Introduction
Obesity rates have dramatically increased over the last two decades [1]. By 2023, it is anticipated that more than one billion individuals globally will be affected by this trend [2]. The situation is not different in the Arabian Gulf countries such as the United Arab Emirates (UAE), where obesity prevalence is believed to have at least doubled between 1989 and 2017 [3], reaching 24% among males and 40% among females [4].
In addition to diet, lifestyle, and genetics, gut microbiota has surfaced as a potential factor influencing obesity and related metabolic disturbances [5,6]. Proposed mechanisms involve the gut microbiota’s impact on gut peptides, appetite, energy balance, and fat storage [7–12] through a bidirectional signaling axis known as the “microbiota-gut-brain axis” [10,13].
Given the rising prevalence of obesity and the challenges of adhering to lifestyle, dietary, and pharmacotherapy interventions [14], bariatric surgery has gained recognition as an effective long-term adjunct for managing obesity and its associated comorbidities [15,16]. Bariatric surgery can be restrictive or malabsorptive [17]. Sleeve gastrectomy (SG) is a restrictive surgery. The precise mechanisms responsible for weight loss and metabolic alterations associated with SG have not been entirely clarified. Nonetheless, potential factors that may play a role include changes in food preferences, decreased food intake, heightened satiety, improved gastric emptying, reduced nutrient absorption, and increased energy expenditure [18–22]. These mechanisms may, in part, be a result of post-bariatric surgery alterations in gut peptides such as glucagon-like peptide 1 (GLP-1) and peptide YY (PYY) [23–27], and, as mentioned above, these could be influenced by the gut microbiota. Recent findings indicate a substantial modification in the composition and diversity of the gut microbiota after bariatric surgery. Key reported changes encompass increased levels of Proteobacteria [new nomenclature Pseudomonadota] and Bacteroidetes [Bacteroidota] [28–30], reduced levels of Clostridium and the overall Firmicutes [Bacillota] phylum [29,31,32], and variations in Bacteroides and Prevotella based on the consumed diet [33–36]. The composition and diversity of the gut microbiota are thought to play a significant role in weight regulation [37]. Specifically, a higher Firmicutes to Bacteroidetes ratio (F/B), referring to the relative abundance or proportion of the two major phyla of bacteria in the human gut microbiota, has been linked to obesity [38], although there is no consensus on the use of this ratio [39,40]. Ethnicity greatly influences microbiota composition and diversity [41,42], making it crucial to conduct ethnic-specific research. In this context, we embarked on a prospective study to explore the effect of SG on energy homeostasis, gut peptides, hormones, and gut microbiota in a sample of Emiratis with obesity. We aim to understand the unique mechanisms relevant to weight loss following SG, beyond the restrictive effect of the surgery on food intake. Through an in-depth exploration of these mechanisms, we aspire to lay the groundwork for formulating less invasive anti-obesogenic treatment strategies.
Materials and methods
As illustrated in the supporting information (S1 Flow Chart).
Study design
The study design involved a three-month prospective, pre-post investigation.
We recruited Emiratis with obesity undergoing SG from two hospitals in the UAE, one in Sharjah and one in Dubai, between October 27, 2019, and March 26, 2021.
The study was registered in ClinicalTrials.gov (ID: NCT04200521)
Sample size calculation
The sample size was determined by the F/B ratio, which is the principal consequence of this research. By employing traditional calculations based on normal distributions and the findings of Damms-Machado et al., in which the mean (SD) fecal F/B changed significantly from 5.9 (2.1) to 10.4 (1.4) in the three months after bariatric surgery [29], it was determined that a sample size of two patients would be sufficient to achieve 80% power at a two-sided 5% significance level to identify an effect of comparable magnitude. Considering the anticipated high dropout rate, the minimal sample size was multiplied by 15. Thus, we targeted thirty adult Emiratis with obesity of both sexes and resided in the UAE intending to undergo SG. A total of 43 eligible participants were recruited; however, only 19 of them underwent SG and were subsequently included in the study.
Informed consent
Information for participants.
We provided each participant with an information sheet that detailed the purpose, duration, procedures, and protocols involved, potential risks and benefits, and their rights as participants, including the right to withdraw at any time without penalty.
Consent process.
The consent process involved training the research staff, explaining the study in detail, answering any questions, and ensuring that participants fully understood the information provided. We conducted this interaction in the participants’ preferred language to ensure their comprehension.
Documentation.
Participants who agreed to take part in the study signed a written informed consent form. We securely stored these forms to ensure confidentiality and adhere to ethical standards.
Ethical considerations
The Dubai Health Care Regulatory Research Ethics Committee (DHCR-REC), the Research Ethics Committee of the Ministry of Health and Prevention of the UAE (MOHP/DXB-REC-52/2018), and the Zayed University Ethical Committee Board (ZU19_51_F) granted ethical approvals for the study. Before enrollment, written informed consent was obtained from all patients. In studies involving human subjects, every procedure was conducted in adherence to the ethical principles set forth by the relevant national and institutional research committees, in addition to the 1964 Helsinki Declaration.
Inclusivity in global research
Additional information regarding the ethical, cultural, and scientific considerations specific to inclusivity in global research is included in the Supporting Information (S1 Checklist).
Inclusion criteria
Patients determined to undergo sleeve gastrectomy (SG) by the multidisciplinary team at the partner hospitals were evaluated for their eligibility for the study by the research team. Inclusion criteria entailed Emirati participants residing in the UAE, with stable body weight for the past three months and free of antibiotics for at least three months, aged between 18 to 60 years, of either sex, with a body mass index (BMI) of ≥ 35 kg/m2, and consenting to participate in the study.
Exclusion criteria
The study excluded individuals who were either pregnant or nursing during that period.
Additionally, we excluded individuals who had used antibiotics within the previous three months due to potential alterations in gut microbiota composition that could influence study results. We also excluded participants who experienced significant weight loss of 5% or more in the three months before surgery, as this could potentially impact baseline metabolic parameters.
Moreover, individuals with excessive alcohol consumption, defined as exceeding two standard drinks per day for males and one standard drink per day for females, were excluded from the study [43].
Study variables
General demographics and medical history.
The subsequent information was gathered: age, sex, comorbidities (including hypertension, dyslipidemia, diabetes mellitus type 2 (T2DM), and cardiovascular disease), surgical history within the previous five years, weight loss of at least 5% over the previous three months, present medication usage (including antibiotic consumption), prebiotic and probiotic supplement consumption, nutritional supplement intake, and level of physical activity.
We analyzed physical activity levels using two questions from the initial screening questionnaire. “Do you integrate physical activity into your daily routine?” and “Do you incorporate any exercise program?” required a “yes” or “no” response. Sedentary was defined as “no” to both questions, light physical activity as “yes” to the first question, moderate physical activity as “yes” to the second question, and high physical activity as “yes” to both.
Nutritional assessment.
Anthropometrics: The participant’s body weight was determined to the nearest kilogram using a portable Seca 762 scale (Vogel & Halke, Hamburg, Germany) while fasting and wearing lightweight apparel, and removing their footwear. Height was determined to the nearest millimeter using a portable Seca stadiometer while they were shoeless. BMI was calculated as weight (kg)/ height (m2). Using measuring tapes (Seca 201- Ergonomic circumference measuring tape), the waist circumference (WC) was determined at the midpoint, halfway between the right iliac crest and the lower costal region, with an accuracy of 0.1 cm [44]. The WC was then, classified and categorized as appropriate or increased based on the cut-off points of < 94 cm and ≥ 94 cm for males and < 80 and ≥ 80 cm for females, respectively [45]. Waist-to-height ratio (WHtR) was determined by dividing WC by height in centimeters; abdominal obesity was defined as a WHtR value of 0.5; a WHtR value below 0.5 was regarded as normal [46].
Body composition: Percentage body fat (PBF), fat mass (FM), and fat-free mass (FFM) were evaluated using a bioelectrical impedance analyzer (BC-420 MA, Tanita Corporation, Tokyo, Japan), following the manufacturer’s instructions. We defined elevated PBF among males and females using cut-offs of ≥ 25% and ≥ 35%, respectively [47].
Resting energy expenditure (REE): Resting energy expenditure was assessed using a portable indirect calorimeter (Metabolic Analyser - Indirect Calorimetry; Breezing, Halberstadt, Germany). We conducted this measurement under resting conditions, requiring participants to refrain from eating or drinking for a minimum of 2 hours before the analysis.
Before each measurement, the breezing device was calibrated according to the manufacturer’s instructions. Calibration involved setting up the device to accurately measure oxygen (VO2) and carbon dioxide (VCO2) levels. Participants were instructed to wear a mouthpiece provided with the calorimeter. A nose clip was utilized to ensure that participants breathed exclusively through the mouth, minimizing the risk of air leakage and optimizing measurement accuracy. Participants stayed in a comfortable position to minimize movement. The device continuously records gas exchange data during a designated period of normal breathing (3 minutes). Finally, the collected data is analyzed by the device’s software to determine participants’ REE by analyzing the oxygen and carbon dioxide content of breath. This process provides a reliable assessment of their resting metabolic rate [48].
Food intake: Was assessed using three 24-hour dietary recalls, during which the patient described and quantified the foods and beverages over the past 24 hours for three consecutive days, including two weekdays and one weekend day. During the interview, the dietitian probed the participants more than once to cover forgotten eating occasions and foods and assisted the participant in estimating portion size to provide comprehensive information about the intake. Daily energy (Kcal) and fiber intakes (g/1000 Kcal) were calculated using the food composition database of the Nutritics software (using the Middle Eastern version of their software program).
Appetite: The assessment was conducted using the Simplified Nutritional Appetite Questionnaire (SNAQ) [49]. SNAQ is a quick and easy-to-administer self-assessment screening tool composed of four items (1, 2, 4, and 6) of the Council on Nutrition Appetite Questionnaire (CNAQ). These items assess appetite, satiety, the taste of food, and the number of meals per day, whereby each question has five letter answer options ranging from a to e, which respectively stand for a = 1; b = 2; c = 3; d = 4, and e = 5. The sum of these items constitutes the SNAQ score, which ranges from 4 to 20, with higher scores indicating better appetite and a lower risk of malnutrition. A cut-off of fourteen indicates a risk of malnutrition and predicts a significant risk of at least 5% weight loss within six months.
We translated the questionnaire into Arabic according to international procedures [50].
Two independent translators first translated it both forward and backward, compared the two versions, and made necessary adjustments to the Arabic version.
Postoperative bariatric diet meal planning (S1 File): Participants start with clear liquids (Stage 1) and advance to full liquids (Stage 2) within several days post-surgery, ensuring at least 60 ounces (1,800 cc) of fluid intake daily to avoid dehydration. They then progress through pureed foods (Stage 3), soft foods (Stage 4), and finally all solids (Stage 5) throughout 4 to 6 weeks, with typical daily caloric intake increasing from 400 kcal/day initially to 1,200–1,500 kcal/day several months post-surgery [51].
Biochemical evaluation
Gut peptides (total GLP-1, GLP-2, PYY, CCK, GHR) and hormones (insulin and leptin).
A total of 3 vacutainers of ethylenediaminetetraacetic acid (EDTA) tubes of venous blood samples were collected after a 12-hour overnight fast by a registered nurse/phlebotomist using a 22-gauge needle. We immediately stored the collected blood in EDTA-coated tubes on ice and centrifuged it at 3000 rpm for 15 minutes at a refrigerated temperature. Then, plasma was collected and stored at −20 ºC for later analysis at the Research Institute for Medical and Health Sciences (RIMHS), Sharjah University. Total GLP-1, GLP-2, PYY, GHR, insulin, and leptin were measured using sandwich-based enzyme-linked immunosorbent assay (ELISA) kits (Diametra Millipore, St. Louis, Missouri, USA). We measured CCK using a competitive ELISA kit from ABclonal Technology (Wuhan, China), adhering to the manufacturer’s instructions. The minimum detectable concentrations of these assays are 1.5 pm; 0.3 ng/ml; 6.5 pg/mL; 50 pg/mL; 2 µ U/mL; 0.2 ng/mL, 4.93 pg/ml for GLP-1, GLP-2, PYY, GHR, insulin, leptin, and CCK, respectively.
The measurement of fasting plasma glucose (FPG) levels was conducted following the manufacturer’s instructions using a portable Lux Metre Blood Test (Biochemical Systems International, S.p.A.; Arezzo, Italy). In addition, the Homeostatic Model Assessment of Insulin Resistance (HOMA-IR) was computed by multiplying fasting serum insulin (mU/mL) by FPG (mg/dL) and dividing the result by 405 [52]. HOMA-IR values exceeding 1.8 were regarded as suggestive of insulin resistance [53].
Gut microbiota
Stool sample collection.
Stool samples were self-collected into Zymo (DNA/RNA) Shield™ fecal collection tubes and stored at room temperature without refrigeration. From each participant, two samples were obtained: one was subjected to analysis, and the other one was retained to verify accuracy. Collection of samples adhered to established protocols. To collect 1 gram of feces, a spoon was affixed to the cap of each collection tube, which was pre-filled with 9 ml of DNA/RNA Shield™. We collected sixty-two stool samples from the 43 eligible subjects three days pre-surgery and from the 19 included participants three months post-operatively. The analysis included paired samples from the nineteen participants who underwent SG.
DNA isolation and sequencing of amplicons of the V3–V4 region of the 16S rRNA gene.
The DNA isolation and sequencing of the barcoded amplicons of the V3-V4 region of the 16S rRNA gene were performed by the established protocols supplied by Illumina (Illumina, Eindhoven, The Netherlands) [54]. The steps taken were as follows: the sequencing procedure was executed utilizing the Illumina MiSeq system (San Diego, CA, USA), employing barcodes, and the 2x300 bp protocol. The software Quantitative Insights Into Microbial Ecology (QIIME 2) (https://qiime2.org) software [55] was utilized to analyze the raw sequences. The latter were classified utilizing the Silva database (version 132) as the reference database for the 16S rRNA gene.
We filtered the reads and took only those included in at least 20% of the samples. We use the old nomenclature for phyla, providing new names in brackets in the text for proper comparison with previous literature.
Diversity analysis.
α Diversity indices: The following diversity indices (Observed Operational Taxonomic Units (OTUs), Chao1 index, Phylogenetic diversity (Faith’s PD), Pielou’s evenness, and Shannon diversity index) were calculated with QIIME 2 and displayed in R Studio using the R software (4.1.x) (R Core Team, http://www.R-project.org): We created boxplots using the ggplot2 package for R. We set the depth of rarefication at 3800.
β Diversity indices: Principal Coordinate Analysis (PCoA) was performed using QIIME2 software to visualize dissimilarities in β-diversity. The analysis utilized Unweighted UniFrac, Weighted UniFrac, Bray-Curtis dissimilarity, and Jaccard similarity matrices and were converted to 2D-plots in R using the phyloseq package. Eigenvalues were computed to determine the variance explained by each principal coordinate.
We used Permutational Multivariate Analysis of Variance (PERMANOVA) [56] to assess the statistically significant differences in β diversity between the two groups.
When there were more than two categories, we used the non-parametric Kruskal-Wallis method to calculate the p-value in pairwise comparisons for the diversity between each category.
The non-parametric Wilcoxon signed-rank test for paired samples corrected with the Benjamini–Hochberg false discovery rate (FDR) for multiple comparisons was used in R to analyze differences in microbiota in participants pre- and post-surgery (at phylum and genus levels). The composition of the two groups was visualized using the Krona tool (https://github.com/marbl/Krona/wiki/KronaTools). We calculated the F/B ratio by dividing the relative abundances of Firmicutes and Bacteroidetes.
We investigated the microbial taxa with differential abundances between pre- and post-surgery using linear discriminant analysis (LDA) effect size (LEfSe) (version 1.0 http://huttenhower.sph.harvard.edu/galaxy/). Taxa in each group were deemed enriched if their absolute log10 LDA scores were greater than 2.
A Spearman correlation analysis, for non-parametric data, as is usual for microbiota analyses, was carried out between genera and continuous variables (BMI, WC, WHtR, PBF, REE, energy intake, fiber intake/1000 kcal, appetite, insulin, leptin, GLP1, GLP2, PYY, CCK, and GHR) to pinpoint the microorganisms that correlated positively or negatively with surgical results. The Spearman rho ranges between values of | 0.20 | and | 0.39 | ; | 0.4 | and | 0.69 | ; | 0.70 | and | 0.89; | 0.90 | and | 1.00 | denote a weak, moderate, strong, and very strong correlation, respectively [57]. At a cut-off of < 0.05 for q-values (adjusted p-values after the False Discovery Rate (FDR) correction), were considered significantly different. We generated a Spearman correlation analysis heatmap in R software using the corrplot package.
Statistical analysis
The general characteristics of the participants were analyzed utilizing version 21 of the Statistical Package for the Social Sciences software (SPSS Inc. Chicago, IL, USA). The normality of the data was assessed using the Shapiro-Wilk test before selecting the appropriate statistical analyses. The Shapiro-Wilk test evaluates the null hypothesis (H₀) that the data follows a normal distribution. A P-value greater than 0.05 indicates that we fail to reject the null hypothesis, suggesting that the data does not significantly deviate from normality and can be considered normally distributed (weight (W = 0.94; p = 0.32), BMI (W = 0.95; p = 0.44), WC (W = 0.96; p = 0.58), WHtR (W = 0.97; p = 0.84), fat mass (FM) (W = 0.9; p = 0.06), fat-free mass (FFM) (W = 0.92; p = 0.15), PBF (W = 0.96; p = 0.61), REE (W = 0.95; p = 0.48), EI (W = 0.95; p = 0.44), Fiber intake (W = 0.94; p = 0.29), Fiber/1000Kcal (W = 0.94; p = 0.27), appetite score (W = 0.94; p = 0.32), HOMA-IR (W = 0.92; p = 0.14), GLP1 (W = 0.95; p = 0.54), GLP2 (W = 0.94; p = 0.3), CCK (W = 0.92; p = 0.15), leptin (W = 0.92; p = 0.16), insulin (W = 0.95; p = 0.52), and GHR (W = 0.94; p = 0.36)). Conversely, a P-value less than 0.05 means we reject the null hypothesis, indicating that the data significantly deviates from normality and should be considered non-normally distributed (FPG (W = 0.54; p < 0.001) and PYY (W = 0.9; p = 0.001)).
Continuous variables are presented as mean ± standard error (SE) for normally distributed data and as median with interquartile range (IQR) for non-normally distributed data. Within-subject changes after bariatric surgery were evaluated by a paired sample T-test for normally distributed continuous variables, the Wilcoxon signed-rank test (NPT) for non- normally distributed data, and the McNemar test for categorical variables. A two-tailed p-value less than 0.05 was deemed to indicate statistical significance.
Results
Effects of bariatric surgery on anthropometrics and plasma levels of gut hormones
The baseline characteristics of the participants are detailed in Table 1 along with the study variables comparing the conditions before and after bariatric surgery.
[Figure omitted. See PDF.]
Ultimately, 19 subjects were included pre- and post-surgery: mean age of 29.78 ± 2.29; included 7 males (36.8%) and 12 females (63.2%). Among the nineteen participants enrolled in the study, n = 1 had hypertension, and n = 1 had dyslipidemia; however, none were diagnosed with T2DM or cardiovascular disease. Regarding surgical history, two participants had undergone cholecystectomy, one had a history of appendectomy, and one had a prior caesarean section. Preoperative medication regimens included one participant being prescribed antihypertensive medication, another receiving antacid therapy, and a third utilizing antihistamine medication. Additionally, one participant was taking multivitamins before surgery. Following surgery, the majority of participants received multivitamins and iron supplements, while one continued antacid therapy, another continued antihypertensive medication, and a third continued antihistamine therapy. Notably, none of the participants reported the use of prebiotics, probiotics, or fiber supplements either before or after the surgical intervention. There were no significant differences in the level of physical activity observed between the pre- and post-surgery groups. Furthermore, no one consumed alcohol, and there was no difference in smoking habits. (S1 Table). Additionally, none of the study participants had postoperative complications. Three months post-surgery, the mean BMI fell from 43 to 35 kg/m2 (t = 20.36; p < 0.001); similarly, waist circumference (t = 9.97; p < 0.001), waist-to-height ratio (t = 9.53; p < 0.001), and PBF (t = 5.44; p < 0.001) showed significant improvements. Appetite reduced significantly post-surgery. Participants reduced their total daily energy intake from (245 4Kcal± 171) pre-surgery to (75 6Kcal± 67.6) (t = 10.93; p < 0.001) at three months post-surgery. We noted no significant differences regarding REE and FPG.
Post-operatively, the HOMA-IR displayed a significant reduction; however, it remained above 1.8. Moreover, fasting levels of insulin (t = 6.83; p < 0.001), leptin (t = 10.71; p < 0.001), GLP-1 (t = 5.7; p < 0.001), GLP-2 (t = 5.07; P < 0.001), and CCK (t = 2.42; P = 0.026) decreased significantly, and GHR (t = −2.97; p = 0.008) and PYY (Z = −3.54; p < 0.001) levels significantly increased.
α- and β-diversity (microbial diversity and richness, microbial dissimilarities)
α-Diversity.
The α-diversity of the pre-and post-surgery samples (Fig 1) was calculated using Kruskall-Wallis for five different metrics, including Shannon diversity (Fig 1A; pre (Mean (SEM) = 6.47 ± 0.14, post = 6.73 ± 0.10; p = 0.21), Chao1 (Fig 1B; pre = 213.2 ± 16.14, post = 241.16 ± 16.83; p = 0.23), Pielou’s evenness (Fig 1C; pre = 0.85 ± 0.009, post = 0.87 ± 0.008; p = 0.31), observed OTUs (Fig 1D; pre = 199.31 ± 14.7, post = 222.05 ± 13.74; p = 0.28) and Faith’s PD (Fig 1E, pre = 13.2 ± 0.71, post = 14.58 ± 0.93; p = 0.28). We noted no statistically significant differences in the five-diversity metrics between pre- and post-surgery.
[Figure omitted. See PDF.]
(A) Shannon; (B) Chao1; (C) Pielou’s evenness; (D) Observed OTUs; (E) Faith’s PD.
β-diversity.
Regardingβ-diversity (Fig 2), no significant differences were observed for Unweighted UniFrac and Bray-Curtis dissimilarity metrics. However, significant differences were identified for Weighted UniFrac and Jaccard metrics between pre-and post-surgery ((Fig 2B; p = 0.012; (Eigenheim-values PC1: 0.80, PC2: 0.22 and Fig 2D; p = 0.006; Eigenheim-values PC1: 0.82, PC2: 0.74, respectively). All p values were calculated with the statistic test PERMANOVA.
[Figure omitted. See PDF.]
The total variance explained by the first two components is displayed on the PC1 and PC2 axes, respectively. Eigenvalues for the first five axes (PC1 to PC5) are provided in S3 Table. Pre-surgery (blue spheres) and post-surgery (red spheres).
Pseudo-F for PERMANOVA for the different analyses were as follows: Unweighted UniFrac: F = 1.960337; Weighted Uni: F = 1.691676; Bray: F = 1.272821 and Jacc: F = 1.314021. Eigenheim values for the first five principal coordinates are detailed in S3 Table.
Relative abundance at the phylum level
Bacteroidetes [Bacteroidota], Firmicutes [Bacillota], and Proteobacteria [Pseudomonadota] dominated the microbiome composition of obese Emirati participants pre- and post-surgery (S1A and S1B Fig). The detailed proportions of the top 7 phyla in the pre-and post-surgery samples are provided in (Table 2). Proteobacteria [Pseudomonadota] and Saccharibacteria [Saccharimonadota] exhibited a statistically significant increase in their proportion post-surgery (Z = −1.9; q = 0.013 and Z = −2.4; q = 0.027, respectively). No changes were noted regarding the relative abundance of Firmicutes and Bacteroidetes nor the F/B ratio between pre- and post-surgery samples.
[Figure omitted. See PDF.]
Relative abundance at the genus level
A total of 28 genera showed differences in abundance between pre- and post-surgery with absolute LDA scores > 2, using LefSe (Fig 3A). In this case, of the twenty-eight genera, the same five genera significantly decreased post-surgery as for the Wilcoxon signed-rank test (Table 3 and Fig 3B–I), while 11 genera that were significantly increased by the Wilcoxon signed-rank test also increased post-surgery as identified by LEfSe.
[Figure omitted. See PDF.]
The whiskers extend to the most extreme data points not considered outliers (1.5 times the interquartile range), and the outliers are plotted individually. (A) LefSe discriminative analysis between pre- and obese post-surgery samples (absolute LDA score > 2). The LDA score (log10) for the more prevalent genera in the pre-surgery is represented on a positive scale (green), and the LDA score for the more prevalent genera in the post-surgery is defined on a negative scale (red). (B) Paired boxplots of Alistipes in pre- and post-surgery samples. (C) Paired boxplots of Lachnospira in pre- and post-surgery samples. (D) Paired boxplots of Saccharibacteria in pre- and post-surgery samples. (E) Paired boxplots of Lachnospiraceae UCG 004 in pre- and post-surgery samples. (F) Relative abundance of Alistipes in pre- and post-surgery samples. Means (straight line) and medians (dotted line) are shown for both groups. (G) Relative abundance of Lachnospira in pre- and post-surgery samples. Means (straight line) and medians (dotted line) are shown for both groups. (H) Relative abundance of Saccharibacteria in pre- and post-surgery samples. Means (straight line) and medians (dotted line) are shown for both groups. (I) Relative abundance of Lachnospiraceae UCG 004 in pre- and post-surgery samples. Means (straight line) and medians (dotted line) are shown for both groups.
[Figure omitted. See PDF.]
Changes noted at the level of the following genera were only statistically significant using LEfSe, including (Lachnospiraceae and Lachnospiraceae ND3007 group, Roseburia, Anaerostipes (pre-surgery); Fusobacterium, Intestinimonas, an uncharacterized genus in Christensenellaceae, Ruminiclostridium 9, Odoribacter, Veillonella, Phascolarctobacterium, Akkermansia (post-surgery).
Using the non-parametric Wilcoxon signed-rank test (for paired samples), 19 genera significantly changed between pre-and post-surgery, as depicted in (Table 3 and Fig 3B–I). Of the 19 genera, five decreased post-operatively (Lachnospiraceae UCG-004, Lachnospira, Erysipelotrichaceae UCG-003, Dialister, and Lachnospiraceae FCS020 group), while the remaining 14 increased post-surgery.
Correlations between microbial taxa and clinical parameters
The taxa that show statistically significant correlations with clinical parameters are provided in the S2 Table and S2 Fig, along with the correlation coefficients (rho).
Several taxa that significantly increased post-surgery (uncharacterized genera in Ruminococcaceae, Desulfovibrio, Akkermansia, Carnobacteriaceae, Streptococcus, and Sellimonas) were negatively correlated with energy intake, BMI, and appetite. Roseburia, which decreased post-surgery, was positively correlated with appetite, while PYY was positively correlated with genera that increased post-surgery (Carnobacteriaceae, Streptococcus, Eisenbergiella, Hungatella, Veillonella) and negatively correlated with Dialister that decreased post-surgery. Lachnoclostridium and Flavonifractor showed a negative correlation with leptin. Hungetella and Eisenbergiella, which were significantly higher post-surgery, showed a positive correlation with GHR.
There were no observed correlations among the gut microbiota at the genus level and the subsequent anthropometric and dietary variables (WC, WHtR, PBF) REE, fiber/ 1000 kcal; or biochemical variables (insulin, GLP1, GLP2, and CCK).
Discussion
There is a growing body of evidence linking the gut microbiota to obesity; however, the impact of bariatric surgery on the gut microbiota and the mechanism of action is understudied. Moreover, the integrated communication between the gut microbiota and the metabolic and neuroendocrine systems, along with its connection to obesity, remains to be uncovered. Some of the significant determinants of gut microbiota composition are geography, ethnicity, and cultural practices; thus, gut bacteria composition must be studied in several areas of the globe. We explored the impact of bariatric surgery on the gut microbiota and the factors that influence it, such as energy balance, gut peptides, and hormones, in a sample of Emiratis with obesity. Participants achieved significant weight loss post-surgery, with significant differences in the gut microbiota composition.
Obesity is associated with dysbiosis and less bacterial α-diversity. Post-surgery, our sample experiences an increase in α-diversity without reaching statistical significance. This was consistent with the literature, where patients’ post-surgery, α-diversity, and metabolic profiles improved [58]. However, the published data on the matter is inconsistent. While some research indicated a significant increase [59–63], others found no change or even a decrease [32,42,58,64] post-surgery on gut bacteria. According to Palleja et al. [65], Roux-en-Y gastric bypass leads to an increase in microbial diversity, which remains stable even one year postoperatively. Another comparative study between RYGB and sleeve gastrectomy (SG) also reported greater microbial diversity in the RYGB group, whereas no significant changes were observed in microbial diversity before and after SG in obese individuals [66]. The discrepancies may be the result of many factors, such as the type of surgery conducted [58,66], the sample characteristics at baseline, and the diets followed post-surgery. Therefore, changes in α-diversity appear not to be a reliable indicator of the effectiveness of bariatric surgery in humans [67]. Additionally, the alterations in gut microbiota following RYGB, including specific taxonomic shifts (e.g., greater abundances of Escherichia, Bacteroides, and Veillonella), may play a role in the differing metabolic outcomes between the two procedures [68].
Most literature pinpoints a major change in the microbial community after bariatric surgery, suggesting that the latter significantly impacts the gut microbiota, facilitates weight loss, and ameliorates metabolic profile [59,67]. We observed significant changes in β-diversity post-surgery, denoting a heterogeneous microbiota [69]. However, not all four metrics were significant, with the weighted UniFrac (which considers abundance) being significant, but also Jaccard similarity (which does not take abundance into account). Therefore, we also looked at which taxa were differentially abundant after SG surgery.
We observed significant differences in the relative abundance of several bacterial taxa. In pre-and post-surgery, Bacteroidetes and Firmicutes dominated the microbiome composition, with Proteobacteria the third most phylum, but trailing by an order of magnitude. The other phyla were even less abundant. Post-surgery, Proteobacteria, and Saccharibacteria significantly increased. The rise in Proteobacteria is consistent with earlier findings [30,63,70–72]. Increases in luminal acidity and dissolved oxygen following SG favor the growth of these microbes [30]. A rise in Proteobacteria after bariatric surgery is associated with favorable outcomes, including weight loss [73], enhanced glucose homeostasis, and reduced systemic inflammation [74], even though this phylum is also known to harbor (potential) pathogenic species.
Despite some previous reports that obesity is associated with a higher F/B ratio that decreases after surgery [18,75], F/B remained constant post-surgery in our study. F/B may not be associated with weight loss or predict the outcome post-surgery [39,72,76]. Instead, obesity may be caused by an enhanced energy harvest from one’s diet, not just by a simple rise in F/B ratio but also by gut bacteria tuned towards creating SCFA. Therefore, looking into the connection between energy harvest associated with bacterial SCFA synthesis and obesity is warranted [77]. In line with the literature, we observed significant decreases in several genera, including Lachnospiraceae [78], Dialister, Roseburia, Anaeostipes, and Eryspelotrichaceae [18]. Noteworthy, increased appetite positively correlated with Roseburia in our study. This is in line with previous findings showing that this genus predominated in the microbiota pre-surgery and decreased post-surgery which could explain the decrease in appetite noted post-surgery [18,71]. Furthermore, bariatric surgery induced an increase in the relative abundance of Butyricimonas, which has been linked to decreased appetite [79]. As expected, Akkermansia, which is usually low in obesity, increased post-surgery [72] approximately 6-fold (not significant in the Wilcoxon test, but an LDA of −3.5 in LEfSe). The mucin-degrading properties of Akkermansia reduce inflammation and alterations in adipose tissue, and this bacterium has been strongly linked to favorable metabolic outcomes and a decline in body weight [67,80]. There is a significant correlation between an increase in Akkermansia abundance and a reduction in the taste of energy-dense foods [32]. This is consistent with the hypothesis that Akkermansia prefers an environment with lower food intake, as in our context, where it negatively correlates with energy intake. Akkermansia enrichment is a potential therapy for obesity management. Indeed, a randomized clinical trial showed that short-term oral administration of pasteurized Akkermensia to overweight and obese participants led to weight loss and improved insulin sensitivity [81].
Several studies [18,71] reported increased Alistipes (phylum Bacteroidetes), Veilonella, and Streptococcus post-surgery. We found similar results. Increases in these strains are usually associated with metabolic improvements and more significant weight loss [18,71]. Furthermore, we found a positive correlation between PYY (the satiety hormone) and Veilonella and Streptococcus in our study. Specifically, greater abundances of Veillonella and Streptococcus have been associated with T2DM remission following Roux-en-Y gastric bypass (RYGB) [82].
The gut-brain axis (GBA), which controls hunger and food intake, and the intestinal microbiota consist of bidirectional communication [83]. Some bacterial strains can affect the production of gut hormones, such as PYY, GLP-1, leptin, and GHR, that control hunger and satiety in the brain [83,84] and regulate energy and glucose homeostasis [75,85]. Most evidence refers to increased GLP-1 post-surgery [86]. Interestingly, we observed a decrease in fasting GLP-1, as also shown in some studies [75,87], suggesting that enhanced insulin sensitivity may lead to the downregulation of GLP-1. Methodological differences might explain this discrepancy, i.e., assessing fasting post-prandial GLP-1 response vs. testing a metabolite or the active form of GLP-1. Therefore, it remains challenging to draw firm conclusions about GLP-1 post-SG [87]. Our participants also showed increased plasma PYY post-surgery, as demonstrated elsewhere [24,75,85], partially attributed to weight loss and enhanced satiety. PYY is a neuropeptide that has satiating and reducing effects on body weight and adiposity [88]. Conversely, GHR promotes food intake and attenuates the anorectic effect of PYY and GLP-1 [89]. Gastric cells primarily. Removing part of the stomach may lower GHR, as shown in most SG studies [85]. Yet, we unexpectedly found that GHR increased. A possible explanation is that the majority of GHR-producing cells are located in the stomach’s fundus, whereas circulating acyl GHR may come primarily from the duodenum, which remains intact in SG, leading to a compensatory upregulation of duodenal GHR production [88]. In GHR-deficient mice, SG was similarly effective in inducing weight loss, raising the question of whether this hormone is a crucial facilitator of weight reduction [87], especially since weight loss post-surgery is independent of GHR [90].
The considerable reduction in insulin levels in our study is one of the most significant outcomes of bariatric surgery [91]. Similarly, post-surgery, there was a significant decrease in HOMA-IR levels [92]. Enhanced insulin sensitivity promotes weight loss and decreased appetite [90]. Similarly, leptin resistance improved post-surgery. Leptin stimulates anorexigenic neurons and reduces appetite [84]. As discussed above, changes in the colonic microbiome post-surgery correlate with variations in appetite. Sanmiguel et al. [32] found a link between having higher levels of certain types of colonic bacterial species and losing your appetite after SG. These bacteria included Odoribacter. We report a similar finding, where appetite also dropped significantly post-surgery, associated with an increase in Roseburia and Butyricimonas. Future studies should explore causal processes linking the effects of increasing these bacteria on appetite.
Energy intake, BMI, and appetite negatively correlated with several taxa that significantly increased post-surgery were with. Among them were Streptococcus, Akkermansia, a genus in Carnobacteriaceae, and a genus in Ruminococcaceae. It has been demonstrated that dietary fiber affects hunger, body weight, and gut microbiota composition [93]. Participants consumed more fiber/1000 kcal post-SG, which could play a role. However, they did not meet the recommended daily level. Interestingly, although we detected several significant gut microbiota correlations with clinical parameters, none were reported for resting energy expenditure [94,95].
The question of whether the changes noticed post-bariatric surgery are permanent or reversible remains debatable and potentially surgery-specific [96]. The most extensive modifications in the composition of the gut microbiota are usually present post-RYGB [42]. SG induces less drastic alterations in the diversity of the adaptive gut microbiota than RYGB because it does not alter the gut’s anatomical structure [ 42,79]. Apart from the type of surgical intervention [30,97], the inconsistencies in the impact of bariatric surgery on the gut microbiota are partly due to the differences in participant characteristics, such as geographic location [98], ethnicity, sex, age, initial BMI, initial microbial composition, gastrointestinal comorbidities, medication use, prebiotics/probiotics use, differences in diet, as well as study design, sampling time, sample storage method, sequencing methodology, bioinformatics and statistical software used [42], all of which can change gut microbial composition.
Strengths and limitations
To the best of our knowledge, this is the only study that has examined the effect of bariatric surgery on the following gut peptides (GHR, GLP-1, GLP-2, CCK, PYY) and hormones including insulin, and leptin, on Emiratis with obesity post-SG-surgery. Furthermore, this study’s strengths include a novel research question targeting ethnic-specific research, a clear data-extraction process, well-defined inclusion and exclusion criteria, and a wide range of parameters assessed through valid methods. On the other hand, our results are limited by the small number of patients and the limited follow-up period, although small samples are common in such studies [29,33,97,99]. Furthermore, changes in these subjects’ gut microbiota are evident. According to studies using several follow-up time points, most microbiome alterations occur within three months post-surgery, followed by a steady microbiome for the next year [100]. We did not measure the post-prandial secretion of gut hormones in response to a standardized meal test. Additionally, other gut hormones such as the appetite reducing GIP-oxyntomodulin hybrid peptide were not measured, which could provide further insights into metabolic changes post-surgery [101]. Another limitation is that we did not account for diet, a flaw that is present in the majority, if not all, of human bariatric studies. Diet following bariatric surgery may alter significantly over time, which could impact the gut microbiota and should be investigated. Furthermore, our study did not include other bariatric surgery techniques, such as Roux-en-Y gastric bypass (RYGB), limiting the opportunity to investigate consistency or potential variations between diverse bariatric surgery procedures [34]. Despite the observational study design’s inability to establish causality for changes reported post-surgery, it allows for the formulation of several hypotheses. Another limitation of this study is the potential lack of generalizability of the findings. The study exclusively focused on the Emirati population, which was chosen due to the scarcity of gut microbiota data in the GCC region and the unique ethnic influences that could impact microbiota composition. Although this population provides valuable insights into how gut microbiota and bariatric surgery outcomes interact in a largely understudied region, the results may not be directly applicable to other ethnic or regional populations with different genetic, dietary, and lifestyle factors. Future studies are recommended to explore these associations across diverse ethnic groups and geographic regions to enhance the generalizability of the findings and to determine if region-specific factors or genetic predispositions contribute to the observed effects.
Practical implications
Understanding changes post-bariatric surgery in gut peptides, hormones, and gut microbiota and the interplay between these factors can lead to non-invasive, anti-obesogenic personalized interventions, including targeted probiotics, prebiotics, dietary, and pharmacological interventions. Based on our results, we suggest interventions to reduce Roseburia levels and/or increase Butrycimonas, ultimately influencing appetite. Furthermore, the administration of probiotics rich in Akkermensia to treat obesity appears promising and warrants empirical research.
Conclusion
We discovered that bariatric surgery significantly alters the plasma levels of gut peptides and hormones, as well as the composition of the gut microbiota. We speculate that the microbiota modifications could, at least in part, be responsible for the positive outcomes of bariatric surgery, including weight loss. Most importantly, we could not confirm the association between obesity and an increased F/B ratio [102,103]. Further research is necessary to understand the interplay between the gut microbiota and changes in gut peptides, hormones, human metabolism, and energy homeostasis following bariatric surgery.
Supporting information
S1 Flow Chart. Overview of the study process.
https://doi.org/10.1371/journal.pone.0318699.s001
(DOCX)
S1 Fig. Krona plot of the intestinal bacterial communities pre (A) and post-surgery (B) at the phylum level.
The _ in both plots indicates “uncharacterized.”
https://doi.org/10.1371/journal.pone.0318699.s002
S2 Fig. Heatmap of Spearman correlation analysis between gut microbiota (at genus level) and anthropometrics and clinical variables.
The asterisk (*) denotes statistical significance at a cut-off of < 0.05 for q-values (adjusted p-values after the False Discovery Rate (FDR) correction) and the negative and positive values are the rho ( = correlation factor), which can be positive for positive correlations and negative for negative correlations.
https://doi.org/10.1371/journal.pone.0318699.s003
S1 Table. Lifestyle factors for pre-and post-bariatric surgery.
https://doi.org/10.1371/journal.pone.0318699.s004
(DOCX)
S2 Table. Spearman correlation coefficient (rho) for the taxa showing significant negative and positive correlations with clinical data according to the heatmap (S2 Fig).
https://doi.org/10.1371/journal.pone.0318699.s005
(DOCX)
S3 Table. Eigenvalues of principal coordinates (PCs) for different beta diversity metrics.
https://doi.org/10.1371/journal.pone.0318699.s006
(DOCX)
S1 File. Postoperative bariatric diet meal planning.
https://doi.org/10.1371/journal.pone.0318699.s007
(DOC)
S1 Checklist. Inclusivity in global research.
https://doi.org/10.1371/journal.pone.0318699.s008
(DOCX)
Acknowledgments
We thank the research assistants and students from the College of Natural and Health Sciences department at Zayed University in Dubai campus for their assistance in data collection, Dr. Sulaiman al Habib Hospital (Dubai, UAE), Al Qassimi Hospital (Sharjah, UAE), and the Research Institute for Medical and Health Sciences (Sharjah, UAE) for their collaboration and involvement in the study.
References
1. 1. Hruby A, Hu FB. The epidemiology of obesity: a big picture. PharmacoEcon. 2015;33(7):673–89. pmid:25471927
* View Article
* PubMed/NCBI
* Google Scholar
2. 2. World Obesity Atlas. One Billion People Globally Estimated to be Living with Obesity by 2030: World Obesity Federation; 2022. Available from: https://www.worldobesityday.org/.
* View Article
* Google Scholar
3. 3. Nawar R, Ibrahim E, Abusnana S, Al Awadi F, Al Hammadi FH, Farghaly M, et al. Understanding the Gaps in Obesity Management in the UAE: Perceptions, Barriers, and Attitudes. Dubai Diabetes Endocrinol J. 2021;27(2):37–49.
* View Article
* Google Scholar
4. 4. Sulaiman N, Elbadawi S, Hussein A, Abusnana S, Madani A, Mairghani M, et al. Prevalence of overweight and obesity in United Arab Emirates Expatriates: the UAE national diabetes and lifestyle study. Diabetolo Metabolic Syndrome. 2017;9(1):1–9.
* View Article
* Google Scholar
5. 5. Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr. 2011;94(1):58–65. pmid:21543530
* View Article
* PubMed/NCBI
* Google Scholar
6. 6. Baothman OA, Zamzami MA, Taher I, Abubaker J, Abu-Farha M. The role of Gut Microbiota in the development of obesity and Diabetes. Lipids Health Dis. 2016;15:108. pmid:27317359
* View Article
* PubMed/NCBI
* Google Scholar
7. 7. Karlsson F, Tremaroli V, Nielsen J, Bäckhed F. Assessing the human gut microbiota in metabolic diseases. Diabetes. 2013;62(10):3341–9. pmid:24065795
* View Article
* PubMed/NCBI
* Google Scholar
8. 8. Duca FA, Lam TK. Gut microbiota, nutrient sensing and energy balance. Diabetes Obes Metab. 2014;16(Suppl 1):68–76. pmid:25200299
* View Article
* PubMed/NCBI
* Google Scholar
9. 9. Bakker GJ, Zhao J, Herrema H, Nieuwdorp M. Gut Microbiota and Energy Expenditure in Health and Obesity. J Clin Gastroenterol. 2015;49(Suppl 1):S13–9. pmid:26447957
* View Article
* PubMed/NCBI
* Google Scholar
10. 10. Bauer PV, Hamr SC, Duca FA. Regulation of energy balance by a gut–brain axis and involvement of the gut microbiota. Cell Mol Life Sci. 2016;73(4):737–55.
* View Article
* Google Scholar
11. 11. Covasa M, Stephens RW, Toderean R, Cobuz C. Intestinal sensing by gut microbiota: targeting gut peptides. Front Endocrinol. 2019;10:82. pmid:30837951
* View Article
* PubMed/NCBI
* Google Scholar
12. 12. Blaut M. Gut microbiota and energy balance: role in obesity. Proc Nutr Soc. 2015;74(3):227–34. pmid:25518735
* View Article
* PubMed/NCBI
* Google Scholar
13. 13. Kairupan TS, Amitani H, Cheng KC, Runtuwene J, Asakawa A, Inui A. Role of gastrointestinal hormones in feeding behavior and obesity treatment. J Gastroenterol. 2016;51(2):93–103. pmid:26346735
* View Article
* PubMed/NCBI
* Google Scholar
14. 14. Kral JG, Kava RA, Catalano PM, Moore BJ. Severe obesity: the neglected epidemic. Obesity facts. 2012;5(2):254–69. pmid:22647306
* View Article
* PubMed/NCBI
* Google Scholar
15. 15. Sjöström L, Peltonen M, Jacobson P, Sjöström CD, Karason K, Wedel H, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307(1):56–65. pmid:22215166
* View Article
* PubMed/NCBI
* Google Scholar
16. 16. Mitka M. Bariatric surgery continues to show benefits for patients with diabetes. JAMA. 2012;307(18):1901–2. pmid:22570449
* View Article
* PubMed/NCBI
* Google Scholar
17. 17. Buchwald H, Avidor Y, Braunwald E, Jensen MD, Pories W, Fahrbach K, et al. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004;292(14):1724–37. pmid:15479938
* View Article
* PubMed/NCBI
* Google Scholar
18. 18. Anhê FF, Varin TV, Schertzer JD, Marette A. The Gut Microbiota as a Mediator of Metabolic Benefits after Bariatric Surgery. Can J Diabetes. 2017;41(4):439–47. pmid:28552651
* View Article
* PubMed/NCBI
* Google Scholar
19. 19. Murphy EF, Quigley EM. The microbiota and bariatric surgery: it’s a bug’s life. Gastroenterology. 2012;142(2):399–401; discussion 401. pmid:22192432
* View Article
* PubMed/NCBI
* Google Scholar
20. 20. Miras AD, le Roux CW. Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol. 2013;10(10):575–84. pmid:23835488
* View Article
* PubMed/NCBI
* Google Scholar
21. 21. Shao Y, Ding R, Xu B, Hua R, Shen Q, He K, et al. Alterations of Gut Microbiota After Roux-en-Y Gastric Bypass and Sleeve Gastrectomy in Sprague-Dawley Rats. Obes Surg. 2017;27(2):295–302. pmid:27440168
* View Article
* PubMed/NCBI
* Google Scholar
22. 22. le Roux CW, Bloom SR. Why Do Patients Lose Weight after Roux-en-Y Gastric Bypass? J Clin Endocrinol Metabolism. 2005;90(1):591–2. pmid:15643022
* View Article
* PubMed/NCBI
* Google Scholar
23. 23. Li W, Richard D. Effects of Bariatric Surgery on Energy Homeostasis. Can J Diabetes. 2017;41(4):426–31. pmid:28739098
* View Article
* PubMed/NCBI
* Google Scholar
24. 24. Federico A, Dallio M, Tolone S, Gravina AG, Patrone V, Romano M, et al. Gastrointestinal Hormones, Intestinal Microbiota and Metabolic Homeostasis in Obese Patients: Effect of Bariatric Surgery. In Vivo. 2016;30(3):321–30. pmid:27107092
* View Article
* PubMed/NCBI
* Google Scholar
25. 25. Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides. 2016;77:28–37. pmid:26344355
* View Article
* PubMed/NCBI
* Google Scholar
26. 26. De Lorenzo A, Soldati L, Sarlo F, Calvani M, Di Lorenzo N, Di Renzo L. New obesity classification criteria as a tool for bariatric surgery indication. World J Gastroenterol. 2016;22(2):681–703. pmid:26811617
* View Article
* PubMed/NCBI
* Google Scholar
27. 27. Ravussin Y, Koren O, Spor A, LeDuc C, Gutman R, Stombaugh J, et al. Responses of gut microbiota to diet composition and weight loss in lean and obese mice. Obesity (Silver Spring, Md.). 2012;20(4):738–47. PMID:21593810
* View Article
* PubMed/NCBI
* Google Scholar
28. 28. Kikuchi R, Irie J, Yamada-Goto N, Kikkawa E, Seki Y, Kasama K, et al. The impact of laparoscopic sleeve gastrectomy with duodenojejunal bypass on intestinal microbiota differs from that of laparoscopic sleeve gastrectomy in Japanese patients with obesity. Clin Drug Investig. 2018;38(6):545–52. pmid:29508311
* View Article
* PubMed/NCBI
* Google Scholar
29. 29. Damms-Machado A, Mitra S, Schollenberger AE, Kramer KM, Meile T, Konigsrainer A, et al. Effects of surgical and dietary weight loss therapy for obesity on gut microbiota composition and nutrient absorption. Biomed Res Int. 2015;2015:806248. pmid:25710027
* View Article
* PubMed/NCBI
* Google Scholar
30. 30. Medina DA, Pedreros JP, Turiel D, Quezada N, Pimentel F, Escalona A, et al. Distinct patterns in the gut microbiota after surgical or medical therapy in obese patients. PeerJ. 2017;5:e3443. pmid:28649469
* View Article
* PubMed/NCBI
* Google Scholar
31. 31. Murphy R, Tsai P, Jüllig M, Liu A, Plank L, Booth M. Differential Changes in Gut Microbiota After Gastric Bypass and Sleeve Gastrectomy Bariatric Surgery Vary According to Diabetes Remission. Obes Surg. 2017;27(4):917–25. pmid:27738970
* View Article
* PubMed/NCBI
* Google Scholar
32. 32. Sanmiguel CP, Jacobs J, Gupta A, Ju T, Stains J, Coveleskie K, et al. Surgically induced changes in gut microbiome and hedonic eating as related to weight loss: preliminary findings in obese women undergoing bariatric surgery. Psychosom Med. 2017;79(8):880–7. pmid:28570438
* View Article
* PubMed/NCBI
* Google Scholar
33. 33. Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A. 2009;106(7):2365–70. pmid:19164560
* View Article
* PubMed/NCBI
* Google Scholar
34. 34. Furet JP, Kong LC, Tap J, Poitou C, Basdevant A, Bouillot JL, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59(12):3049–57. pmid:20876719
* View Article
* PubMed/NCBI
* Google Scholar
35. 35. Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–14.
* View Article
* Google Scholar
36. 36. Kong LC, Tap J, Aron-Wisnewsky J, Pelloux V, Basdevant A, Bouillot JL, et al. Gut microbiota after gastric bypass in human obesity: increased richness and associations of bacterial genera with adipose tissue genes. Am J Clin Nutr. 2013;98(1):16–24. pmid:23719559
* View Article
* PubMed/NCBI
* Google Scholar
37. 37. Davis CD. The Gut Microbiome and Its Role in Obesity. Nutr Today. 2016;51(4):167–74. pmid:27795585
* View Article
* PubMed/NCBI
* Google Scholar
38. 38. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Human gut microbes associated with obesity. Nature. 2006;444(7122):1022–3. pmid:17183309
* View Article
* PubMed/NCBI
* Google Scholar
39. 39. Schwiertz A, Taras D, Schafer K, Beijer S, Bos NA, Donus C, et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring). 2010;18(1):190–5. pmid:19498350
* View Article
* PubMed/NCBI
* Google Scholar
40. 40. Guida S, Venema K. Gut microbiota and obesity: Involvement of the adipose tissue. J Funct Foods. 2015;14:407–23.
* View Article
* Google Scholar
41. 41. Deschasaux M, Bouter KE, Prodan A, Levin E, Groen AK, Herrema H, et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat Med. 2018;24(10):1526–31. pmid:30150717
* View Article
* PubMed/NCBI
* Google Scholar
42. 42. Morales-Marroquin E, Hanson B, Greathouse L, de la Cruz-Munoz N, Messiah SE. Comparison of methodological approaches to human gut microbiota changes in response to metabolic and bariatric surgery: A systematic review. Obes Rev. 2020;21(8):e13025. pmid:32249534
* View Article
* PubMed/NCBI
* Google Scholar
43. 43. Snetselaar LG, de Jesus JM, DeSilva DM, Stoody EE. Dietary Guidelines for Americans, 2020–2025: Understanding the Scientific Process, Guidelines, and Key Recommendations. Nutrition Today. 2021;56(6):287–95. pmid:34987271
* View Article
* PubMed/NCBI
* Google Scholar
44. 44. Van der Kooy K, Seidell JC. Techniques for the measurement of visceral fat: a practical guide. Int J Obes Relat Metabolic Disorders. 1993;17(4):187–96pmid:8387967
* View Article
* PubMed/NCBI
* Google Scholar
45. 45. Ford ES. Prevalence of the metabolic syndrome defined by the International Diabetes Federation among adults in the US. Diabetes Care. 2005;28(11):2745–9. pmid:16249550
* View Article
* PubMed/NCBI
* Google Scholar
46. 46. Ashwell M, Gibson S. Waist-to-height ratio as an indicator of ‘early health risk’: simpler and more predictive than using a ‘matrix’based on BMI and waist circumference. BMJ Open. 2016;6(3):e010159. pmid:26975935
* View Article
* PubMed/NCBI
* Google Scholar
47. 47. Romero-Corral A, Somers VK, Sierra-Johnson J, Thomas RJ, Collazo-Clavell M, Korinek J, et al. Accuracy of body mass index in diagnosing obesity in the adult general population. Int J Obes. 2008;32(6):959–66.
* View Article
* Google Scholar
48. 48. Xian X, Quach A, Bridgeman D, Tsow F, Forzani E, Tao N. Personalized indirect calorimeter for energy expenditure (EE) measurement. Global J Obes Diabetes Metabolic Syndrome. 2015;2(1):004–8.
* View Article
* Google Scholar
49. 49. Wilson M-MG, Thomas DR, Rubenstein LZ, Chibnall JT, Anderson S, Baxi A, et al. Appetite assessment: simple appetite questionnaire predicts weight loss in community-dwelling adults and nursing home residents. Am J Clin Nutr. 2005;82(5):1074–81. pmid:16280441
* View Article
* PubMed/NCBI
* Google Scholar
50. 50. World Health Organization. Translation and cross-cultural adaptation of the World Health Organization Disability Assessment Schedule –WHODAS 2.0 2017. Available from: https://www.scielo.br/j/fp/a/6h8GLr8crZcMwtY9f9hfrND/?format=pdf&lang=en.
* View Article
* Google Scholar
51. 51. Mechanick JI, Apovian C, Brethauer S, Garvey WT, Joffe AM, Kim J, et al. Clinical practice guidelines for the perioperative nutrition, metabolic, and nonsurgical support of patients undergoing bariatric procedures - 2019 update: cosponsored by American association of clinical endocrinologists/American college of endocrinology, the obesity society, American society for metabolic & bariatric surgery, obesity medicine association, and American society of anesthesiologists - executive summary. Endocr Pract. 2019;25(12):1346–59. pmid:31682518
* View Article
* PubMed/NCBI
* Google Scholar
52. 52. Wallace TM, Levy JC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care. 2004;27(6):1487–95. pmid:15161807
* View Article
* PubMed/NCBI
* Google Scholar
53. 53. Esteghamati A, Ashraf H, Esteghamati A-R, Meysamie A, Khalilzadeh O, Nakhjavani M, et al. Optimal threshold of homeostasis model assessment for insulin resistance in an Iranian population: the implication of metabolic syndrome to detect insulin resistance. Diabetes Res Clin Pract. 2009;84(3):279–87. pmid:19359063
* View Article
* PubMed/NCBI
* Google Scholar
54. 54. Ahmad MA, Karavetian M, Moubareck CA, Wazz G, Mahdy T, Venema K. Association of the gut microbiota with clinical variables in obese and lean Emirati subjects. Front Microbiol. 2023;14.
* View Article
* Google Scholar
55. 55. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37(8):852–7. pmid:31341288
* View Article
* PubMed/NCBI
* Google Scholar
56. 56. Anderson M. Permutational Multivariate Analysis of Variance (PERMANOVA). 2017;1–15.
57. 57. Schober P, Boer C, Schwarte LA. Correlation Coefficients: Appropriate Use and Interpretation. Anesth Analg. 2018;126(5):1763–8. pmid:29481436
* View Article
* PubMed/NCBI
* Google Scholar
58. 58. Seganfredo F, Blume C, Moehlecke M, Giongo A, Casagrande D, Spolidoro J, et al. Weight‐loss interventions and gut microbiota changes in overweight and obese patients: a systematic review. Obes Rev. 2017;18(8):832–51.
* View Article
* Google Scholar
59. 59. Wang FG, Bai RX, Yan WM, Yan M, Dong LY, Song MM. Differential composition of gut microbiota among healthy volunteers, morbidly obese patients and post-bariatric surgery patients. Exp Ther Med. 2019;17(3):2268–78. pmid:30867711
* View Article
* PubMed/NCBI
* Google Scholar
60. 60. Guo Y, Liu CQ, Shan CX, Chen Y, Li HH, Huang ZP, et al. Gut microbiota after Roux-en-Y gastric bypass and sleeve gastrectomy in a diabetic rat model: Increased diversity and associations of discriminant genera with metabolic changes. Diabetes Metab Res Rev. 2017;33(3).
* View Article
* Google Scholar
61. 61. Basso N, Soricelli E, Castagneto-Gissey L, Casella G, Albanese D, Fava F, et al. Insulin Resistance, Microbiota, and Fat Distribution Changes by a New Model of Vertical Sleeve Gastrectomy in Obese Rats. Diabetes. 2016;65(10):2990–3001. pmid:27431457
* View Article
* PubMed/NCBI
* Google Scholar
62. 62. Campisciano G, Palmisano S, Cason C, Giuricin M, Silvestri M, Guerra M, et al. Gut microbiota characterisation in obese patients before and after bariatric surgery. Benef Microbes. 2018;9(3):367–73. pmid:29482339
* View Article
* PubMed/NCBI
* Google Scholar
63. 63. Guo Y, Huang ZP, Liu CQ, Qi L, Sheng Y, Zou DJ. Modulation of the gut microbiome: a systematic review of the effect of bariatric surgery. Eur J Endocrinol. 2018;178(1):43–56. pmid:28916564
* View Article
* PubMed/NCBI
* Google Scholar
64. 64. Ward EK, Schuster DP, Stowers KH, Royse AK, Ir D, Robertson CE, et al. The effect of PPI use on human gut microbiota and weight loss in patients undergoing laparoscopic Roux-en-Y gastric bypass. Obes Surg. 2014;24(9):1567–71. pmid:24849914
* View Article
* PubMed/NCBI
* Google Scholar
65. 65. Palleja A, Kashani A, Allin KH, Nielsen T, Zhang C, Li Y, et al. Roux-en-Y gastric bypass surgery of morbidly obese patients induces swift and persistent changes of the individual gut microbiota. Genome Med. 2016;8(1):1–13.
* View Article
* Google Scholar
66. 66. Castaner O, Goday A, Park YM, Lee SH, Magkos F, Shiow STE, et al. The Gut Microbiome Profile in Obesity: A Systematic Review. Int J Endocrinol. 2018;2018:4095789. pmid:29849617
* View Article
* PubMed/NCBI
* Google Scholar
67. 67. Cook J, Lehne C, Weiland A, Archid R, Ritze Y, Bauer K, et al. Gut microbiota, probiotics and psychological states and behaviors after bariatric surgery—a systematic review of their interrelation. Nutrients. 2020;12(8):2396. pmid:32785153
* View Article
* PubMed/NCBI
* Google Scholar
68. 68. Hamamah S, Hajnal A, Covasa M. Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets. Nutrients. 2024;16(7):1071. pmid:38613104
* View Article
* PubMed/NCBI
* Google Scholar
69. 69. Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23(7):859–68. pmid:28628112
* View Article
* PubMed/NCBI
* Google Scholar
70. 70. Cortez RV, Petry T, Caravatto P, Pessôa R, Sanabani SS, Martinez MB, et al. Shifts in intestinal microbiota after duodenal exclusion favor glycemic control and weight loss: a randomized controlled trial. Surg Obes Relat Dis. 2018;14(11):1748–54. pmid:30174193
* View Article
* PubMed/NCBI
* Google Scholar
71. 71. Ulker I, Yildiran H. The effects of bariatric surgery on gut microbiota in patients with obesity: a review of the literature. Biosci Microbiota Food Health. 2018;38(1):3–9.
* View Article
* Google Scholar
72. 72. Shen N, Caixàs A, Ahlers M, Patel K, Gao Z, Dutia R, et al. Longitudinal changes of microbiome composition and microbial metabolomics after surgical weight loss in individuals with obesity. Surg Obes Relat Dis. 2019;15(8):1367–73. pmid:31296445
* View Article
* PubMed/NCBI
* Google Scholar
73. 73. Luijten J, Vugts G, Nieuwenhuijzen GAP, Luyer MDP. The Importance of the Microbiome in Bariatric Surgery: a Systematic Review. Obes Surg. 2019;29(7):2338–49.
* View Article
* Google Scholar
74. 74. Debédat J, Clément K, Aron-Wisnewsky J. Gut Microbiota Dysbiosis in Human Obesity: Impact of Bariatric Surgery. Curr Obes Rep. 2019;8(3):229–42. pmid:31197613
* View Article
* PubMed/NCBI
* Google Scholar
75. 75. Sweeney TE, Morton JM. Metabolic surgery: action via hormonal milieu changes, changes in bile acids or gut microbiota? A summary of the literature. Best Pract Res Clin Gastroenterol. 2014;28(4):727–40. pmid:25194186
* View Article
* PubMed/NCBI
* Google Scholar
76. 76. Duncan SH, Lobley G, Holtrop G, Ince J, Johnstone A, Louis P, et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes. 2008;32(11):1720–4.
* View Article
* Google Scholar
77. 77. Jinatham V, Kullawong N, Kespechara K, Gentekaki E, Popluechai S. Comparison of Gut Microbiota between Lean and Obese Adult Thai Individuals. Microbiol Biotechnol Lett. 2018;46(3):277–87.
* View Article
* Google Scholar
78. 78. Patrone V, Vajana E, Minuti A, Callegari ML, Federico A, Loguercio C, et al. Postoperative Changes in Fecal Bacterial Communities and Fermentation Products in Obese Patients Undergoing Bilio-Intestinal Bypass. Front Microbiol. 2016;7:200. pmid:26941724
* View Article
* PubMed/NCBI
* Google Scholar
79. 79. Oduro-Donkor D, Turner MC, Farnaud S, Renshaw D, Kyrou I, Hanson P, 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(5):363–73. pmid:32840125
* View Article
* PubMed/NCBI
* Google Scholar
80. 80. Schneeberger M, Everard A, Gómez-Valadés AG, Matamoros S, Ramírez S, Delzenne NM, et al. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci Rep. 2015;5(1):16643. pmid:26563823
* View Article
* PubMed/NCBI
* Google Scholar
81. 81. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096–103. pmid:31263284
* View Article
* PubMed/NCBI
* Google Scholar
82. 82. Davies N, O’Sullivan JM, Plank LD, Murphy R. Gut Microbial Predictors of Type 2 Diabetes Remission Following Bariatric Surgery. Obes Surg. 2020;30(9):3536–48. pmid:32447634
* View Article
* PubMed/NCBI
* Google Scholar
83. 83. Ballini A, Dipalma G, Isacco CG, Boccellino M, Di Domenico M, Santacroce L, et al. Oral Microbiota and Immune System Crosstalk: A Translational Research. Biology (Basel). 2020;9(6):131.
* View Article
* Google Scholar
84. 84. Han H, Yi B, Zhong R, Wang M, Zhang S, Ma J, et al. From gut microbiota to host appetite: gut microbiota-derived metabolites as key regulators. Microbiome. 2021;9(1):162. pmid:34284827
* View Article
* PubMed/NCBI
* Google Scholar
85. 85. McCarty TR, Jirapinyo P, Thompson CC. Effect of Sleeve Gastrectomy on Ghrelin, GLP-1, PYY, and GIP Gut Hormones: A Systematic Review and Meta-analysis. Ann Surg. 2020;272(1):72–80. pmid:31592891
* View Article
* PubMed/NCBI
* Google Scholar
86. 86. D’Alessio D. Is GLP-1 a hormone: Whether and When? J Diabetes Investig. 2016;7(Suppl 1):50–5. pmid:27186356
* View Article
* PubMed/NCBI
* Google Scholar
87. 87. Farey JE, Preda TC, Fisher OM, Levert-Mignon AJ, Stewart RL, Karsten E, et al. Effect of Laparoscopic Sleeve Gastrectomy on Fasting Gastrointestinal, Pancreatic, and Adipose-Derived Hormones and on Non-Esterified Fatty Acids. Obes Surg. 2017;27(2):399–407. pmid:27465935
* View Article
* PubMed/NCBI
* Google Scholar
88. 88. Yousseif A, Emmanuel J, Karra E, Millet Q, Elkalaawy M, Jenkinson AD, et al. Differential Effects of Laparoscopic Sleeve Gastrectomy and Laparoscopic Gastric Bypass on Appetite, Circulating Acyl-ghrelin, Peptide YY3-36 and Active GLP-1 Levels in Non-diabetic Humans. Obes Surg. 2014;24(2):241–52. pmid:23996294
* View Article
* PubMed/NCBI
* Google Scholar
89. 89. Tsoli M, Chronaiou A, Kehagias I, Kalfarentzos F, Alexandrides TK. Hormone changes and diabetes resolution after biliopancreatic diversion and laparoscopic sleeve gastrectomy: a comparative prospective study. Surg Obes Relat Dis. 2013;9(5):667–77. pmid:23466015
* View Article
* PubMed/NCBI
* Google Scholar
90. 90. Huang R, Ding X, Fu H, Cai Q. Potential mechanisms of sleeve gastrectomy for reducing weight and improving metabolism in patients with obesity. Surg Obes Relat Dis. 2019;15(10):1861–71. pmid:31375442
* View Article
* PubMed/NCBI
* Google Scholar
91. 91. Tabasi M, Ashrafian F, Khezerloo JK, Eshghjoo S, Behrouzi A, Javadinia SA, et al. Changes in Gut Microbiota and Hormones After Bariatric Surgery: a Bench-to-Bedside Review. Obes Surg. 2019;29(5):1663–74. pmid:30793228
* View Article
* PubMed/NCBI
* Google Scholar
92. 92. Magouliotis DE, Tasiopoulou VS, 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(5):1345–57. pmid:28265960
* View Article
* PubMed/NCBI
* Google Scholar
93. 93. Reimer RA, Willis HJ, Tunnicliffe JM, Park H, Madsen KL, Soto-Vaca A. Inulin-type fructans and whey protein both modulate appetite but only fructans alter gut microbiota in adults with overweight/obesity: A randomized controlled trial. Mol Nutr Food Res. 2017;61(11). pmid:28730743
* View Article
* PubMed/NCBI
* Google Scholar
94. 94. Davies Naomi K, O’Sullivan JM, Plank LD, Murphy R. Altered gut microbiome after bariatric surgery and its association with metabolic benefits: A systematic review. Surg Obes Relat Dis. 2019;15(4):656–65.
* View Article
* Google Scholar
95. 95. Kocełak P, Zak-Gołąb A, Zahorska-Markiewicz B, Aptekorz M, Zientara M, Martirosian G, et al. Resting energy expenditure and gut microbiota in obese and normal weight subjects. Eur Rev Med Pharmacol Sci. 2013;17(20):2816–21pmid:24174366
* View Article
* PubMed/NCBI
* Google Scholar
96. 96. Gutiérrez-Repiso C, Moreno-Indias I, Tinahones FJ. Shifts in gut microbiota and their metabolites induced by bariatric surgery. Impact of factors shaping gut microbiota on bariatric surgery outcomes. Rev Endocr Metab Disord. 2021;22(4):1137–56. pmid:34287758
* View Article
* PubMed/NCBI
* Google Scholar
97. 97. Tremaroli V, Karlsson F, Werling M, Stahlman M, Kovatcheva-Datchary P, Olbers T, et al. 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(2):228–38. pmid:26244932
* View Article
* PubMed/NCBI
* Google Scholar
98. 98. Medina DA, Li T, Thomson P, Artacho A, Perez-Brocal V, Moya A. Cross-regional view of functional and taxonomic microbiota composition in obesity and post-obesity treatment shows country specific microbial contribution. Front Microbiol. 2019;10:2346. pmid:31681211
* View Article
* PubMed/NCBI
* Google Scholar
99. 99. Graessler J, Qin Y, Zhong H, Zhang J, Licinio J, Wong ML, et al. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: correlation with inflammatory and metabolic parameters. Pharmacogenomics J. 2013;13(6):514–22. pmid:23032991
* View Article
* PubMed/NCBI
* Google Scholar
100. 100. Yu D, Shu XO, Howard EF, Long J, English WJ, Flynn CR. Fecal metagenomics and metabolomics reveal gut microbial changes after bariatric surgery. Surg Obes Relat Dis. 2020;16(11):1772–82. pmid:32747219
* View Article
* PubMed/NCBI
* Google Scholar
101. 101. Bhat VK, Kerr BD, Flatt PR, Gault VA. A novel GIP-oxyntomodulin hybrid peptide acting through GIP, glucagon and GLP-1 receptors exhibits weight reducing and anti-diabetic properties. Biochem Pharmacol. 2013;85(11):1655–62. pmid:23518155
* View Article
* PubMed/NCBI
* Google Scholar
102. 102. Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3(4):213–23. pmid:18407065
* View Article
* PubMed/NCBI
* Google Scholar
103. 103. le Roux CW, Bueter M, Theis N, Werling M, Ashrafian H, Löwenstein C, et al. Gastric bypass reduces fat intake and preference. Am J Physiol Regul Integr Comp Physiol. 2011;301(4):R1057–66. pmid:21734019
* View Article
* PubMed/NCBI
* Google Scholar
Citation: Ali Ahmad M, Venema K, Ayoub Moubareck C, Wazz G, Mahdy T, Karavetian M (2025) Changes in energy homeostasis, gut peptides, and gut microbiota in Emiratis with obesity after bariatric surgery. PLoS ONE 20(2): e0318699. https://doi.org/10.1371/journal.pone.0318699
About the Authors:
Manal Ali Ahmad
Roles: Conceptualization, Data curation, Formal analysis, Methodology, Writing – original draft
Affiliation: School of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
ORICD: https://orcid.org/0000-0002-9754-1545
Koen Venema
Roles: Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Supervision, Validation, Visualization, Writing – review & editing
Affiliation: Wageningen Food and Biobased Research, Wageningen University and Research, Wageningen, The Netherlands
Carole Ayoub Moubareck
Roles: Funding acquisition
Affiliation: College of Natural and Health Sciences, Zayed University, Dubai, United Arab Emirates
Gabi Wazz
Roles: Writing – review & editing
Affiliation: Center of Excellence in Bariatric and Metabolic Surgery, Dr. Sulaiman Al Habib Hospital, Dubai, United Arab Emirates
Tarek Mahdy
Roles: Writing – review & editing
Affiliation: Department of Bariatric Surgery, Sharjah University, Sharjah, United Arab Emirates
Mirey Karavetian
Roles: Conceptualization, Data curation, Funding acquisition, Methodology, Project administration, Supervision, Writing – review & editing
Affiliation: Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
ORICD: https://orcid.org/0000-0001-5916-6740
[/RAW_REF_TEXT]
1. Hruby A, Hu FB. The epidemiology of obesity: a big picture. PharmacoEcon. 2015;33(7):673–89. pmid:25471927
2. World Obesity Atlas. One Billion People Globally Estimated to be Living with Obesity by 2030: World Obesity Federation; 2022. Available from: https://www.worldobesityday.org/.
3. Nawar R, Ibrahim E, Abusnana S, Al Awadi F, Al Hammadi FH, Farghaly M, et al. Understanding the Gaps in Obesity Management in the UAE: Perceptions, Barriers, and Attitudes. Dubai Diabetes Endocrinol J. 2021;27(2):37–49.
4. Sulaiman N, Elbadawi S, Hussein A, Abusnana S, Madani A, Mairghani M, et al. Prevalence of overweight and obesity in United Arab Emirates Expatriates: the UAE national diabetes and lifestyle study. Diabetolo Metabolic Syndrome. 2017;9(1):1–9.
5. Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr. 2011;94(1):58–65. pmid:21543530
6. Baothman OA, Zamzami MA, Taher I, Abubaker J, Abu-Farha M. The role of Gut Microbiota in the development of obesity and Diabetes. Lipids Health Dis. 2016;15:108. pmid:27317359
7. Karlsson F, Tremaroli V, Nielsen J, Bäckhed F. Assessing the human gut microbiota in metabolic diseases. Diabetes. 2013;62(10):3341–9. pmid:24065795
8. Duca FA, Lam TK. Gut microbiota, nutrient sensing and energy balance. Diabetes Obes Metab. 2014;16(Suppl 1):68–76. pmid:25200299
9. Bakker GJ, Zhao J, Herrema H, Nieuwdorp M. Gut Microbiota and Energy Expenditure in Health and Obesity. J Clin Gastroenterol. 2015;49(Suppl 1):S13–9. pmid:26447957
10. Bauer PV, Hamr SC, Duca FA. Regulation of energy balance by a gut–brain axis and involvement of the gut microbiota. Cell Mol Life Sci. 2016;73(4):737–55.
11. Covasa M, Stephens RW, Toderean R, Cobuz C. Intestinal sensing by gut microbiota: targeting gut peptides. Front Endocrinol. 2019;10:82. pmid:30837951
12. Blaut M. Gut microbiota and energy balance: role in obesity. Proc Nutr Soc. 2015;74(3):227–34. pmid:25518735
13. Kairupan TS, Amitani H, Cheng KC, Runtuwene J, Asakawa A, Inui A. Role of gastrointestinal hormones in feeding behavior and obesity treatment. J Gastroenterol. 2016;51(2):93–103. pmid:26346735
14. Kral JG, Kava RA, Catalano PM, Moore BJ. Severe obesity: the neglected epidemic. Obesity facts. 2012;5(2):254–69. pmid:22647306
15. Sjöström L, Peltonen M, Jacobson P, Sjöström CD, Karason K, Wedel H, et al. Bariatric surgery and long-term cardiovascular events. JAMA. 2012;307(1):56–65. pmid:22215166
16. Mitka M. Bariatric surgery continues to show benefits for patients with diabetes. JAMA. 2012;307(18):1901–2. pmid:22570449
17. Buchwald H, Avidor Y, Braunwald E, Jensen MD, Pories W, Fahrbach K, et al. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004;292(14):1724–37. pmid:15479938
18. Anhê FF, Varin TV, Schertzer JD, Marette A. The Gut Microbiota as a Mediator of Metabolic Benefits after Bariatric Surgery. Can J Diabetes. 2017;41(4):439–47. pmid:28552651
19. Murphy EF, Quigley EM. The microbiota and bariatric surgery: it’s a bug’s life. Gastroenterology. 2012;142(2):399–401; discussion 401. pmid:22192432
20. Miras AD, le Roux CW. Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol. 2013;10(10):575–84. pmid:23835488
21. Shao Y, Ding R, Xu B, Hua R, Shen Q, He K, et al. Alterations of Gut Microbiota After Roux-en-Y Gastric Bypass and Sleeve Gastrectomy in Sprague-Dawley Rats. Obes Surg. 2017;27(2):295–302. pmid:27440168
22. le Roux CW, Bloom SR. Why Do Patients Lose Weight after Roux-en-Y Gastric Bypass? J Clin Endocrinol Metabolism. 2005;90(1):591–2. pmid:15643022
23. Li W, Richard D. Effects of Bariatric Surgery on Energy Homeostasis. Can J Diabetes. 2017;41(4):426–31. pmid:28739098
24. Federico A, Dallio M, Tolone S, Gravina AG, Patrone V, Romano M, et al. Gastrointestinal Hormones, Intestinal Microbiota and Metabolic Homeostasis in Obese Patients: Effect of Bariatric Surgery. In Vivo. 2016;30(3):321–30. pmid:27107092
25. Meek CL, Lewis HB, Reimann F, Gribble FM, Park AJ. The effect of bariatric surgery on gastrointestinal and pancreatic peptide hormones. Peptides. 2016;77:28–37. pmid:26344355
26. De Lorenzo A, Soldati L, Sarlo F, Calvani M, Di Lorenzo N, Di Renzo L. New obesity classification criteria as a tool for bariatric surgery indication. World J Gastroenterol. 2016;22(2):681–703. pmid:26811617
27. Ravussin Y, Koren O, Spor A, LeDuc C, Gutman R, Stombaugh J, et al. Responses of gut microbiota to diet composition and weight loss in lean and obese mice. Obesity (Silver Spring, Md.). 2012;20(4):738–47. PMID:21593810
28. Kikuchi R, Irie J, Yamada-Goto N, Kikkawa E, Seki Y, Kasama K, et al. The impact of laparoscopic sleeve gastrectomy with duodenojejunal bypass on intestinal microbiota differs from that of laparoscopic sleeve gastrectomy in Japanese patients with obesity. Clin Drug Investig. 2018;38(6):545–52. pmid:29508311
29. Damms-Machado A, Mitra S, Schollenberger AE, Kramer KM, Meile T, Konigsrainer A, et al. Effects of surgical and dietary weight loss therapy for obesity on gut microbiota composition and nutrient absorption. Biomed Res Int. 2015;2015:806248. pmid:25710027
30. Medina DA, Pedreros JP, Turiel D, Quezada N, Pimentel F, Escalona A, et al. Distinct patterns in the gut microbiota after surgical or medical therapy in obese patients. PeerJ. 2017;5:e3443. pmid:28649469
31. Murphy R, Tsai P, Jüllig M, Liu A, Plank L, Booth M. Differential Changes in Gut Microbiota After Gastric Bypass and Sleeve Gastrectomy Bariatric Surgery Vary According to Diabetes Remission. Obes Surg. 2017;27(4):917–25. pmid:27738970
32. Sanmiguel CP, Jacobs J, Gupta A, Ju T, Stains J, Coveleskie K, et al. Surgically induced changes in gut microbiome and hedonic eating as related to weight loss: preliminary findings in obese women undergoing bariatric surgery. Psychosom Med. 2017;79(8):880–7. pmid:28570438
33. Zhang H, DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, et al. Human gut microbiota in obesity and after gastric bypass. Proc Natl Acad Sci U S A. 2009;106(7):2365–70. pmid:19164560
34. Furet JP, Kong LC, Tap J, Poitou C, Basdevant A, Bouillot JL, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: links with metabolic and low-grade inflammation markers. Diabetes. 2010;59(12):3049–57. pmid:20876719
35. Huttenhower C, Gevers D, Knight R, Abubucker S, Badger JH, Chinwalla AT, et al. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486(7402):207–14.
36. Kong LC, Tap J, Aron-Wisnewsky J, Pelloux V, Basdevant A, Bouillot JL, et al. Gut microbiota after gastric bypass in human obesity: increased richness and associations of bacterial genera with adipose tissue genes. Am J Clin Nutr. 2013;98(1):16–24. pmid:23719559
37. Davis CD. The Gut Microbiome and Its Role in Obesity. Nutr Today. 2016;51(4):167–74. pmid:27795585
38. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Human gut microbes associated with obesity. Nature. 2006;444(7122):1022–3. pmid:17183309
39. Schwiertz A, Taras D, Schafer K, Beijer S, Bos NA, Donus C, et al. Microbiota and SCFA in lean and overweight healthy subjects. Obesity (Silver Spring). 2010;18(1):190–5. pmid:19498350
40. Guida S, Venema K. Gut microbiota and obesity: Involvement of the adipose tissue. J Funct Foods. 2015;14:407–23.
41. Deschasaux M, Bouter KE, Prodan A, Levin E, Groen AK, Herrema H, et al. Depicting the composition of gut microbiota in a population with varied ethnic origins but shared geography. Nat Med. 2018;24(10):1526–31. pmid:30150717
42. Morales-Marroquin E, Hanson B, Greathouse L, de la Cruz-Munoz N, Messiah SE. Comparison of methodological approaches to human gut microbiota changes in response to metabolic and bariatric surgery: A systematic review. Obes Rev. 2020;21(8):e13025. pmid:32249534
43. Snetselaar LG, de Jesus JM, DeSilva DM, Stoody EE. Dietary Guidelines for Americans, 2020–2025: Understanding the Scientific Process, Guidelines, and Key Recommendations. Nutrition Today. 2021;56(6):287–95. pmid:34987271
44. Van der Kooy K, Seidell JC. Techniques for the measurement of visceral fat: a practical guide. Int J Obes Relat Metabolic Disorders. 1993;17(4):187–96pmid:8387967
45. Ford ES. Prevalence of the metabolic syndrome defined by the International Diabetes Federation among adults in the US. Diabetes Care. 2005;28(11):2745–9. pmid:16249550
46. Ashwell M, Gibson S. Waist-to-height ratio as an indicator of ‘early health risk’: simpler and more predictive than using a ‘matrix’based on BMI and waist circumference. BMJ Open. 2016;6(3):e010159. pmid:26975935
47. Romero-Corral A, Somers VK, Sierra-Johnson J, Thomas RJ, Collazo-Clavell M, Korinek J, et al. Accuracy of body mass index in diagnosing obesity in the adult general population. Int J Obes. 2008;32(6):959–66.
48. Xian X, Quach A, Bridgeman D, Tsow F, Forzani E, Tao N. Personalized indirect calorimeter for energy expenditure (EE) measurement. Global J Obes Diabetes Metabolic Syndrome. 2015;2(1):004–8.
49. Wilson M-MG, Thomas DR, Rubenstein LZ, Chibnall JT, Anderson S, Baxi A, et al. Appetite assessment: simple appetite questionnaire predicts weight loss in community-dwelling adults and nursing home residents. Am J Clin Nutr. 2005;82(5):1074–81. pmid:16280441
50. World Health Organization. Translation and cross-cultural adaptation of the World Health Organization Disability Assessment Schedule –WHODAS 2.0 2017. Available from: https://www.scielo.br/j/fp/a/6h8GLr8crZcMwtY9f9hfrND/?format=pdf&lang=en.
51. Mechanick JI, Apovian C, Brethauer S, Garvey WT, Joffe AM, Kim J, et al. Clinical practice guidelines for the perioperative nutrition, metabolic, and nonsurgical support of patients undergoing bariatric procedures - 2019 update: cosponsored by American association of clinical endocrinologists/American college of endocrinology, the obesity society, American society for metabolic & bariatric surgery, obesity medicine association, and American society of anesthesiologists - executive summary. Endocr Pract. 2019;25(12):1346–59. pmid:31682518
52. Wallace TM, Levy JC, Matthews DR. Use and abuse of HOMA modeling. Diabetes Care. 2004;27(6):1487–95. pmid:15161807
53. Esteghamati A, Ashraf H, Esteghamati A-R, Meysamie A, Khalilzadeh O, Nakhjavani M, et al. Optimal threshold of homeostasis model assessment for insulin resistance in an Iranian population: the implication of metabolic syndrome to detect insulin resistance. Diabetes Res Clin Pract. 2009;84(3):279–87. pmid:19359063
54. Ahmad MA, Karavetian M, Moubareck CA, Wazz G, Mahdy T, Venema K. Association of the gut microbiota with clinical variables in obese and lean Emirati subjects. Front Microbiol. 2023;14.
55. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37(8):852–7. pmid:31341288
56. Anderson M. Permutational Multivariate Analysis of Variance (PERMANOVA). 2017;1–15.
57. Schober P, Boer C, Schwarte LA. Correlation Coefficients: Appropriate Use and Interpretation. Anesth Analg. 2018;126(5):1763–8. pmid:29481436
58. Seganfredo F, Blume C, Moehlecke M, Giongo A, Casagrande D, Spolidoro J, et al. Weight‐loss interventions and gut microbiota changes in overweight and obese patients: a systematic review. Obes Rev. 2017;18(8):832–51.
59. Wang FG, Bai RX, Yan WM, Yan M, Dong LY, Song MM. Differential composition of gut microbiota among healthy volunteers, morbidly obese patients and post-bariatric surgery patients. Exp Ther Med. 2019;17(3):2268–78. pmid:30867711
60. Guo Y, Liu CQ, Shan CX, Chen Y, Li HH, Huang ZP, et al. Gut microbiota after Roux-en-Y gastric bypass and sleeve gastrectomy in a diabetic rat model: Increased diversity and associations of discriminant genera with metabolic changes. Diabetes Metab Res Rev. 2017;33(3).
61. Basso N, Soricelli E, Castagneto-Gissey L, Casella G, Albanese D, Fava F, et al. Insulin Resistance, Microbiota, and Fat Distribution Changes by a New Model of Vertical Sleeve Gastrectomy in Obese Rats. Diabetes. 2016;65(10):2990–3001. pmid:27431457
62. Campisciano G, Palmisano S, Cason C, Giuricin M, Silvestri M, Guerra M, et al. Gut microbiota characterisation in obese patients before and after bariatric surgery. Benef Microbes. 2018;9(3):367–73. pmid:29482339
63. Guo Y, Huang ZP, Liu CQ, Qi L, Sheng Y, Zou DJ. Modulation of the gut microbiome: a systematic review of the effect of bariatric surgery. Eur J Endocrinol. 2018;178(1):43–56. pmid:28916564
64. Ward EK, Schuster DP, Stowers KH, Royse AK, Ir D, Robertson CE, et al. The effect of PPI use on human gut microbiota and weight loss in patients undergoing laparoscopic Roux-en-Y gastric bypass. Obes Surg. 2014;24(9):1567–71. pmid:24849914
65. Palleja A, Kashani A, Allin KH, Nielsen T, Zhang C, Li Y, et al. Roux-en-Y gastric bypass surgery of morbidly obese patients induces swift and persistent changes of the individual gut microbiota. Genome Med. 2016;8(1):1–13.
66. Castaner O, Goday A, Park YM, Lee SH, Magkos F, Shiow STE, et al. The Gut Microbiome Profile in Obesity: A Systematic Review. Int J Endocrinol. 2018;2018:4095789. pmid:29849617
67. Cook J, Lehne C, Weiland A, Archid R, Ritze Y, Bauer K, et al. Gut microbiota, probiotics and psychological states and behaviors after bariatric surgery—a systematic review of their interrelation. Nutrients. 2020;12(8):2396. pmid:32785153
68. Hamamah S, Hajnal A, Covasa M. Influence of Bariatric Surgery on Gut Microbiota Composition and Its Implication on Brain and Peripheral Targets. Nutrients. 2024;16(7):1071. pmid:38613104
69. Liu R, Hong J, Xu X, Feng Q, Zhang D, Gu Y, et al. Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention. Nat Med. 2017;23(7):859–68. pmid:28628112
70. Cortez RV, Petry T, Caravatto P, Pessôa R, Sanabani SS, Martinez MB, et al. Shifts in intestinal microbiota after duodenal exclusion favor glycemic control and weight loss: a randomized controlled trial. Surg Obes Relat Dis. 2018;14(11):1748–54. pmid:30174193
71. Ulker I, Yildiran H. The effects of bariatric surgery on gut microbiota in patients with obesity: a review of the literature. Biosci Microbiota Food Health. 2018;38(1):3–9.
72. Shen N, Caixàs A, Ahlers M, Patel K, Gao Z, Dutia R, et al. Longitudinal changes of microbiome composition and microbial metabolomics after surgical weight loss in individuals with obesity. Surg Obes Relat Dis. 2019;15(8):1367–73. pmid:31296445
73. Luijten J, Vugts G, Nieuwenhuijzen GAP, Luyer MDP. The Importance of the Microbiome in Bariatric Surgery: a Systematic Review. Obes Surg. 2019;29(7):2338–49.
74. Debédat J, Clément K, Aron-Wisnewsky J. Gut Microbiota Dysbiosis in Human Obesity: Impact of Bariatric Surgery. Curr Obes Rep. 2019;8(3):229–42. pmid:31197613
75. Sweeney TE, Morton JM. Metabolic surgery: action via hormonal milieu changes, changes in bile acids or gut microbiota? A summary of the literature. Best Pract Res Clin Gastroenterol. 2014;28(4):727–40. pmid:25194186
76. Duncan SH, Lobley G, Holtrop G, Ince J, Johnstone A, Louis P, et al. Human colonic microbiota associated with diet, obesity and weight loss. Int J Obes. 2008;32(11):1720–4.
77. Jinatham V, Kullawong N, Kespechara K, Gentekaki E, Popluechai S. Comparison of Gut Microbiota between Lean and Obese Adult Thai Individuals. Microbiol Biotechnol Lett. 2018;46(3):277–87.
78. Patrone V, Vajana E, Minuti A, Callegari ML, Federico A, Loguercio C, et al. Postoperative Changes in Fecal Bacterial Communities and Fermentation Products in Obese Patients Undergoing Bilio-Intestinal Bypass. Front Microbiol. 2016;7:200. pmid:26941724
79. Oduro-Donkor D, Turner MC, Farnaud S, Renshaw D, Kyrou I, Hanson P, 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(5):363–73. pmid:32840125
80. Schneeberger M, Everard A, Gómez-Valadés AG, Matamoros S, Ramírez S, Delzenne NM, et al. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci Rep. 2015;5(1):16643. pmid:26563823
81. Depommier C, Everard A, Druart C, Plovier H, Van Hul M, Vieira-Silva S, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med. 2019;25(7):1096–103. pmid:31263284
82. Davies N, O’Sullivan JM, Plank LD, Murphy R. Gut Microbial Predictors of Type 2 Diabetes Remission Following Bariatric Surgery. Obes Surg. 2020;30(9):3536–48. pmid:32447634
83. Ballini A, Dipalma G, Isacco CG, Boccellino M, Di Domenico M, Santacroce L, et al. Oral Microbiota and Immune System Crosstalk: A Translational Research. Biology (Basel). 2020;9(6):131.
84. Han H, Yi B, Zhong R, Wang M, Zhang S, Ma J, et al. From gut microbiota to host appetite: gut microbiota-derived metabolites as key regulators. Microbiome. 2021;9(1):162. pmid:34284827
85. McCarty TR, Jirapinyo P, Thompson CC. Effect of Sleeve Gastrectomy on Ghrelin, GLP-1, PYY, and GIP Gut Hormones: A Systematic Review and Meta-analysis. Ann Surg. 2020;272(1):72–80. pmid:31592891
86. D’Alessio D. Is GLP-1 a hormone: Whether and When? J Diabetes Investig. 2016;7(Suppl 1):50–5. pmid:27186356
87. Farey JE, Preda TC, Fisher OM, Levert-Mignon AJ, Stewart RL, Karsten E, et al. Effect of Laparoscopic Sleeve Gastrectomy on Fasting Gastrointestinal, Pancreatic, and Adipose-Derived Hormones and on Non-Esterified Fatty Acids. Obes Surg. 2017;27(2):399–407. pmid:27465935
88. Yousseif A, Emmanuel J, Karra E, Millet Q, Elkalaawy M, Jenkinson AD, et al. Differential Effects of Laparoscopic Sleeve Gastrectomy and Laparoscopic Gastric Bypass on Appetite, Circulating Acyl-ghrelin, Peptide YY3-36 and Active GLP-1 Levels in Non-diabetic Humans. Obes Surg. 2014;24(2):241–52. pmid:23996294
89. Tsoli M, Chronaiou A, Kehagias I, Kalfarentzos F, Alexandrides TK. Hormone changes and diabetes resolution after biliopancreatic diversion and laparoscopic sleeve gastrectomy: a comparative prospective study. Surg Obes Relat Dis. 2013;9(5):667–77. pmid:23466015
90. Huang R, Ding X, Fu H, Cai Q. Potential mechanisms of sleeve gastrectomy for reducing weight and improving metabolism in patients with obesity. Surg Obes Relat Dis. 2019;15(10):1861–71. pmid:31375442
91. Tabasi M, Ashrafian F, Khezerloo JK, Eshghjoo S, Behrouzi A, Javadinia SA, et al. Changes in Gut Microbiota and Hormones After Bariatric Surgery: a Bench-to-Bedside Review. Obes Surg. 2019;29(5):1663–74. pmid:30793228
92. Magouliotis DE, Tasiopoulou VS, 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(5):1345–57. pmid:28265960
93. Reimer RA, Willis HJ, Tunnicliffe JM, Park H, Madsen KL, Soto-Vaca A. Inulin-type fructans and whey protein both modulate appetite but only fructans alter gut microbiota in adults with overweight/obesity: A randomized controlled trial. Mol Nutr Food Res. 2017;61(11). pmid:28730743
94. Davies Naomi K, O’Sullivan JM, Plank LD, Murphy R. Altered gut microbiome after bariatric surgery and its association with metabolic benefits: A systematic review. Surg Obes Relat Dis. 2019;15(4):656–65.
95. Kocełak P, Zak-Gołąb A, Zahorska-Markiewicz B, Aptekorz M, Zientara M, Martirosian G, et al. Resting energy expenditure and gut microbiota in obese and normal weight subjects. Eur Rev Med Pharmacol Sci. 2013;17(20):2816–21pmid:24174366
96. Gutiérrez-Repiso C, Moreno-Indias I, Tinahones FJ. Shifts in gut microbiota and their metabolites induced by bariatric surgery. Impact of factors shaping gut microbiota on bariatric surgery outcomes. Rev Endocr Metab Disord. 2021;22(4):1137–56. pmid:34287758
97. Tremaroli V, Karlsson F, Werling M, Stahlman M, Kovatcheva-Datchary P, Olbers T, et al. 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(2):228–38. pmid:26244932
98. Medina DA, Li T, Thomson P, Artacho A, Perez-Brocal V, Moya A. Cross-regional view of functional and taxonomic microbiota composition in obesity and post-obesity treatment shows country specific microbial contribution. Front Microbiol. 2019;10:2346. pmid:31681211
99. Graessler J, Qin Y, Zhong H, Zhang J, Licinio J, Wong ML, et al. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: correlation with inflammatory and metabolic parameters. Pharmacogenomics J. 2013;13(6):514–22. pmid:23032991
100. Yu D, Shu XO, Howard EF, Long J, English WJ, Flynn CR. Fecal metagenomics and metabolomics reveal gut microbial changes after bariatric surgery. Surg Obes Relat Dis. 2020;16(11):1772–82. pmid:32747219
101. Bhat VK, Kerr BD, Flatt PR, Gault VA. A novel GIP-oxyntomodulin hybrid peptide acting through GIP, glucagon and GLP-1 receptors exhibits weight reducing and anti-diabetic properties. Biochem Pharmacol. 2013;85(11):1655–62. pmid:23518155
102. Turnbaugh PJ, Backhed F, Fulton L, Gordon JI. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe. 2008;3(4):213–23. pmid:18407065
103. le Roux CW, Bueter M, Theis N, Werling M, Ashrafian H, Löwenstein C, et al. Gastric bypass reduces fat intake and preference. Am J Physiol Regul Integr Comp Physiol. 2011;301(4):R1057–66. pmid:21734019
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
© 2025 Ahmad et al. This is an open access article distributed under the terms of the Creative Commons Attribution License: http://creativecommons.org/licenses/by/4.0/ (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
Abstract
Background
Obesity is a growing health concern worldwide, including United Arab Emirates. Bariatric surgery is an effective treatment option, with to date unclear weight loss mechanisms. In this prospective study, we explored post-bariatric surgery changes in energy homeostasis, gut peptides, hormones, and gut microbiota.
Method
We recruited 19 Emirati adults who were planning to undergo sleeve gastrectomy (SG). We assessed the energy requirements using 24-hour diet recalls, indirect calorimetry for resting energy expenditure (REE), and a questionnaire for appetite.
Anthropometrics included body mass index (BMI), waist circumference, waist-to-height ratio, fat mass, fat-free mass, and percentage of body fat. Gut peptides, including peptide YY (PYY), glucagon-like peptide-1/2 (GLP-1/2), ghrelin (GHR), cholecystokinin (CCK), insulin, and leptin, were quantified using ELISA. Gut microbiota composition at phylum and genus levels, including the Firmicutes/Bacteroidetes (F/B) ratio and alpha (α) and beta (β) diversity, was determined by sequencing amplicons of the V3-V4 region of the 16S rRNA at baseline and three months post-surgery. Comparisons used paired sample T-test, Wilcoxon, and McNemar test. QIIME 2 was used to identify taxa and their relative abundance; subsequent analyses were done in R for (α) and (β) diversity (package qiime2R) and Wilcoxon signed-rank test in R for differences in microbiota at phylum and genus levels. We conducted Spearman correlation analyses between genera and energy homeostasis, appetite, anthropometrics, hormones, and gut peptides.
Results
At three months post-SG, energy intake, appetite, all anthropometric indices, insulin, leptin, and GLP-1 significantly decreased; PYY and GHR significantly increased, and REE was stable. β-diversity of the gut microbiota and its composition at phylum and genus levels significantly changed post-surgery, yet F/B remained constant. Energy intake, BMI, and appetite negatively correlated with several taxa that significantly increased post-SG.
Conclusion
Gut peptides, hormones, and microbiota change partly account for bariatric surgery’s weight-loss benefits. Understanding these alterations can inform personalized interventions targeting obesity.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer