical bowel preparation, nor the use of rectal enemas. In colonic surgery, bowel cleansing can be safely omitted and induces no lower complication rate [6]. In 2010, Watanabe et al. first published a study based on a Japanese population evaluating the impact of bowel preparation in this specific setting [7]. They reported decreased populations of Bifidobacteria, Clostridium coccoides, Clostridium leptum, Enterobacteriaceae, and Lactobacillus postoperatively, with no change in Enterococcus and Staphylococcus populations. This alteration with reduced levels of short-chain fatty acids (SCFAs) could result in impairment of the intestinal barrier, thus leading to bacterial translocation and possible infectious complications. Similar results were shown in another Chinese RCT with significant reduction in the whole bacterial abundance, especially regarding Bacteroides and Peptostreptococcus, and bacillus/coccus ratio [8]. Further data were analyzed considering the days required for bowel preparation. Sixty patients undergoing colorectal resection were enrolled in Nanfang Hospital from March 2010 to March 2011 and were randomly assigned to receive 3 days vs. 1 day of bowel preparation. They observed postoperative changes in both groups; the postoperative population of Bifidobacterium and Lactobacillus showed a significant decrease, which was more significant in the group undergoing fast 1-day preparation, while E. coli and Staphylococcus were higher than the preoperative level, which was more significant in the 3-day preparation. Significant results emerged in terms of postoperative infections with lower incidence in case of fast preparation [9].
The adoption of antibiotic prophylaxis is another cornerstone of the pre-operative phase that could change from a microbiota preservation perspective. The lack of a targeting approach to pathogens still makes it a debated point despite the evidence in preventing major infections.
The linkage between antibiotic administration and changes in gut microbiota diversity and consequent antibiotic-associated diarrhea is a concept learned long ago [10]. The mechanisms of interaction of antibiotic topical or systemic therapy preoperatively are not yet well understood and could lead to a prevalence of microbes responsible for short- and long-term post-operative consequences; surprisingly, it appears that antibiotic therapy does not lead to a significant risk of C. difficile colitis [11].
It is clear that particular attention was given to the possible combined effect of intestinal mechanical preparation and antibiotic prophylaxis. Kiran et al. [12] conducted a comparative study in 2015 for 8442 patients undergoing elective colorectal resection, evaluating the impact of preoperative osmotic preparation and antibiotics, osmotic preparation alone and no bowel preparation in terms of outcomes, particularly anastomotic leak, surgical site infection (SSI) and ileus. On multivariable analysis, mechanical bowel preparation with antibiotics, but not without, was independently associated with reduced anastomotic leak, surgical site infections and postoperative ileus. These data were in accordance with subsequent results from a large metanalysis [13]. However, the more recent MOBILE trial challenged the relative evidence that the combination of osmotic preparation and antibiotic together lead to better outcomes. They included 396 patients undergoing elective colectomy, randomly assigned to combined preparation (196 patients) and no bowel preparation (200), and no significant reduction of surgical site infections and overall morbidity after combined preparation [14]. This raised the question of assessing the benefits by combining the two factors, and the ORALEV study recently confirmed that the administration of oral antibiotics as prophylaxis the day before colon surgery significantly reduces the incidence of surgical-site infections without mechanical bowel preparation, since mechanical bowel preparation was used in both groups [15].
Interesting results are expected from the ongoing MOBILE2 trial, which compares mechanical and antibiotic bowel preparation versus bowel preparation alone in patients undergoing rectal surgery. The aim of the study is to examine if oral antibiotics reduce the overall complications, SSIs or anastomotic leakages after rectal surgery, and also if there are any adverse effects related to the oral antibiotics [16].
These studies suggest that there is ongoing interest in the best practice to prevent dangerous infections while preserving our “good” commensals, and there is still a long way to go to achieve consensual international recommendations.

4. Which Microbes and Which Molecular Pathways after Gastrointestinal Surgery

The first systematic review assessing pre- and post-surgical microbiome changes after gastrointestinal surgery was published in 2021. They included 14 studies, all reporting post-surgical changes in the microbiome profiles. In 9 of the 14 studies, the prevalence of specific bacteria had significantly changed after surgery. Improved outcome was associated with higher levels of beneficial bacteria and greater microbiome diversity post-surgery [17]. The key recommendations reported by the authors are the development of a standardized protocol for microbiome sampling methods, choice of sample sites within the GIT, analysis methodology and time frames of testing.
Eight studies reported inflammatory bowel disease (IBD) surgical approach and its consequences [18,19,20,21,22,23,24,25,26]. Specific changes reported enhance the idea that surgery can somehow normalize a state of dysbiosis that characterizes these diseases. For Crohn’s disease, great variability is observed in the case of ileocolic anastomosis or ileostomy. Interesting features have been found in patients who develop post-surgical disease recurrence, with higher counts of nitrate-reducing bacteria such as E. coli 3 months after surgery. Recurrence was associated with increased levels of facultative anaerobes and nitrate reducers (Streptococcaceae and Enterobacteriaceae) [22] rather than anaerobic spore-forming bacteria such as C. coccoides, and other potentially beneficial Clostridiales such as F. prausnitzii, which were increased in patients in remission [23,24].
Four studies, two European and two asian [27,28,29,30], assessed microbiome changes following colorectal surgery, giving attention to some key players: the decrease in Firmicutes, Bacteroides, Clostridium leptum (Firmicutes), Bifidobacterium (Actinobacteria), Atopobium cluster (Actinobacteria), Prevotella (Bacteroidetes) and Escherichia Coli (Proteobacteria); conversely, an increase was shown for Enterobacteriaceae (Proteobacteria), Clostridia (Firmicutes) and Gram-negative anaerobes (including Bacteroides, Fusobacteria and Pseudomonas). There were less marked associations with microbial changes compared to the IBD population. In one study [28], surgical site infections within 1 week of surgery were associated with increased numbers of facultative anaerobes and nitrate reducers such as the Proteobacteria Pseudomonas and Enterobacter, Bacteriodes spp and Firmicutes Staphylococcus, Enterococcus and Klebsiella. Another study [31] found a similar trend towards reduced post-surgical infectious complications in the group who had decreased Enterobacteriaceae, Staphylococcus and Pseudomonas and an increase in Bifidobacterium within 2 weeks after surgery.
A more recent study by Kong et al. [32] analyzed the compositional shifts of the intestinal microbiota in fecal samples from 43 CRC patients undergoing radical surgery through 16S rRNA amplicon sequencing preoperatively compared to postoperatively. After CRC surgery, they observed a reduced ratio of Bacteroidetes/Firmicutes; the numbers of beneficial obligate anaerobes, including Bacteroides, Bifidobacterium, Faecalibacterium, Parabacteroides and Prevotella, were also reduced postoperatively. Moreover, radical surgery not only removed the tumor-associated lesions, but also eliminated well-known tumor-associated bacteria including Enterococcus and Fusobacterium, possibly aiding in recurrence prevention. Furthermore, butyrate-producing bacteria (Bacillus, Bilophila, Barnesiella) were decreased, whereas conditional pathogens, including Escherichia-Shigella, Enterobacteriaceae and Streptococcus, were enhanced. These results are contrary to another Chinese study that revealed a decrease in Escherichia-Shigella and an increase in Enterococcus and Parabacteroides [33]. Thus, these perturbations of intestinal microbiota after CRC surgery could promote adverse inflammatory outcomes in these patients. A further systematic review was recently conducted, which included 6 randomized controlled trials and 27 prospective cohort studies, reporting a total of 968 patients. Gastrointestinal surgery was associated with an increase in α diversity and a shift in β diversity postoperatively. Multiple bacterial taxa were identified to consistently trend toward an increase or decrease postoperatively. A difference in microbiota across geographic provenance was also observed. There was a distinct lack of studies showing correlation with clinical outcomes or performing microbiome functional analysis. Furthermore, there was a lack of standardization in sampling, analytical methodology and reporting [34].

4.1. Microbiota Involvement in Anastomotic Leakage and Surgical Site Infections

The process of tissue repair begins immediately after intestinal resection and anastomosis. Some evidence in animal models, such as germ-free mice, showed that the absence of commensals could delay physiological repair processes [35]. AL occurrence was usually linked to poor surgical technique-related factors, including ischemia, increased suture tension, device deployment, suture type or placement and stapling method. However, recent vast improvements in surgical technology did not lead to the expected better outcomes [36].
The available literature supports an important role for intestinal microbiota in anastomotic healing and susceptibility to anastomotic leakage (AL). All this knowledge mainly derives from animal studies conducted in rodents [37,38,39]. Butyrate-producing bacteria assist in epithelial repair, while specific collagenolytic pathogens, such as E. faecalis and P. aeruginosa, have been involved in AL pathophysiology. However, their presence should be added to other factors to develop AL.
A recent study assessed the efficacy of bioresorbable sheath (C-Seal) in preventing colorectal AL in 123 patients undergoing colorectal resection. The C-seal was introduced to help reduce the risk of anastomotic leakage. It comprises an intraluminal biodegradable soft sheath that is stapled to the colorectal anastomosis [40,41]. They collected mucosa-associated microbiota from the stapled colorectal “donut” and performed a 16s MiSeq sequencing. Results showed that in non-C-seal population AL was related to reduced microbial diversity and increased abundance of Bacteroidaceae and Lachnospiraceae compared to patients without AL. AL was not associated with intestinal microbiota in C-seal patients who presented higher AL rates. Although C-seal placement did not reduce AL occurrence, it obliterated the association between intestinal microbiota and AL [42,43].
A more recent study compared preoperative microbiota composition in patients eligible for CRC surgery. They showed that patients developing AL had increased dysbiosis-related bacteria, as Acinetobacter Iwo and Hafina alvei; low abundance of protective bacteria Barnesiella intestihominis and Faecalibacterium prausnitzii were reported as well [44]. A similar preoperative assessment was conducted by Liu Y et al. [45].
Based on these data, there is an emerging concept on the possibility of preoperative microbiota screening predictive for AL development; furthermore, a preoperative modulation could be applied to prevent AL and surgical site infections (SSIs).
In addition to AL, the problem of SSIs is emerging as a possible correlate of dysbiosis status. The infectious agents involved have long been reported, but only recently has microbiota analysis been proposed to interpret these data. Ohigashi et al. [28] is currently one of the first studies reporting fecal microbiota with SSI-related bacteria in CRC patients after surgery. SSIs occurred in 6 out of 81 patients, and the causative bacteria were identified to be S. aureus, P. aeruginosa and Enterococcus spp., which were also enriched in the analysis of fecal samples after CRC surgery. The MIRACLe protocol was proposed to evaluate the effects of a novel perioperative treatment for the implementation of the gut microbiota, to prevent anastomotic fistula and leakage (AL) in patients undergoing laparoscopic colorectal resections for cancer. This study was conducted in a single Italian center, including 60 patients undergoing elective colorectal surgery and receiving a novel perioperative preparation following the MIRACLe (Microbiota Implementation to Reduce Anastomotic Colorectal Leaks) protocol (oral antibiotics, mechanical bowel preparation and perioperative probiotics), compared to a group of 500 patients (control group), who received a standard ERAS protocol. In the MIRACLe Group, only one anastomotic leak was registered. The MIRACLe group showed encouraging results, with significant lower incidence of AL, surgical site infections, reoperations and post-operative mortality [46]. Another protocol has been recently proposed to assess whether a presurgical nutritional intervention, based on a high-fiber diet rich in polyunsaturated fatty acids (PUFAs), can reduce disturbances of the gut microbiota composition and, consequently, the rate of post-surgical complications in patients with CRC. This is expected to be the first study protocol evaluating the impact of a presurgical nutritional intervention based on high-fiber intake with high levels of PUFAs on altered gut microbiota and its relationship with post-surgical complications, anastomotic leaks and site infections. The ongoing METABIOTE trial is an observational prospective cohort study which aims to define the predictive value of prognostic markers, including gut microbiota, sarcopenia, metabolic syndrome and obesity. All consecutive patients with a non-metastatic sporadic CRC scheduled for surgery at the Digestive Surgery department at the University Hospital of Clermont-Ferrand (France) are systematically asked to participate in the study, aiming at inclusion of 300 patients from 2019 to 2021. The primary outcome is the 5-year overall survival (OS). Other outcomes are 5-year CRC-related OS, 5-year disease-free survival, 30-day postoperative morbidity, 90-day postoperative mortality and length of hospital stay [47].

4.2. Long-Term Microbiota Modifications after Gastrointestinal Surgery

On the basis of the above, a change in the microbiota affects the immediate post-operative phases, but it is expected that it may also persist in the long term. In this respect, it is the oncology field that has been most studied, in order to identify a prognostically favorable microbial profile.
The first bacterium studied in CRC patients in relation to oncological outcomes is Fusobacterium nucleatum (F. nucleatum), which is known for its role in tumorigenesis. Bacteroides fragilis (B. fragilis) was then correlated as well with reduced disease-free and overall survival, especially enterotoxigenic B. fragilis (ETBF) was related to CRC progression [48]. Another bacterial genus of particular interest concerning CRC outcomes is Bifidobacterium, with its abundance having been reduced in CRC patients compared with healthy individuals, as reported by a cohort study of 1313 CRC patients. They reported the detection of Bifidobacterium in 30% of tumors with no statistically significant association between its abundance and survival outcomes or molecular and clinicopathological features of CRC. [49]
After individual key players, the concept of co-abundance groups (CAGs) was introduced. Flemer et al. [50] provided an analysis based on CAGs of various bacteria, and they reported that Bacteroidetes, Prevotella and pathogen CAGs were correlated with longer survival in CRC patients.
A Chinese group [51] was the first to evaluate the long-term metabolic status and microbial composition of CRC patients after curative surgery. They showed that metabolic syndrome was more prevalent among CRC patients after right hemicolectomy (RH) but not after lower anterior resection (LAR) compared to controls over a follow-up period of 5 years. The RH group also showed dysbiosis represented by lower bacterial richness and diversity as opposed to the LAR group. The RH group also showed microbiota alterations postoperatively with a reduction in the Firmicures/Bacteroidetes ratio, with a higher level of Fusobacterium and lower numbers of butyrate-producing Faecalibacterium prausnitzii and Roseburia. Abbas et al. [52] recently published an observational cohort study evaluating gut microbiota changes before and after elective oncologic colon surgery in adults previously receiving different antibiotic prophylaxis. They performed metataxonomic analysis based on sequencing of the bacterial 16S rRNA gene marker. Despite the small population of 27 patients, they observed large and significant increases in the genus Enterococcus between the preoperative/intraoperative samples and the postoperative sample, mostly represented by Enterococcus faecalis. There were also significant differences in the postoperative microbiome between patients who received standard prophylaxis and carbapenems, specifically in the family Erysipelotrichaceae.
Similar evidence was suggested by the Malaysian population studied by Png et al. [53]. They evaluated forty-nine fecal samples from 25 noncancer (NC) individuals and 12 CRC patients, before and 6 months after surgery, performing analysis through bacterial 16S rRNA gene sequencing. Bacterial richness and diversity were reduced, while pro-carcinogenic bacteria such as Bacteroides fragilis and Odoribacter splanchnicus were increased in CRC patients compared to the NC group. These differences were no longer observed after surgery. The comparison between the preoperative and postoperative CRC population confirmed increased abundance of probiotic bacteria after surgery. Concomitantly, bacteria associated with CRC progression were observed to have increased after surgery, implying persistent dysbiosis. The increasing use of advanced latest-generation analysis techniques will lead to further analysis of the microbial tissue characterizing this population in the long term, providing new insights.

5. Bariatric Surgery

5.1. Microbiota and Obesity

In order to consider the impact of bariatric surgery on the microbiota, it is necessary to re-evaluate the microbial balance during obesity.
Obesity is a chronic disease that recognizes a multifactorial etiology: environmental, dietary, lifestyle, host and genetic factors contribute to its establishment [54].
In the current understanding of this disease, gut microbiota plays an accepted role in the development of obesity; in fact, it is considered, by definition, a state of dysbiosis [55].
In obesity, a reduction in microbial genetic richness and compositional and functional alterations has been observed. Dysbiosis contributes to a state of low-grade inflammation, increased body weight and fat mass, and increased risk of type 2 diabetes [56].
The microbiota of individuals with obesity is believed to have an increased capacity for energy harvest [57].
Indeed, the gut microbiota produces short-chain fatty acids; these are bacterial metabolites derived from the fermentation of otherwise indigestible oligosaccharides, dietary plant fibers and undigested proteins. Short-chain fatty acids are involved in an important part of the pathophysiology of obesity, namely stimulation of the production of satiety hormones (GLP-1 and PYY), as well as playing a role in lipid metabolism, inflammation and insulin sensitivity.
In addition, specific microbial profiles have been observed in obesity. As widely accepted, obesity is commonly associated with an increased ratio of Firmicutes to Bacteroidetes, a dysbiotic energy-harvesting microbiome [58].
The first treatment of overweight and obesity is usually clinical management, traditionally based on lifestyle interventions [59]. Weight loss through dietary interventions alters the composition of the gut microbiota, counteracting the high Firmicutes/Bacteroidetes ratio widely reported in obesity and increasing the benefits of the phylum Verrucomicrobia [60], but also promoting functional changes in the microbiota and altering its derived metabolites [61,62]. When nonsurgical alternatives have failed, bariatric surgery (BS) is considered the gold standard treatment, with great performance in remediating the condition in the short and long term [63].

5.2. Evidence after Surgery

In the last few decades, BS has increased dramatically worldwide and now appears to be not only a treatment for weight loss but a solution to reduce cardiovascular risks and diabetes, thus leading to this surgical procedure being considered a “metabolic surgery”.
First, bariatric surgery has a profound effect on biochemical and anthropometric parameters, improving the health status of patients in the years following surgery [64].
The known mechanisms by which bariatric surgery succeeds in achieving its therapeutic effects are: altered diet (significantly energy restricted with increased protein intake), changes in the anatomy and physiology of the gastrointestinal tract (GIT) induced by the type of procedure performed (thus, with specific changes in food digestion and absorption) and lack of adequate nutritional supplementation [65].
A link between GM change and improved body composition after surgery-induced weight loss is evidenced by animal and human fecal transplants from BS patients to mice reared without any exposure to the microorganisms. In these studies, after transplantation and over three months, mice who received microbiota from BS patients gained less fat mass than mice colonized with microbial communities from obese patients [56].
Studies using different sequencing methods have reported elevated microbial gene richness (MGR) and diversity of gut microbial populations after Roux-en-Y gastric bypass (RYGB) and vertical sleeve gastrectomy (VSG) and a shift from “obese” to “less obese” microbial structure [66].
One of the most interesting studies in this area, using high-resolution sequencing technology, has shown that bariatric surgery, including both adjustable gastric banding (AGB) and RYGB, improves MGR. In most patients it is partially restored, and most maintain low MGR (even for longer periods, such as 5 years) while losing a lot of weight. [64].
At the phylum level, bariatric surgery appears to reduce the abundance of the phylum Firmicutes, while Proteobacteria showed an opposite pattern [67].
This change in the composition and richness of the gut microbiota after surgery appears to be more profound in Roux-en-Y gastric bypass (RYGB) than in sleeve gastrectomy [68,69]. In other studies, changes in gut microbiota among patients undergoing these two types of interventions appear to be similar [70,71]. A recent study, analyzing the fecal metagenome and metabolome of patients with severe obesity after BS, demonstrated long-term improvement in GM even after a longer period (up to 4 years). [72]
Having ascertained the actual change in GM after BS and having hypothesized a possible role in improving body composition, the main point to be understood is whether there are interventions that can be used in clinical practice before BS that can enhance this change. Several studies have found that the use of probiotics has the potential to reduce SIBO and improve gastrointestinal symptoms after bariatric surgery [73,74]. Another recent study showed that L. acidophilus and B. lactis supplementation is effective in reducing swelling but without affecting the development of SIBO in the early postoperative period [75]. The use of prebiotics before BS could also improve metabolic effects, such as increased postprandial secretion of insulin, GLP-1, and PYY, which usually increases after surgery. This might be a starting point to investigate, with new studies, whether the use of prebiotics in the late postoperative period might be more effective in patients with a weak response to insulin and incretins, and thus insufficient weight loss or diabetes [76].
Another interesting aspect is the possible effect of gut microbiota on the nervous system and cognitive physiology. Evidence from animals and humans implies that the gut microbiota affects brain structure and cognitive functions [77]. A recent longitudinal study highlights the crucial role of the gut microbiota–mycobiota in human somatic and cognitive health, particularly memory, suggesting that the complex ecology of the gut–brain axis evolves dynamically to adapt body and brain physiology in response to BS [78].

6. Summary

The gut microbiota acts as an ecosystem with a very important role in the well-being of the entire organism. It has also been shown that the microbiota has a great influence on the healing process of an intestinal anastomosis. There is a strict relationship between GM and the surgical approach considering that surgical intervention is a curative trauma for this ecosystem. The goal of the future research should be evaluating the “good” microbes improving surgical outcomes. At the same time, pathogens are still the mainstay of mortality in this setting and should not be underestimated. Further large studies will highlight the possible microbiota profiles predictive of digestive surgery outcomes, and everyday practice will be influenced by this rapidly growing evidence. Using the increasingly available and inexpensive methods of personalized microbiota analysis, we can advance the understanding of the intestinal microbiota in health and disease and ultimately guide clinical care. In addition, new studies are needed to determine whether the use of pharmacological interventions that modify the gut microbiota can improve some post-surgical outcomes by increasing the prevalence of protective versus pathogenic microbial species.

Author Contributions

Conceptualization and writing—original draft preparation, G.G., C.B., L.C., M.S. and C.C.; writing—review and editing, C.P., C.F.M.J., A.C. and D.C.; supervision, G.E., V.S. and C.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.A.; Hirschfield, G.M.; Hold, G.L.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2015, 65, 330–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Cani, P.D.; Knauf, C. How gut microbes talk to organs: The role of endocrine and nervous routes. Mol. Metab. 2016, 5, 743–752. [Google Scholar] [CrossRef] [PubMed]
  3. Bachmann, R.; Leonard, D.; Delzenne, N.; Kartheuser, A.; Cani, P.D. Novel insight into the role of microbiota in colorectal surgery. Gut 2017, 66, 738–749. [Google Scholar] [CrossRef] [PubMed]
  4. Gustafsson, U.; Scott, M.; Schwenk, W.; Demartines, N.; Roulin, D.; Francis, N.; McNaught, C.; MacFie, J.; Liberman, A.; Soop, M.; et al. Guidelines for perioperative care in elective colonic surgery: Enhanced Recovery After Surgery (ERAS®) Society recommendations. Clin. Nutr. 2012, 31, 783–800. [Google Scholar] [CrossRef]
  5. 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]
  6. Leenen, J.P.L.; Hentzen, J.E.K.R.; Ockhuijsen, H.D.L. Effectiveness of mechanical bowel preparation versus no preparation on anastomotic leakage in colorectal surgery: A systematic review and meta-analysis. Updates Surg. 2018, 71, 227–236. [Google Scholar] [CrossRef]
  7. Watanabe, M.; Murakami, M.; Nakao, K.; Asahara, T.; Nomoto, K.; Tsunoda, A. Randomized clinical trial of the influence of mechanical bowel preparation on faecal microflora in patients undergoing colonic cancer resection. Br. J. Surg. 2010, 97, 1791–1797. [Google Scholar] [CrossRef]
  8. Wang, S.; Wang, M.L.; Li, Y.; Zhou, Y.B.; Wang, D.S. Clinical study on risk factor associated with gut flora change in patients with rectal cancer during perioperative period. Zhonghua Wei Chang Wai Ke Za Zhi 2012, 15, 570–573. [Google Scholar]
  9. Wu, Y.J.; Wu, C.T.; Zhang, X.B.; Ou, W.T.; Huang, P. Clinical study of different bowel preparations on changes of gut flora in patients undergoing colorectal resection. Zhonghua Wei Chang Wai Ke Za Zhi 2012, 15, 574–577. [Google Scholar]
  10. Young, V.B.; Schmidt, T.M. Antibiotic-Associated Diarrhea Accompanied by Large-Scale Alterations in the Composition of the Fecal Microbiota. J. Clin. Microbiol. 2004, 42, 1203–1206. [Google Scholar] [CrossRef] [Green Version]
  11. Yeom, C.H.; Cho, M.M.; Baek, S.K.; Bae, O.S. Risk Factors for the Development of Clostridium difficile-associated Colitis after Colorectal Cancer Surgery. J. Korean Soc. Coloproctol. 2010, 26, 329–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Kiran, R.P.; Murray, A.C.A.; Chiuzan, C.; Estrada, D.; Forde, K. Combined Preoperative Mechanical Bowel Preparation with Oral Antibiotics Significantly Reduces Surgical Site Infection, Anastomotic Leak, and Ileus after Colorectal Surgery. Ann. Surg. 2015, 262, 416–425. [Google Scholar] [CrossRef]
  13. Rollins, K.; Javanmard-Emamghissi, H.; Acheson, A.G.; Lobo, D.N. The Role of Oral Antibiotic Preparation in Elective Colorectal Surgery: A Meta-analysis. Ann. Surg. 2019, 270, 43–58. [Google Scholar] [CrossRef] [PubMed]
  14. Koskenvuo, L.; Lehtonen, T.; Koskensalo, S.; Rasilainen, S.; Klintrup, K.; Ehrlich, A.; Pinta, T.; Scheinin, T.; Sallinen, V. Mechanical and oral antibiotic bowel preparation versus no bowel preparation for elective colectomy (MOBILE): A multicentre, randomised, parallel, single-blinded trial. Lancet 2019, 394, 840–848. [Google Scholar] [CrossRef]
  15. Basany, E.E.; Solís-Peña, A.; Pellino, G.; Kreisler, E.; Fraccalvieri, D.; Muinelo-Lorenzo, M.; Maseda-Díaz, O.; García-González, J.M.; Santamaría-Olabarrieta, M.; Codina-Cazador, A.; et al. Preoperative oral antibiotics and surgical-site infections in colon surgery (ORALEV): A multicentre, single-blind, pragmatic, randomised controlled trial. Lancet Gastroenterol. Hepatol. 2020, 5, 729–738. [Google Scholar] [CrossRef]
  16. 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]
  17. Ferrie, S.; Webster, A.; Wu, B.; Tan, C.; Carey, S. Gastrointestinal surgery and the gut microbiome: A systematic literature review. Eur. J. Clin. Nutr. 2020, 75, 12–25. [Google Scholar] [CrossRef]
  18. Almeida, M.G.; Kiss, D.R.; Zilberstein, B.; Quintanilha, A.G.; Teixeira, M.G.; Habr-Gama, A. Intestinal Mucosa-Associated Microflora in Ulcerative Colitis Patients Before and After Restorative Proctocolectomy with an Ileoanal Pouch. Dis. Colon Rectum 2008, 51, 1113–1119. [Google Scholar] [CrossRef]
  19. Neut, C.; Bulois, P.; Desreumaux, P.; Membré, J.M.; Lederman, E.; Gambiez, L.; Cortot, A.; Quandalle, P.; van Kruiningen, H.; Colombel, J.-F. Changes in the bacterial flora of the neoterminal ileum after ileocolonic resection for Crohn’s disease. Am. J. Gastroenterol. 2002, 97, 939–946. [Google Scholar] [CrossRef]
  20. Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humaran, L.G.; Gratadoux, J.-J.; Blugeon, S.; Bridonneau, C.; Furet, J.-P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [Green Version]
  21. Marteau, P.; Lémann, M.; Seksik, P.; Laharie, D.; Colombel, J.F.; Bouhnik, Y.; Cadiot, G.; Soulé, J.C.; Bourreille, A.; Metman, E.; et al. Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn’s disease: A randomised, double blind, placebo controlled GETAID trial. Gut 2006, 55, 842–847. [Google Scholar] [CrossRef] [PubMed]
  22. De Cruz, P.; Kang, S.; Wagner, J.; Buckley, M.; Sim, W.H.; Prideaux, L.; Lockett, T.; McSweeney, C.; Morrison, M.; Kirkwood, C.D.; et al. Association between specific mucosa-associated microbiota in Crohn’s disease at the time of resection and subsequent disease recurrence: A pilot study. J. Gastroenterol. Hepatol. 2015, 30, 268–278. [Google Scholar] [CrossRef]
  23. Dey, N.; Soergel, D.A.W.; Repo, S.; Brenner, S.E. Association of gut microbiota with post-operative clinical course in Crohn’s disease. BMC Gastroenterol. 2013, 13, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Laffin, M.; Perry, T.; Park, H.; Gillevet, P.M.; Sikaroodi, M.; Kaplan, G.; Fedorak, R.N.; Kroeker, K.; Dieleman, L.A.; Dicken, B.; et al. Endospore-Forming Bacteria are Associated with Maintenance of Remission following Intestinal Resection in Crohn’s Disease. Gastroenterology 2017, 152, S192–S193. [Google Scholar] [CrossRef]
  25. Wright, E.K.; Kamm, M.A.; Wagner, J.; Teo, S.-M.; De Cruz, P.; Hamilton, A.L.; Ritchie, K.J.; Inouye, M.; Kirkwood, C.D. Microbial Factors Associated with Postoperative Crohn’s Disease Recurrence. J. Crohn’s Colitis 2016, 11, 191–203. [Google Scholar] [CrossRef] [Green Version]
  26. Mondot, S.; Lepage, P.; Seksik, P.; Allez, M.; Treton, X.; Bouhnik, Y.; Colombel, J.F.; Leclerc, M.; Pochart, P.; Dore, J.; et al. Structural robustness of the gut mucosal microbiota is associated with Crohn’s disease remission after surgery. Gut 2015, 65, 954–962. [Google Scholar] [CrossRef]
  27. Feng, X.; Su, Y.; Jiang, J.; Li, N.; Ding, W.; Wang, Z.; Hu, X.; Zhu, W.; Li, J. Changes in Fecal and Colonic Mucosal Microbiota of Patients with Refractory Constipation after a Subtotal Colectomy. Am. Surg. 2015, 81, 198–206. [Google Scholar] [CrossRef]
  28. Ohigashi, S.; Sudo, K.; Kobayashi, D.; Takahashi, T.; Nomoto, K.; Onodera, H. Significant Changes in the Intestinal Environment After Surgery in Patients with Colorectal Cancer. J. Gastrointest. Surg. 2013, 17, 1657–1664. [Google Scholar] [CrossRef]
  29. Mangell, P.; Thorlacius, H.; Syk, I.; Ahrné, S.; Molin, G.; Olsson, C.; Jeppsson, B. Lactobacillus plantarum 299v Does Not Reduce Enteric Bacteria or Bacterial Translocation in Patients Undergoing Colon Resection. Am. J. Dig. Dis. 2012, 57, 1915–1924. [Google Scholar] [CrossRef]
  30. Quintanilha, A.G.; dos Santos, M.A.; Avila-Campos, M.J.; Saad, W.A.; Pinotti, H.W.; Zilberstein, B. Chagasic Megacolon and Proximal Jejunum Microbiota. Scand. J. Gastroenterol. 2000, 35, 632–636. [Google Scholar] [CrossRef]
  31. Ucmak, H.; Sucakli, M.H.; Buyukbese, M.A.; Toprak, R.; kuş, S. Infections of gastrointestinal laparoscopic surgery: Systematic review. J. Med. Res. Sci. 2012. [Google Scholar]
  32. Kong, C.; Gao, R.; Qin, H. Abstract 2833: Alterations in intestinal microbiota of colorectal cancer patients receiving radical surgery combined with adjuvant CapeOx therapy. Sci. China Life Sci. 2019, 62, 1178–1193. [Google Scholar] [CrossRef] [PubMed]
  33. Liu, C.; Zhang, Y.; Shang, Y.; Wu, B.; Yang, E.; Luo, Y.-Y.; Li, X. Intestinal bacteria detected in cancer and adjacent tissue from patients with colorectal cancer. Oncol. Lett. 2018, 17, 1115–1127. [Google Scholar] [CrossRef]
  34. Tarazi, M.; Jamel, S.; Mullish, B.H.; Markar, S.R.; Hanna, G.B. Impact of gastrointestinal surgery upon the gut microbiome: A systematic review. Surgery 2021, 171, 1331–1340. [Google Scholar] [CrossRef]
  35. Hooper, L.V.; Wong, M.H.; Thelin, A.; Hansson, L.; Falk, P.G.; Gordon, J.I. Molecular Analysis of Commensal Host-Microbial Relationships in the Intestine. Science 2001, 291, 881–884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Daams, F.; Luyer, M.; Lange, J.F. Colorectal anastomotic leakage: Aspects of prevention, detection and treatment. World J. Gastroenterol. 2013, 19, 2293–2297. [Google Scholar] [CrossRef]
  37. Shogan, B.D.; Smith, D.P.; Christley, S.; Gilbert, J.A.; Zaborina, O.; Alverdy, J.C. Intestinal anastomotic injury alters spatially defined microbiome composition and function. Microbiome 2014, 2, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Stern, J.R.; Olivas, A.D.; Valuckaite, V.; Zaborina, O.; Alverdy, J.C.; An, G. Agent-based model of epithelial host–pathogen interactions in anastomotic leak. J. Surg. Res. 2013, 184, 730–738. [Google Scholar] [CrossRef] [Green Version]
  39. Shogan, B.D.; Belogortseva, N.; Luong, P.M.; Zaborin, A.; Lax, S.; Bethel, C.; Ward, M.; Muldoon, J.P.; Singer, M.; An, G.; et al. Collagen degradation and MMP9 activation by Enterococcus faecalis contribute to intestinal anastomotic leak. Sci. Transl. Med. 2015, 7, 286ra68. [Google Scholar] [CrossRef] [Green Version]
  40. Morks, A.N.; Havenga, K.; Ten Cate Hoedemaker, H.O.; Ploeg, R.J. C-seal for prevention of anastomotic leakage following colorectal anastomosis. Ned. Tijdschr. Geneeskd. 2011, 155, 2812. [Google Scholar]
  41. Bakker, I.S.; Morks, A.N.; Ten Cate Hoedemaker, H.O.; Burgerhof, J.G.M.; Leuvenink, H.; van Praagh, J.B.; Ploeg, R.J.; Havenga, K.; van Etten, B.; Lange, J.F.M.; et al. Randomized clinical trial of biodegradeable intraluminal sheath to prevent anastomotic leak after stapled colorectal anastomosis. Br. J. Surg. 2017, 104, 1010–1019. [Google Scholar] [CrossRef] [PubMed]
  42. Van Praagh, J.B.; de Goffau, M.C.; Bakker, I.S.; Harmsen, H.J.; Olinga, P.; Havenga, K. Intestinal microbiota and anastomotic leakage of stapled colorectal anastomoses: A pilot study. Surg. Endosc. 2016, 30, 2259–2265. [Google Scholar] [CrossRef]
  43. Van Praagh, J.B.; de Goffau, M.; Bakker, I.S.; van Goor, H.; Harmsen, H.J.M.; Olinga, P.; Havenga, K. Mucus Microbiome of Anastomotic Tissue During Surgery Has Predictive Value for Colorectal Anastomotic Leakage. Ann. Surg. 2019, 269, 911–916. [Google Scholar] [CrossRef]
  44. Palmisano, S.; Campisciano, G.; Iacuzzo, C.; Bonadio, L.; Zucca, A.; Cosola, D.; Comar, M.; de Manzini, N. Role of preoperative gut microbiota on colorectal anastomotic leakage: Preliminary results. Updates Surg. 2020, 72, 1013–1022. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, Y.; He, W.; Yang, J.; He, Y.; Wang, Z.; Li, K. The effects of preoperative intestinal dysbacteriosis on postoperative recovery in colorectal cancer surgery: A prospective cohort study. BMC Gastroenterol. 2021, 21, 446. [Google Scholar] [CrossRef] [PubMed]
  46. Carlini, M.; Grieco, M.; Spoletini, D.; Menditto, R.; Napoleone, V.; Brachini, G.; Mingoli, A.; Marcellinaro, R. Implementation of the gut microbiota prevents anastomotic leaks in laparoscopic colorectal surgery for cancer:the results of the MIRACLe study. Updates Surg. 2022, 74, 1253–1262. [Google Scholar] [CrossRef]
  47. Veziant, J.; Poirot, K.; Chevarin, C.; Cassagnes, L.; Sauvanet, P.; Chassaing, B.; Robin, F.; Godfraind, C.; Barnich, N.; Pezet, D.; et al. Predictive Value of Innovative Prognostic Markers (Gut Microbiota, Sarcopenia, Metabolic Syndrome and Obesity) on Surgical and Oncologic Results in the Management of Sporadic Colorectal Adenocarcinoma. BMJ Open 2020, 10. [Google Scholar] [CrossRef] [Green Version]
  48. Wei, Z.; Cao, S.; Liu, S.; Yao, Z.; Sun, T.; Li, Y.; Li, J.; Zhang, D.; Zhou, Y. Could gut microbiota serve as prognostic biomarker associated with colorectal cancer patients’ survival? A pilot study on relevant mechanism. Oncotarget 2016, 7, 46158–46172. [Google Scholar] [CrossRef] [Green Version]
  49. Kosumi, K.; Hamada, T.; Koh, H.; Borowsky, J.; Bullman, S.; Twombly, T.S.; Nevo, D.; Masugi, Y.; Liu, L.; da Silva, A.; et al. The Amount of Bifidobacterium Genus in Colorectal Carcinoma Tissue in Relation to Tumor Characteristics and Clinical Outcome. Am. J. Pathol. 2018, 188, 2839–2852. [Google Scholar] [CrossRef] [Green Version]
  50. Flemer, B.; Herlihy, M.; O’Riordain, M.; Shanahan, F.; O’Toole, P.W. Tumour-associated and non-tumour-associated microbiota: Addendum. Gut Microbes 2018, 9, 369–373. [Google Scholar] [CrossRef]
  51. Lin, X.-H.; Jiang, J.-K.; Luo, J.-C.; Lin, C.-C.; Ting, P.-H.; Yang, U.-C.; Lan, Y.-T.; Huang, Y.-H.; Hou, M.-C.; Lee, F.-Y. The long term microbiota and metabolic status in patients with colorectal cancer after curative colon surgery. PLoS ONE 2019, 14, e0218436. [Google Scholar] [CrossRef] [PubMed]
  52. Abbas, M.; Gaïa, N.; Buchs, N.C.; Delaune, V.; Girard, M.; Andrey, D.O.; Meyer, J.; Schrenzel, J.; Ris, F.; Harbarth, S.; et al. Changes in the gut bacterial communities in colon cancer surgery patients: An observational study. Gut Pathog. 2022, 14, 2. [Google Scholar] [CrossRef] [PubMed]
  53. Png, C.-W.; Chua, Y.-K.; Law, J.-H.; Zhang, Y.; Tan, K.-K. Alterations in co-abundant bacteriome in colorectal cancer and its persistence after surgery: A pilot study. Sci. Rep. 2022, 12, 9829. [Google Scholar] [CrossRef] [PubMed]
  54. Khan, M.J.; Gerasimidis, K.; Edwards, C.A.; Shaikh, M.G. Role of Gut Microbiota in the Aetiology of Obesity: Proposed Mechanisms and Review of the Literature. J. Obes. 2016, 2016, 7353642. [Google Scholar] [CrossRef]
  55. Cuevas-Sierra, A.; Ramos-Lopez, O.; Riezu-Boj, J.I.; Milagro, F.I.; Martinez, J.A. Diet, Gut Microbiota, and Obesity: Links with Host Genetics and Epigenetics and Potential Applications. Adv. Nutr. 2019, 10, S17–S30. [Google Scholar] [CrossRef] [Green Version]
  56. Debédat, J.; Clément, K.; Aron-Wisnewsky, J. Gut Microbiota Dysbiosis in Human Obesity: Impact of Bariatric Surgery. Curr. Obes. Rep. 2019, 8, 229–242. [Google Scholar] [CrossRef]
  57. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An Obesity-Associated Gut Microbiome with Increased Capacity for Energy Harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  58. Ley, R.E.; Bäckhed, F.; Turnbaugh, P.; Lozupone, C.A.; Knight, R.D.; Gordon, J.I. Obesity alters gut microbial ecology. Proc. Natl. Acad. Sci. USA 2005, 102, 11070–11075. [Google Scholar] [CrossRef] [Green Version]
  59. Semlitsch, T.; Stigler, F.L.; Jeitler, K.; Horvath, K.; Siebenhofer-Kroitzsch, A. Management of overweight and obesity in primary care—A systematic overview of international evidence-based guidelines. Obes. Rev. 2019, 20, 1218–1230. [Google Scholar] [CrossRef]
  60. Rinninella, E.; Cintoni, M.; Raoul, P.; Ianiro, G.; Laterza, L.; Lopetuso, L.R.; Ponziani, F.R.; Gasbarrini, A.; Mele, M.C. Gut Microbiota during Dietary Restrictions: New Insights in Non-Communicable Diseases. Microorganisms 2020, 8, 1140. [Google Scholar] [CrossRef]
  61. Fabbiano, S.; Suárez-Zamorano, N.; Chevalier, C.; Lazarević, V.; Kieser, S.; Rigo, D.; Leo, S.; Veyrat-Durebex, C.; Gaïa, N.; Maresca, M.; et al. Functional Gut Microbiota Remodeling Contributes to the Caloric Restriction-Induced Metabolic Improvements. Cell Metab. 2018, 28, 907–921.e7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Gibiino, G.; De Siena, M.; Sbrancia, M.; Binda, C.; Sambri, V.; Gasbarrini, A.; Fabbri, C. Dietary Habits and Gut Microbiota in Healthy Adults: Focusing on the Right Diet. A Systematic Review. Int. J. Mol. Sci. 2021, 22, 6728. [Google Scholar] [CrossRef] [PubMed]
  63. Park, C.H.; Nam, S.-J.; Choi, H.S.; Kim, K.O.; Kim, D.H.; Kim, J.-W.; Sohn, W.; Yoon, J.H.; Jung, S.H.; Hyun, Y.S.; et al. Comparative Efficacy of Bariatric Surgery in the Treatment of Morbid Obesity and Diabetes Mellitus: A Systematic Review and Network Meta-Analysis. Obes. Surg. 2019, 29, 2180–2190. [Google Scholar] [CrossRef]
  64. Aron-Wisnewsky, J.; Prifti, E.; Belda, E.; Ichou, F.; Kayser, B.D.; Dao, M.C.; Verger, E.O.; Hedjazi, L.; Bouillot, J.-L.; Chevallier, J.-M.; et al. Major microbiota dysbiosis in severe obesity: Fate after bariatric surgery. Gut 2018, 68, 70–82. [Google Scholar] [CrossRef] [PubMed]
  65. Lupoli, R.; Lembo, E.; Saldalamacchia, G.; Avola, C.K.; Angrisani, L.; Capaldo, B. Bariatric surgery and long-term nutritional issues. World J. Diabetes 2017, 8, 464–474. [Google Scholar] [CrossRef]
  66. Magouliotis, D.E.; Tasiopoulou, V.S.; Sioka, E.; Chatedaki, C.; Zacharoulis, D. Impact of Bariatric Surgery on Metabolic and Gut Microbiota Profile: A Systematic Review and Meta-analysis. Obes. Surg. 2017, 27, 1345–1357. [Google Scholar] [CrossRef]
  67. Oduro-Donkor, D.; Turner, M.; Farnaud, S.; Renshaw, D.; Kyrou, I.; Hanson, P.; Hattersley, J.; Weickert, M.O.; Menon, V.; Randeva, H.S.; et al. Modification of fecal microbiota as a mediator of effective weight loss and metabolic benefits following bariatric surgery. Expert Rev. Endocrinol. Metab. 2020, 15, 363–373. [Google Scholar] [CrossRef]
  68. Salazar, N.; Ponce-Alonso, M.; Garriga, M.; Sánchez-Carrillo, S.; Hernández-Barranco, A.M.; Redruello, B.; Fernández, M.; Botella-Carretero, J.I.; Vega-Piñero, B.; Galeano, J.; et al. Fecal Metabolome and Bacterial Composition in Severe Obesity: Impact of Diet and Bariatric Surgery. Gut Microbes 2022, 14, 2106102. [Google Scholar] [CrossRef]
  69. Lee, C.J.; Florea, L.; Sears, C.L.; Maruthur, N.; Potter, J.J.; Schweitzer, M.; Magnuson, T.; Clark, J.M. Changes in Gut Microbiome after Bariatric Surgery Versus Medical Weight Loss in a Pilot Randomized Trial. Obes. Surg. 2019, 29, 3239–3245. [Google Scholar] [CrossRef]
  70. Stefura, T.; Zapała, B.; Gosiewski, T.; Skomarovska, O.; Pędziwiatr, M.; Major, P. Changes in the Composition of Oral and Intestinal Microbiota after Sleeve Gastrectomy and Roux-En-Y Gastric Bypass and Their Impact on Outcomes of Bariatric Surgery. Obes. Surg. 2022, 32, 1439–1450. [Google Scholar] [CrossRef]
  71. Tremaroli, V.; Karlsson, F.; Werling, M.; Ståhlman, M.; Kovatcheva-Datchary, P.; Olbers, T.; Fändriks, L.; le Roux, C.W.; Nielsen, J.; Bäckhed, F. Roux-en-Y Gastric Bypass and Vertical Banded Gastroplasty Induce Long-Term Changes on the Human Gut Microbiome Contributing to Fat Mass Regulation. Cell Metab. 2015, 22, 228–238. [Google Scholar] [CrossRef] [Green Version]
  72. Juárez-Fernández, M.; Román-Sagüillo, S.; Porras, D.; García-Mediavilla, M.; Linares, P.; Ballesteros-Pomar, M.; Urioste-Fondo, A.; Álvarez-Cuenllas, B.; González-Gallego, J.; Sánchez-Campos, S.; et al. Long-Term Effects of Bariatric Surgery on Gut Microbiota Composition and Faecal Metabolome Related to Obesity Remission. Nutrients 2021, 13, 2519. [Google Scholar] [CrossRef] [PubMed]
  73. Woodard, G.A.; Encarnacion, B.; Downey, J.R.; Peraza, J.; Chong, K.; Hernandez-Boussard, T.; Morton, J.M. Probiotics Improve Outcomes After Roux-en-Y Gastric Bypass Surgery: A Prospective Randomized Trial. J. Gastrointest. Surg. 2009, 13, 1198–1204. [Google Scholar] [CrossRef] [PubMed]
  74. Chen, J.C.; Lee, W.J.; Tsou, J.J.; Liu, T.P.; Tsai, P.L. Effect of probiotics on postoperative quality of gastric bypass surgeries: A prospective randomized trial. Surg. Obes. Relat. Dis. 2016, 12, 57–61. [Google Scholar] [CrossRef] [PubMed]
  75. Wagner, N.R.F.; Ramos, M.R.Z.; de Oliveira Carlos, L.; da Cruz, M.R.R.; Taconeli, C.A.; Filho, A.J.B.; Nassif, L.S.; Schieferdecker, M.E.M.; Campos, A.C.L. Effects of Probiotics Supplementation on Gastrointestinal Symptoms and SIBO after Roux-en-Y Gastric Bypass: A Prospective, Randomized, Double-Blind, Placebo-Controlled Trial. Obes. Surg. 2020, 31, 143–150. [Google Scholar] [CrossRef]
  76. Calikoglu, F.; Barbaros, U.; Uzum, A.K.; Tutuncu, Y.; Satman, I. The Metabolic Effects of Pre-probiotic Supplementation after Roux-en-Y Gastric Bypass (RYGB) Surgery: A Prospective, Randomized Controlled Study. Obes. Surg. 2020, 31, 215–223. [Google Scholar] [CrossRef]
  77. Sharon, G.; Sampson, T.R.; Geschwind, D.H.; Mazmanian, S.K. The Central Nervous System and the Gut Microbiome. Cell 2016, 167, 915–932. [Google Scholar] [CrossRef] [Green Version]
  78. Enaud, R.; Cambos, S.; Viaud, E.; Guichoux, E.; Chancerel, E.; Marighetto, A.; Etchamendy, N.; Clark, S.; Mohammedi, K.; Cota, D.; et al. Gut Microbiota and Mycobiota Evolution Is Linked to Memory Improvement after Bariatric Surgery in Obese Patients: A Pilot Study. Nutrients 2021, 13, 4061. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gibiino, G.; Binda, C.; Cristofaro, L.; Sbrancia, M.; Coluccio, C.; Petraroli, C.; Jung, C.F.M.; Cucchetti, A.; Cavaliere, D.; Ercolani, G.; et al. Dysbiosis and Gastrointestinal Surgery: Current Insights and Future Research. Biomedicines 2022, 10, 2532. https://doi.org/10.3390/biomedicines10102532

AMA Style

Gibiino G, Binda C, Cristofaro L, Sbrancia M, Coluccio C, Petraroli C, Jung CFM, Cucchetti A, Cavaliere D, Ercolani G, et al. Dysbiosis and Gastrointestinal Surgery: Current Insights and Future Research. Biomedicines. 2022; 10(10):2532. https://doi.org/10.3390/biomedicines10102532

Chicago/Turabian Style

Gibiino, Giulia, Cecilia Binda, Ludovica Cristofaro, Monica Sbrancia, Chiara Coluccio, Chiara Petraroli, Carlo Felix Maria Jung, Alessandro Cucchetti, Davide Cavaliere, Giorgio Ercolani, and et al. 2022. "Dysbiosis and Gastrointestinal Surgery: Current Insights and Future Research" Biomedicines 10, no. 10: 2532. https://doi.org/10.3390/biomedicines10102532

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