Next Article in Journal
Exposure to Environmentally Relevant Levels of PFAS Causes Metabolic Changes in the Freshwater Amphipod Austrochiltonia subtenuis
Next Article in Special Issue
Associations between Periodontitis, COVID-19, and Cardiometabolic Complications: Molecular Mechanisms and Clinical Evidence
Previous Article in Journal
Anti-Diabetic and Cytotoxic Evaluation of Phlomis stewartii Plant Phytochemicals on Cigarette Smoke Inhalation and Alloxan-Induced Diabetes in Wistar Rats
Previous Article in Special Issue
Vitamin D Alleviates Anxiety and Depression in Elderly People with Prediabetes: A Randomized Controlled Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechanisms and Outcomes of Metabolic Surgery in Type 2 Diabetes

by
Mansor Fazliana
1,* and
Zubaidah Nor Hanipah
2
1
Nutrition, Metabolism and Cardiovascular Research Centre, Institute for Medical Research, National Institutes of Health, Ministry of Health Malaysia, Selangor 40170, Malaysia
2
Department of Surgery, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia
*
Author to whom correspondence should be addressed.
Metabolites 2022, 12(11), 1134; https://doi.org/10.3390/metabo12111134
Submission received: 12 October 2022 / Revised: 9 November 2022 / Accepted: 14 November 2022 / Published: 17 November 2022
(This article belongs to the Special Issue New Therapeutic Targets and Treatment Options in Metabolic Syndrome)

Abstract

:
This review is aimed at synthesizing the mechanisms and outcomes of metabolic surgery on the endocrine system, microbiome, metabolomics, and at the molecular level. We review the hormonal, adipokine, microbiota, microRNA, and metabolomic changes in human and animal models following metabolic surgery for the treatment of obesity and diabetes. The most relevant studies in this area over the past 17 years have been considered for this review. In most cases, metabolic procedures, especially those that include intestinal bypass components, showed the remission of type 2 diabetes. This involves a variety of weight-independent mechanisms to improve glucose homeostasis, improving insulin sensitivity and secretion, gut microbiota, and bile acid cross-talk.

1. Introduction

Globally rising sedentary lifestyles and obesity have increased, resulting in the tremendous burden of type 2 diabetes (T2D) worldwide. Bay et al. reported in a study conducted in the US that approximately 85% of patients with T2D are either overweight or obese [1]. The literature has shown that weight loss surgery (i.e., bariatric or metabolic surgery) has been proven to be effective and provides good long-term glycemic control in patients with obesity and T2D. Metabolic surgery is now considered a well-supported treatment for T2D in patients with obesity and is endorsed by international diabetes and medical organizations [2].
Metabolic surgery exerts its physiological benefits by mediating intestinal physiology, bile acid metabolism, incretin hormone secretion, neuronal signaling, and microbiome changes. Understanding the mechanisms of metabolic procedures and their outcomes on diabetes will ensure optimal treatment and disease prevention strategies in T2D patients. Although the benefits of metabolic surgery for obesity-related comorbidities are well-established, evidence of its molecular and metabolomic effects is still limited.
This review article summarizes the effects of metabolic surgery on the endocrine system, gut microbiota, microRNA levels, and metabolomics for the treatment of obesity and diabetes, both in clinical trials as well as in animal models.

2. Overview of Bariatric/Metabolic Surgery

Bariatric surgery was initially performed for weight loss (baros = weight) [3]. It was initially applied to the treatment of morbid obesity, as documented in 1954 [4]. The procedure was originally designed to achieve and sustain weight loss, and it was consequently noted to induce improvements in glucose regulation [5,6]. Bariatric surgical procedures included jejunoileal bypass (JIB), Roux-en-Y gastric bypass (RYGB), sleeve gastrectomy (SG), biliopancreatic diversion (BPD), vertical-banded gastroplasty (VBG) or its familiar duodenal switch (DS), and adjustable gastric banding (AGB) [7]. Currently, RYGB and SG are two of the most popular types of bariatric surgeries that are applied in the treatment of obesity and T2D worldwide [8]. RYGB and SG have very different anatomical structures, but to varying degrees both surgeries are effective at inducing weight loss and T2D remission [9].
The surgery itself has led to the mechanical hypothesis that bariatric operations generally promote weight loss by restricting the stomach size and/or bypassing some of the intestines to cause nutrient malabsorption. However, the remission of T2D has been observed after gastric bypass before the occurrence of significant weight reduction. It shows that the glucose improvement resulting from metabolic operations occurs not only because of weight loss but also due to weight-independent factors, such as gut hormones, bile acids, gut microbiota, the nervous system, and other potential underlying mechanisms [10,11,12,13]. Short- and long-term clinical studies have shown that bariatric surgery results in substantial weight loss and either the improvement or remission of T2D [13,14,15]. As clinical data accumulated, some versions of these procedures were ultimately abandoned because of complications related to the surgery.
Many studies have shown that bariatric surgery, which is now known as metabolic surgery, improves glucose homeostasis. Bariatric procedures, especially those with intestinal bypass components, have been shown to lead to the remission in T2D. In a recent 2-year follow-up study, the complete remission of T2D was achieved in nearly half of patients two years after metabolic surgery [14]. A large cohort with prospective randomized interventional data demonstrated that metabolic surgery improves glycemic control, diabetes-related comorbidities, and weight loss to a greater extent than lifestyle/medical intervention for up to 3 years postsurgery [15]. Studies assessing the impact of metabolic surgery on the control of diabetes are summarized in Table 1.
The principal determinants of glucose homeostasis, which include insulin secretion, insulin sensitivity, and insulin-independent glucose disposal, are affected by RYGB, VBG, and BPD [16]. A comprehensive understanding of these effects helped to optimize surgical techniques and devices to provide the maximum antidiabetes impact.
Table 1. Summary of studies assessing the impact of metabolic surgery on the control of diabetes.
Table 1. Summary of studies assessing the impact of metabolic surgery on the control of diabetes.
StudyIntervention/
Surgery Type
Subjects (n)ResultsOutcomes
Vrakopoulou et al. (2021) [17]SG
OAGB
morbidly obese T2D patients
SG (n = 28)
OAGB (n = 25)
SG-10 patients (35.7%) achieved diabetes remission.
OAGB-22 patients (88%) remained off antidiabetic agents.
OAGB was more effective in improving glycemic control and %EWL with an almost immediate resolution of diabetes as well as long-term weight loss.
Moriconi et al. (2021) [18]RYGBRYGB: 88 obese patients
Medical therapy (MT): 25 obese patients
10-year follow-up
Body mass index (BMI), fasting glucose, and hemoglobin A1c (HbA1c) ↓ in RYGB than MT patients. RYGB significantly reduced and sustained glycated hemoglobin (HbA1c) levels compared to medical therapy at a ten-year follow-up.
Weight loss impacted the short-term remission of T2D but had a marginal role in long-term relapse.
Imtiaz et al. (2021) [19]bariatric surgeryRandomized controlled trials (RCTs) among T2D patients
3674 with surgery
1335 without surgery
HbA1C ↓
Improvements were sustained for at least 6 years.
Bariatric surgery lowered A1c in real-world clinical care.
Purnell et al. (2021) [20]RYGB
LAGB
2256 participants, 827 with T2D and severe obesityDiabetes remission occurred in 57% (46% complete, 11% partial) after RYGB and
22.5% (16.9% complete, 5.6% partial) after LAGB.
younger participants, shorter diabetes duration: remission↑
Durable long-term diabetes remission following bariatric surgery was more likely when performed soon after diagnosis, when the diabetes medication burden was low and beta-cell function was preserved.
Huang et al. (2021) [21]single-anastomosis gastric bypass1999 patients from the Asian Diabetes Surgery Summit
1 year
Weight↓
BMI ↓
Blood glucose ↓
HbA1c ↓
Lipid ↓
Blood pressure ↓
Metabolic surgery remarkably improved body weight, T2D, and other metabolic disorders in Asian patients. However, the efficacy of individual procedures varied substantially.
Stenberg et al. (2020) [22]RYGB742 patients at
baseline and 2 years postsurgery
Insulin homeostasis and glucometabolic control were improved and sustained.Positive results were observed in diabetics as well as prediabetics and nondiabetic obese patients, and this improvement was sustained for 2 years after surgery.
Katsogiannos et al. (2020) [23]RYGB19 obese T2D patientsparasympathetic nerve activity ↑ morning cortisol ↓
incretin ↑
glucagon responses ↑
Neurohormonal mechanisms can contribute to the rapid improvement in insulin resistance and glycemia following RYGB in T2D.

3. Mechanisms Involved in the Postoperative Weight and Metabolic Changes

Multiple mechanisms are responsible for postoperative weight and metabolic changes. Obesity and T2D are biologically linked, with obesity being a primary driver of insulin resistance. It is also implicated in β-cell decompensation [24]. Long-term weight loss can dramatically improve the lives of people with established T2D [25,26,27,28]. A negative energy balance and the weight loss it produces have powerful effects on many physiologic factors that could lead to the remission of the T2D disease process [29]. The association between weight loss and its powerful effects in improving these comorbidities is only now being explored. We discuss the mechanisms that are responsible for postoperative weight and metabolic changes. These involve some degree of overlap involving the endocrine, digestive, nervous, and immune systems and metabolomic changes as well as changes on the molecular level. The hormonal and adipokine changes after metabolic surgery include adiponectin, ghrelin, leptin, glucagon-like peptide, secretin, and oxyntomodulin [30,31,32].

3.1. Hormonal Changes

Metabolic surgery improves insulin secretion and sensitivity in patients with T2D, but the effect on patients with normal glucose tolerance or prediabetes (pre-DM) needs further understanding. Stenberg et al. [22] followed 742 patients after RYGB. The glucometabolic control and insulin homeostasis improved in all these patients, and these results were sustained for 2 years. In another study following RYGB in T2D patients, there was a significant improvement in glycemia and insulin resistance, which could be due to neurohormonal mechanisms [33].
The absorption of glucose and protein was greatly accelerated after RYGB but was only modestly accelerated after SG. Insulin, glucagon-like peptide-1 (GLP-1), cholecystokinin (CCK), and peptide YY (PYY) are appetite inhibitors, and their secretions also differed markedly between the procedures [34]. The mechanisms underlying the benefits of metabolic surgery likely involve the bile acid signaling pathway [35].
Sixty-nine subjects who underwent RYGB were compared with matched controls who had never been obese in a 2-year longitudinal study. The RYGB group showed slightly increased HOMA-IR insulin sensitivity, a beneficial body composition, higher insulin clearance, and lower atherogenic lipid and lipoprotein levels as well as benign adipocyte morphology (p < 0.0001 for these parameters). This partly explained why long-term metabolic complications were protected by RYGB [36]. A cross-sectional study of 36 T2D patients was performed to study the absorption rates of glucose and protein as well as the profiles of gastro-entero-pancreatic hormones after metabolic surgery [34]. Metabolic surgery was performed in 24 patients (LSG, n = 12; RYGB, n = 12) and compared to a control group (n = 12). They received continuous infusions of stable isotopes of glucose, glycerol, phenylalanine, tyrosine, and urea before and during a mixed meal containing labeled glucose and intrinsically phenylalanine-labeled caseinate. This study showed that the systemic appearance of ingested glucose was faster after RYGB and SG vs. controls, while the peak glucose appearance rate was 64% higher after RYGB and 23% higher after SG (both p < 0.05). The postprandial glucose and protein absorption and gastro-entero-pancreatic hormone secretions differed after SG and RYGB. RYGB was characterized by the accelerated absorption of glucose and amino acids, whereas protein metabolism after SG did not differ significantly from controls, suggesting that different mechanisms explain the improved glycemic control and weight loss after these surgical procedures [34].
In a long-term study involving 163 patients with T2D, metabolic surgery was able to reduce obesity-related chronic low-grade inflammation. The acute-phase reactants C-reactive proteins (CRP, p < 0.001) and high-sensitivity CRP (hs-CRP, p < 0.001) are commonly used to monitor inflammation and are strongly associated with metabolic syndrome, atherosclerotic cardiovascular disease, and T2D. These markers were significantly reduced for up to 4 years after surgical interventions. The improvement was related to the change in BMI and the remission of T2D in the long term [37].
Ghrelin and GLP-1 have a very important role in this mechanism, which will be discussed further below.
a.
Ghrelin
The main ghrelin functions are targeted at appetite, metabolism, and adiposity. Acylated and deacyl ghrelin are the two main circulating isoforms of this hormone [38]. These isoforms consist of the main elements that are involved in the amelioration of nonalcoholic fatty-liver disease (NAFLD) after bariatric surgery [39]. In a ten-week diabetic rat study, RYGB and SG decreased leptin and ghrelin levels [40]. Ghrelin affects carbohydrate and lipid metabolism in obese patients. After RYGB and LSG surgeries, ghrelin was associated with elevated plasma levels of insulin, leptin, and glucagon [41].
b.
Glucagon-like peptide-1 (GLP-1)
RYGB leads to profound changes in the secretion of gut hormones, with effects on food intake and appetite as well as metabolism [29]. Several studies highlighted the important role of GLP-1 in achieving glycemic control. GLP-1 is elevated after RYGB. Bile acid plasma concentrations were increased after RYGB. In this case, bile acids may act as molecular enhancers of GLP-1 secretion through the activation of TGR5 receptors [42]. The improved glucose tolerance was due to a negative energy balance and the resulting weight loss. In the beginning, these improve hepatic insulin sensitivity, and they later improve peripheral insulin sensitivity. Next, in combination with elevated postprandial insulin secretion, insulin sensitivity is elicited, particularly by magnified GLP-1 responses [29]. Additionally, RYGB causes a weight-loss-independent postprandial insulin secretion, which contributes to the improvement in glycemic control. This action is associated with a ~10-fold increment in the concentrations of the incretin hormone GLP-1 in plasma [43].
A meta-analysis by Jirapinyo et al. of 24 studies involving 368 patients concluded that GLP-1 fasting levels remain unchanged, but the levels increase after RYGB. Interestingly, shorter a Roux limb length is associated with a greater increase in postprandial GLP-1, which may improve glycemic control [29].
Using a model of gastrectomy in lean mice, Larraufie et al. showed that after bariatric surgery GLP-1 is an enhancement factor of insulin secretion, which arises from fast nutrient delivery to the distal gut [44]. In an animal study mentioned earlier, RYGB and SG increased GLP-1 levels [40], while another study showed the complete remission of T2D, which was significantly associated with higher GLP-1 levels [14].
An animal study revealed that the microbiota changes by RYGB play a key role in postsurgical weight loss [45]. Besides bile acids, short-chain fatty acids (SCFAs) were also reported to modify the secretion of GLP-1 [46]. Likely due to RYGB at the point of inclusion of the bypass of the proximal intestine, elevated GLP-1 secretion, altered gut microbiota, and increased SCFA in the gut were reported after pancreatoduodenectomy [47]. The effect of gut microbiota composition on lipid profiles was analyzed after RYGB. It was found that SCFA-producing bacteria promote healthy lipid homeostasis, while the presence of LPS-producing bacteria such Escherichia-Shigella may contribute to the development of atherogenic dyslipidemia [48]. Microbiota changes in metabolic surgery are discussed further in Section 3.3.

3.2. Adipokine Changes

Adipose tissue dysfunctionality could be caused by excessive visceral fat accumulation. This contributes significantly to the onset of obesity-related comorbidities [49]. Higher visceral fat at follow-up exams was significantly associated with reduced remission, which increased the incidence of diabetes, dyslipidemia, and hypertension [50]. Hepatic, adipose, and skeletal muscle tissues are the crucial endocrine organs that produce hepatokines, adipokines, and myokines. They are biomarkers that can be beneficial or detrimental to an organism and act through the autocrine, endocrine, and paracrine pathways [51].
Among the varieties of adipokines produced by adipose tissue are cytokines (TNF-α, TGFβ, IL-R1a, IL-6, and IL-10), chemokines (MCP-1, CCL2, CCL5, macrophage inflammatory protein-2 (MIP-2), IL-8/CXCL8, and IFNγ-inducible protein 10/ CXCL10), acute-phase reactants (haptoglobin, serum amyloid A, C-reactive protein, and plasminogen activator inhibitor 1), damage-associated molecular pattern molecules (calprotectin, DAMPs, HMGB1, tenascin C, and heat shock protein 72), and proinflammatory (leptin, osteopontin, resistin, WNT5A, and chemerin) as well as an anti-inflammatory (ghrelin, adiponectin, SRFP5, lipocalin-2, and omentin) adipokines [30]. Obesity is associated with a changed secretion of adipokines that translates into increased cardiovascular risk in patients. These obese patients have an excess of dysfunctional adiposity [49].
The functions of specific adipokines and their effects on metabolic surgery are further discussed.
a.
Leptin
A product of the obesity gene, leptin takes part in the regulation of body weight by controlling food intake and energy expenditure [52]. Leptin activates a balanced effect on blood pressure control in the healthy state by modulating the endothelial release of nitric oxide as well as sympathetic activity-dependent vasoconstriction and angiotensin II-dependent vasoconstriction [30,53]. In obesity, hyperleptinemia may emerge as a compensatory mechanism to control leptin resistance. This is due to obesity, which activates an organ-specific leptin-resistant state [54]. Šebunova N. et al. analyzed 30 obese bariatric patients and found a remarkable decrease in leptin levels [55].
b.
Adiponectin
Adiponectin is only expressed in adipose tissue, which is detectable in plasma. Plasma adiponectin concentrations are reduced in obese patients. Adiponectin exerts anti-inflammatory actions as well as increases insulin sensitivity [56]. Metabolic syndrome and insulin resistance in humans are best predicted by high-molecular-weight adiponectin [57].
Moreover, the adiponectin/leptin ratio has been recommended as a marker of adipose tissue dysfunction. The ratio correlates with insulin resistance more closely than a surrogate of insulin resistance such as the HOMA index, adiponectin, or leptin alone [58]. Metabolic surgery resulted in weight loss and activated a constant decrease in leptin levels and a parallel increase in adiponectin plasma levels [55,59].
c.
Resistin and visfatin
Adipose-tissue-resident macrophages secrete resistin, which is a polypeptide. Its concentrations are elevated in obesity. This is because the pathophysiology of inflammation-induced insulin resistance in macrophages is regulated by circulating resistin levels [60]. Prospective case-control studies proved an association between an increased risk of developing T2D and subjects with increased resistin levels at baseline. These levels decreased after bariatric surgery [61,62].
Another adipocytokine secreted by adipocytes, inflamed endothelial tissue, and macrophages is visfatin. It increases obesity, insulin resistance, and T2D. Visfatin acts as a proinflammatory intermediary has an important role in vascular inflammation pathogenesis in obesity and T2D, and contributes to atherosclerotic plaque instability. An improvement in insulin resistance and diabetes was reflected in T2D patients who underwent RYGB when the visfatin serum level was decreased [62,63].
d.
Omentin-1 and apelin
Omentin-1, also known as intelectin-1, is an adipokine that is primarily secreted from visceral adipose tissue and consists of 313 amino acids, but it is also expressed in the heart, placenta, and ovaries [64,65]. It is an anti-inflammatory adipokine [66] that is expressed in omental, epicardial, and perivascular adipose tissue [67]. In obesity, the omentin-1 level, which is the major circulating form, is reduced. It is also inversely correlated to waist circumference, BMI, and metabolic syndrome biomarkers [68]. Its expression is reduced in obesity [69]. During diet-induced weight loss, omentin-1 levels usually elevate over time, which is evidence of a link between omentin and obesity [70].
The circulating omentin levels and the related mRNA expression in visceral adipose tissue are distinct in different types of diabetes [68]. There might be a protective action of omentin in metabolic disorders [71]. Omentin reduced insulin resistance in Goto-Kakizaki rats fed a high-fat diet without affecting their lipid profile [72]. Interestingly, a systematic review and meta-analysis found that serum omentin levels are significantly lower in impaired glucose tolerance and T2DM patients but not in type 1 diabetes (T1DM) [73].
In a study, most postbariatric patients displayed an elevation of omentin-1 levels in the immediate postoperative period. This condition even occurred before the induction of weight loss. The increment in omentin-1 levels was even maintained for one year after the bariatric intervention [74].
Another novel adipokine, apelin, has a crucial role in the pathogenesis of insulin resistance as well as T2D. Apelin is secreted from white adipose tissue and is associated with various functions, including food intake and insulin sensitivity [75,76]. The level of apelin in obese patients with T2D is significantly increased compared to healthy people [77]. In obesity and diabetes, insulin could control apelin [78].
Long-term apelin treatment in insulin-resistant obese mice has proven, valuable effects on both glucose and lipid metabolism [79]. During a hyperinsulinemic–euglycemic clamp in nondiabetic human volunteers, apelin perfusion markedly improved insulin sensitivity without causing side effects [80].

3.3. Role of Gut Microbiota, Bile Acids, and Their Cross-Talk

One of the important mechanisms for the improvement in T2D after metabolic surgery is the change in the gut microbiota. The gut microbiota structure has an important role in the improvement in islet β-cell function and the hypoglycemic effect. Body weight gain, blood serum lipids, and fasting blood glucose are effectively decreased by the modified jejunoileal bypass [81]. This potential therapeutic strategy for T2D is explained by the jejunoileal bypass, which has modified and improved the gut microbiota composition [81]. Additionally, insulin resistance, islet β-cell function, and glucose tolerance were significantly improved. In a T2D rat experimental model, it was suggested that the islet β-cell function might be contributed by amino acid metabolism [82].
More evidence suggests that the gut microbiota is associated with the development of several metabolic disorders. Bile acids and nuclear bile acid receptor (FXR) signaling are important for the metabolic benefits of metabolic surgery. Furthermore, the microbiota–bile acid interactions play a role in the mechanisms underlying the effects of metabolic surgery [83]. Metabolic surgery can change the intestinal microorganism pattern in response to gastric restriction or the rearrangement of the intestinal tract. Altered nutrient presentation resulting from an incompletely digested diet that enters the downstream gut after different surgical procedures could alter the gut environment and affect the composition of the intestinal bacteria [84]. In a long-term effect study, it was found that the gut microbiota may play a direct role in the reduction in adiposity after metabolic surgery, as demonstrated in the rodent model. The surgically altered microbiota in recipient mice promoted reduced fat deposition [45].
The transfer of the gut microbiota from RYGB-treated mice to sham-operated, germ-free mice resulted in weight loss and decreased fat mass in the recipient animals. This was potentially due to altered microbial production of short-chain fatty acids. These studies show that changes in the gut microbiota contribute to reduced host weight and adiposity after RYGB surgery [85].
The combination of physiology and the computational modeling of microbiota metabolism would motivate researchers to optimize the diagnosis and treatment of T2D patients in a personalized way [86]. This could be applied in metabolic surgery as an option for the treatments.
In addition to inducing inflammation, the gut microbiota plays an important function in modulating bile acids and including their biosynthesis and biotransformation. Cholic acid, one of the bile acids, regulates the gut microbiota composition in rats, inducing changes similar to those induced by high-fat diets. This explains the relationship between the gastrointestinal microbiota composition and metabolic diseases [87]. Altered bile acid levels and compositions may contribute to improved glucose and lipid metabolism in patients who have had gastric bypass [88]. Postsurgical alterations occur in intestinal anatomy, satiety, the secretion of gastrointestinal peptides, neural signaling, and nutrient absorption. These aspects contribute to weight loss and the associated improvements in systemic metabolism. Additionally, an increase in serum bile acid levels also contributes to improved carbohydrate, lipid, and energy metabolism after bariatric surgery. Bile acids and nuclear bile acid receptor FXR signaling were found to be important molecular underpinnings for the beneficial effects of this weight loss surgery [89].

3.4. Molecular Changes—MicroRNA

MicroRNAs (miRNAs) are expressed in various organs [90]. miRNAs are short pieces of RNA that are recognized as key gene expression regulators and have main roles in the regulation of many biological and pathological processes, including T2D [91]. Owing to their stability and practicality in noninvasive collection methods, circulating miRNAs could be used as biomarkers. This is being explored in a wide range of pathologies, including diabetes and cancer. Furthermore, their levels can be measured by quantitative RT-PCR, which is straightforward, fast, and specific, with sensitive detection and quantification [92].
The differential expression of circulating miRNAs before and after various dietary and bariatric surgery interventions has been reported in a few studies, identifying several weight-loss-related candidate biomarkers. A range of dysregulated miRNA target pathways has also been identified. This is to understand the underlying obesity and obesity-related metabolic disease pathophysiological mechanisms [93].
Dysfunctional adipose tissue is extensively associated with T2D development and is the major source of circulating miRNAs. A specific miRNA, miR-122, which is distinctively found between visceral and subcutaneous adipose tissues, was found in a study [94]. It is involved in weight homeostasis as well as numerous metabolic processes [95]. The ratio of miR-122 between subcutaneous and visceral adipose tissues correlates with the outcome of bariatric surgery [94].
miRNA disorders have been demonstrated in various studies involving β-cell development, insulin production, insulin secretion, insulin sensitivity, insulin resistance, and insulin signaling pathways and finally lead to the development of T2D [96]. These findings support the possible role of miR-33 in monitoring prediabetes onset and progression.
The metabolic responses in the early stages following weight loss after bariatric surgery are evident. These observations correspond with an improvement in diabetes. Seven microRNAs (let-7i-5p, let-7f-5p, miR-7-5p, miR-15b-5p, miR-205-5p, miR-320c, and miR-335-5p) showed significant changes 3 weeks after RYGB surgery among 29 patients with severe obesity and T2D. Altered miRNA functional pathways were associated with liver-, diabetes-, and pituitary-related diseases. Following bariatric surgery, the miRNA expression in natural killer cells and vital intestinal pathology imply mechanistic functions in early diabetes responses [97].
In a diet and cardiovascular study cohort, the baseline levels of miR-223-3p were found to be significantly related to insulin resistance in adipose tissue. Both miR-223-3p-secreting preadipocytes and miR-223-3p-secreting adipocytes caused alterations in the circulating levels. This suggested that inflammation enhances the intracellular accumulation of miR-223-3p. This possibly contributes to preadipocyte dysfunction and body metabolic dysregulation [98].
The usefulness of identifying genetic differences between high- and low-weight-loss groups after bariatric surgery by identifying specific serum miRNA has been demonstrated [95]. The miRNA profile in the serum of plasma is deregulated in the pre-DM state before the development of observable T2D. Undoubtedly, compared with controls, individuals with T2D or pre-DM have a differential profile of circulating miRNAs [99]. Interestingly, a list of miRNAs has been identified in nondiabetic healthy individuals who proceeded to develop pre-DM or T2D [100]. Additionally, multiple circulating miRNA plasma concentrations of healthy individuals have been identified as being markedly different between T2D patients and pre-DM individuals [101,102]. Deregulated plasma levels of miR-15a, miR-30a-5p, miR-150, and miR-375 were detected years before the onset of T2D and pre-DM and could be utilized to assess the risk of developing the disease. This may improve the prediction and prevention among high-risk individuals for T2D [103]. Eikelis et al. also demonstrated that the plasma levels of miR-9, miR-28-3p, miR29a, miR-103, miR-30a-5p, and miR-150 are powerful predictive biomarkers that can discriminate between incident-T2D and non-T2D patients. A potential tool for the early detection of T2D has been developed. It is a multiparameter diagnostic model consisting of miR-148b, miR-223, miR-130a, and miR-19a [104]. miR-132 (mir-132) is an important regulator of liver homeostasis and lipid metabolism. In the same experimental model, an association between miR-132 and the markers of metabolic and cardiovascular disease was found [104]. Part of the blood biochemical changes of diabetes reversal was formed during the resolution of diabetes after bariatric surgery. This process occurred through the miRNA–gene interactions in the pancreatic islet, which is a novel mechanism [105].
Soon, informed decisions about surgery could be facilitated by these miRNAs. These potential miRNA biomarkers could also provide targets for future treatments by opening new genetic pathways that illustrate the pathophysiology of obesity. Table 2 presents differentially expressed miRNAs with their roles/targets after surgery.

3.5. Metabolomics

As a relatively young scientific discipline, metabolomics shows great potential for the comprehensive study of the metabolome’s dynamic alterations. Liquid chromatography–mass spectrometry (LC-MS) and nuclear magnetic resonance (NMR) are the most frequently used techniques to study the main effects of RYGB or SG. NMR can uniquely identify and simultaneously quantify a wide range of analyses of amino acids, carbohydrates, vitamins, thiols, and peptides as well as nucleotides and nucleosides. The LC-MS technique has become a powerful tool for the analysis of the polar metabolites in a complex sample [115,116].
In an exploratory study among 17 diabetic and nondiabetic obese patients undergoing bariatric surgery, untargeted metabolomic profiling in subcutaneous adipose tissue was performed. However, among the 421 metabolites that were identified and analyzed, there were no significant differences between those patients [117]. The dysregulation of lipids and amino acids has been associated with insulin resistance and other pathophysiological processes of T2DM. This may be due to obesity, which may influence subcutaneous adipose tissue metabolism, masking T2DM-dependent dysregulation [118].
In another post-RYGB study, alterations in basal metabolism among overweight diabetic subjects were demonstrated by NMR metabolomic profiling. These changes were associated with energy homeostasis, alterations in lipid metabolism, and decreased branched-chain amino acids [119].
Most methods for the screening and prevention of T2D rely on prediabetic individuals that are already showing a steady decrease in insulin sensitivity. It is important to develop biomarker trajectory models that can accurately complement the existing individual risk assessment methods because these methods may not be as effective as those developed to counter the disease [120].
In a 5-year diabetes remission study, metabolites in the branched-chain amino acid (BCAA) and trimethylamine-N-oxide (TMAO) microbiome-related pathways were found to be predictive of T2D remission and weight loss amounts in severely obese individuals [121]. These metabolites can potentially be used in the clinical management of T2DM patients undergoing bariatric surgery. Additionally, the baseline levels of tryptophan, bilirubin, and indoxyl sulfate measured before surgery as well as the levels of FFA 16:0, FFA 18:3, FFA 17:2, and hippuric acid measured 6 months after surgery best predicted the suitability and efficacy of RYGB for patients with T2DM [122]. A summary of the metabolomic profiles associated with metabolic surgery is presented in Table 3.

4. Conclusions

Multidisciplinary weight management approaches are emerging as feasible and potentially cost-effective treatment options for patients with overweight/obesity and diabetes. Metabolic surgery is a potential and sustainable treatment that can modify a patient’s physiology and glucose regulation mechanism. In most cases, metabolic procedures show the remission of T2D. This involves a variety of weight-independent mechanisms to improve glucose homeostasis, improving insulin sensitivity and secretion.
The potent metabolic effects of metabolic surgery are not only shown by improved obesity, glucose tolerance, and insulin sensitivity. Recent studies show that microbiota–bile acid interactions play a role in the mechanisms underlying the effects of metabolic surgery. Furthermore, metabolic improvement could be monitored by miRNA levels.
The overall message for clinicians is that metabolic surgery can be an excellent and life-extending option for some patients with T2D. However, these procedures are associated with perioperative risks and other aspects that are not discussed in this review, such as permanently changing patients’ relationship with food, sometimes involving psychiatric disorders and requiring lifelong diet support and medical monitoring. These aspects need further discussion in their respective areas.

Author Contributions

Conceptualization, M.F.; writing—original draft preparation, M.F.; writing—review and editing, Z.N.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Ministry of Health of Malaysia, grant number NMRR-20-2496-56353; project code: 21-004.

Acknowledgments

We would like to thank the Director General of Health Malaysia for his permission to publish this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Bays, H.E.; Bazata, D.D.; Clark, N.G.; Gavin, J.R.; Green, A.J.; Lewis, S.J.; Reed, M.L.; Stewart, W.; Chapman, R.H.; Fox, K.M.; et al. Prevalence of Self-Reported Diagnosis of Diabetes Mellitus and Associated Risk Factors in a National Survey in the US Population: SHIELD (Study to Help Improve Early Evaluation and Management of Risk Factors Leading to Diabetes). BMC Public Health 2007, 7, 277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Hanipah, Z.N.; Schauer, P.R. Bariatric Surgery as a Long-Term Treatment for Type 2 Diabetes/Metabolic Syndrome. Annu. Rev. Med. 2020, 71, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Pollard, S. The Current Status of Bariatric Surgery. Frontline Gastroenterol. 2011, 2, 90–95. [Google Scholar] [CrossRef]
  4. Kremen, A.J.; Linner, J.H.; Nelson, C.H. An Experimental Evaluation of the Nutritional Importance of Proximal and Distal Small Intestine. Ann. Surg. 1954, 140, 439–448. [Google Scholar] [CrossRef] [PubMed]
  5. Mason, E.E. History of Obesity Surgery. Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2005, 1, 123–125. [Google Scholar] [CrossRef] [PubMed]
  6. Pories, W.J.; Caro, J.F.; Flickinger, E.G.; Meelheim, H.D.; Swanson, M.S. The Control of Diabetes Mellitus (NIDDM) in the Morbidly Obese with the Greenville Gastric Bypass. Ann. Surg. 1987, 206, 316–323. [Google Scholar] [CrossRef]
  7. Buchwald, H. The Evolution of Metabolic/Bariatric Surgery. Obes. Surg. 2014, 24, 1126–1135. [Google Scholar] [CrossRef]
  8. Nguyen, N.T.; Varela, J.E. Bariatric Surgery for Obesity and Metabolic Disorders: State of the Art. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 160–169. [Google Scholar] [CrossRef]
  9. Sandoval, D.A.; Patti, M.E. Glucose Metabolism after Bariatric Surgery: Implications for T2DM Remission and Hypoglycaemia. Nat. Rev. Endocrinol. 2022. [Google Scholar] [CrossRef]
  10. Shah, S.S.; Todkar, J.S.; Shah, P.S.; Cummings, D.E. Diabetes Remission and Reduced Cardiovascular Risk After Gastric Bypass in Asian Indians with Body Mass Index <35 Kg/M2. Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2010, 6, 332–338. [Google Scholar] [CrossRef]
  11. Thaler, J.P.; Cummings, D.E. Minireview: Hormonal and Metabolic Mechanisms of Diabetes Remission after Gastrointestinal Surgery. Endocrinology 2009, 150, 2518–2525. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Miras, A.D.; le Roux, C.W. Mechanisms Underlying Weight Loss after Bariatric Surgery. Nat. Rev. Gastroenterol. Hepatol. 2013, 10, 575–584. [Google Scholar] [CrossRef] [PubMed]
  13. Seeley, R.J.; Chambers, A.P.; Sandoval, D.A. The Role of Gut Adaptation in the Potent Effects of Multiple Bariatric Surgeries on Obesity and Diabetes. Cell Metab. 2015, 21, 369–378. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. AbdAlla Salman, M.; Rabiee, A.; Salman, A.; Elewa, A.; Tourky, M.; Mahmoud, A.A.; Moustafa, A.; El-Din Shaaban, H.; Ismail, A.A.; Noureldin, K.; et al. Predictors of Type-2 Diabetes Remission Following Bariatric Surgery after a Two-Year Follow Up. Asian J. Surg. 2022. [Google Scholar] [CrossRef] [PubMed]
  15. Kirwan, J.P.; Courcoulas, A.P.; Cummings, D.E.; Goldfine, A.B.; Kashyap, S.R.; Simonson, D.C.; Arterburn, D.E.; Gourash, W.F.; Vernon, A.H.; Jakicic, J.M.; et al. Diabetes Remission in the Alliance of Randomized Trials of Medicine Versus Metabolic Surgery in Type 2 Diabetes (ARMMS-T2D). Diabetes Care 2022, 45, 1574–1583. [Google Scholar] [CrossRef]
  16. Mingrone, G.; Cummings, D.E. Changes of Insulin Sensitivity and Secretion after Bariatric/Metabolic Surgery. Surg. Obes. Relat. Dis. 2016, 12, 1199–1205. [Google Scholar] [CrossRef]
  17. Vrakopoulou, G.Z.; Theodoropoulos, C.; Kalles, V.; Zografos, G.; Almpanopoulos, K. Type 2 Diabetes Mellitus Status in Obese Patients Following Sleeve Gastrectomy or One Anastomosis Gastric Bypass. Sci. Rep. 2021, 11, 4421. [Google Scholar] [CrossRef]
  18. Moriconi, D.; Manca, M.L.; Anselmino, M.; Rebelos, E.; Bellini, R.; Taddei, S.; Ferrannini, E.; Nannipieri, M. Predictors of Type 2 Diabetes Relapse after Roux-En-Y Gastric Bypass: A Ten-Year Follow-up Study. Diabetes Metab. 2021, 48, 101282. [Google Scholar] [CrossRef]
  19. Imtiaz, R.; Doumouras, A.G.; Hong, D.; Anvari, M.; Shah, B.R. Long-Term Impact of Bariatric Surgery on Glycemic Control and Glucose-Lowering Therapy for People with Type 2 Diabetes Population-Based Cohort Study. Surg. Obes. Relat. Dis. 2021, 17, 1049–1056. [Google Scholar] [CrossRef]
  20. Purnell, J.Q.; Dewey, E.N.; Laferrère, B.; Selzer, F.; Flum, D.R.; Mitchell, J.E.; Pomp, A.; Pories, W.J.; Inge, T.; Courcoulas, A.; et al. Diabetes Remission Status During Seven-Year Follow-up of the Longitudinal Assessment of Bariatric Surgery Study. J. Clin. Endocrinol. Metab. 2021, 106, 774–788. [Google Scholar] [CrossRef]
  21. Huang, Y.-M.; Lin, Y.-K.; Lee, W.-J.; Hur, K.Y.; Kasama, K.; Cheng, A.K.S.; Lee, M.-H.; Wong, S.K.-H.; Soong, T.-C.; Lee, K.-T.; et al. Long-Term Outcomes of Metabolic Surgery in Overweight and Obese Patients with Type 2 Diabetes in Asia. Diabetes Obes. Metab. 2021, 23, 742–753. [Google Scholar] [CrossRef] [PubMed]
  22. Stenberg, E.; Rask, E.; Szabo, E.; Näslund, I.; Ottosson, J. The Effect of Laparoscopic Gastric Bypass Surgery on Insulin Resistance and Glycosylated Hemoglobin A1c: A 2-Year Follow-up Study. Obes. Surg. 2020, 30, 3489–3495. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Katsogiannos, P.; Kamble, P.G.; Wiklund, U.; Sundbom, M.; Espes, D.; Hammar, U.; Karlsson, F.A.; Pereira, M.J.; Eriksson, J.W. Rapid Changes in Neuroendocrine Regulation May Contribute to Reversal of Type 2 Diabetes after Gastric Bypass Surgery. Endocrine 2020, 67, 344–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Eckel, R.H.; Kahn, S.E.; Ferrannini, E.; Goldfine, A.B.; Nathan, D.M.; Schwartz, M.W.; Smith, R.J.; Smith, S.R. Obesity and Type 2 Diabetes: What Can Be Unified and What Needs to Be Individualized? J. Clin. Endocrinol. Metab. 2011, 96, 1654–1663. [Google Scholar] [CrossRef] [Green Version]
  25. Knowler, W.C.; Barrett-Connor, E.; Fowler, S.E.; Hamman, R.F.; Lachin, J.M.; Walker, E.A.; Nathan, D.M.; Diabetes Prevention Program Research Group. Reduction in the Incidence of Type 2 Diabetes with Lifestyle Intervention or Metformin. N. Engl. J. Med. 2002, 346, 393–403. [Google Scholar] [CrossRef]
  26. Lindström, J.; Ilanne-Parikka, P.; Peltonen, M.; Aunola, S.; Eriksson, J.G.; Hemiö, K.; Hämäläinen, H.; Härkönen, P.; Keinänen-Kiukaanniemi, S.; Laakso, M.; et al. Sustained Reduction in the Incidence of Type 2 Diabetes by Lifestyle Intervention: Follow-up of the Finnish Diabetes Prevention Study. Lancet Lond. Engl. 2006, 368, 1673–1679. [Google Scholar] [CrossRef]
  27. Li, G.; Zhang, P.; Wang, J.; Gregg, E.W.; Yang, W.; Gong, Q.; Li, H.; Li, H.; Jiang, Y.; An, Y.; et al. The Long-Term Effect of Lifestyle Interventions to Prevent Diabetes in the China Da Qing Diabetes Prevention Study: A 20-Year Follow-up Study. Lancet Lond. Engl. 2008, 371, 1783–1789. [Google Scholar] [CrossRef]
  28. Diabetes Prevention Program Research Group; Knowler, W.C.; Fowler, S.E.; Hamman, R.F.; Christophi, C.A.; Hoffman, H.J.; Brenneman, A.T.; Brown-Friday, J.O.; Goldberg, R.; Venditti, E.; et al. 10-Year Follow-up of Diabetes Incidence and Weight Loss in the Diabetes Prevention Program Outcomes Study. Lancet Lond. Engl. 2009, 374, 1677–1686. [Google Scholar] [CrossRef] [Green Version]
  29. Holst, J.J.; Madsbad, S.; Bojsen-Møller, K.N.; Svane, M.S.; Jørgensen, N.B.; Dirksen, C.; Martinussen, C. Mechanisms in Bariatric Surgery: Gut Hormones, Diabetes Resolution, and Weight Loss. Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2018, 14, 708–714. [Google Scholar] [CrossRef] [Green Version]
  30. Rodríguez, A.; Ezquerro, S.; Méndez-Giménez, L.; Becerril, S.; Frühbeck, G. Revisiting the Adipocyte: A Model for Integration of Cytokine Signaling in the Regulation of Energy Metabolism. Am. J. Physiol. Endocrinol. Metab. 2015, 309, E691–E714. [Google Scholar] [CrossRef]
  31. Luo, J.N.; Tavakkoli, A. Physiologic Mechanisms of Weight Loss Following Metabolic/Bariatric Surgery. Surg. Clin. N. Am. 2021, 101, 223–237. [Google Scholar] [CrossRef] [PubMed]
  32. Estienne, A.; Bongrani, A.; Reverchon, M.; Ramé, C.; Ducluzeau, P.-H.; Froment, P.; Dupont, J. Involvement of Novel Adipokines, Chemerin, Visfatin, Resistin and Apelin in Reproductive Functions in Normal and Pathological Conditions in Humans and Animal Models. Int. J. Mol. Sci. 2019, 20, 4431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Katsogiannos, P.; Kamble, P.G.; Boersma, G.J.; Karlsson, F.A.; Lundkvist, P.; Sundbom, M.; Pereira, M.J.; Eriksson, J.W. Early Changes in Adipose Tissue Morphology, Gene Expression, and Metabolism After RYGB in Patients With Obesity and T2D. J. Clin. Endocrinol. Metab. 2019, 104, 2601–2613. [Google Scholar] [CrossRef] [PubMed]
  34. Svane, M.S.; Bojsen-Møller, K.N.; Martinussen, C.; Dirksen, C.; Madsen, J.L.; Reitelseder, S.; Holm, L.; Rehfeld, J.F.; Kristiansen, V.B.; van Hall, G.; et al. Postprandial Nutrient Handling and Gastrointestinal Hormone Secretion After Roux-En-Y Gastric Bypass vs Sleeve Gastrectomy. Gastroenterology 2019, 156, 1627–1641.e1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Xu, G.; Song, M. Recent Advances in the Mechanisms Underlying the Beneficial Effects of Bariatric and Metabolic Surgery. Surg. Obes. Relat. Dis. 2021, 17, 231–238. [Google Scholar] [CrossRef]
  36. Andersson, D.P.; Dahlman, I.; Eriksson Hogling, D.; Bäckdahl, J.; Toft, E.; Qvisth, V.; Näslund, E.; Thorell, A.; Rydén, M.; Arner, P. Improved Metabolism and Body Composition beyond Normal Levels Following Gastric Bypass Surgery: A Longitudinal Study. J. Intern. Med. 2019, 285, 92–101. [Google Scholar] [CrossRef] [Green Version]
  37. Lautenbach, A.; Stoll, F.; Mann, O.; Busch, P.; Huber, T.B.; Kielstein, H.; Bähr, I.; Aberle, J. Long-Term Improvement of Chronic Low-Grade Inflammation After Bariatric Surgery. Obes. Surg. 2021, 31, 2913–2920. [Google Scholar] [CrossRef]
  38. Gauna, C.; Delhanty, P.J.D.; Hofland, L.J.; Janssen, J.A.M.J.L.; Broglio, F.; Ross, R.J.M.; Ghigo, E.; van der Lely, A.J. Ghrelin Stimulates, Whereas Des-Octanoyl Ghrelin Inhibits, Glucose Output by Primary Hepatocytes. J. Clin. Endocrinol. Metab. 2005, 90, 1055–1060. [Google Scholar] [CrossRef] [Green Version]
  39. Ezquerro, S.; Méndez-Giménez, L.; Becerril, S.; Moncada, R.; Valentí, V.; Catalán, V.; Gómez-Ambrosi, J.; Frühbeck, G.; Rodríguez, A. Acylated and Desacyl Ghrelin Are Associated with Hepatic Lipogenesis, β-Oxidation and Autophagy: Role in NAFLD Amelioration after Sleeve Gastrectomy in Obese Rats. Sci. Rep. 2016, 6, 39942. [Google Scholar] [CrossRef] [Green Version]
  40. Kornyushin, O.; Sonin, D.; Polozov, A.; Masley, V.; Bulavinova, N.; Chervyak, M.; Istomina, M.; Mukhametdinova, D.; Neimark, A.; Cheburkin, Y.; et al. Effect of Sleeve Gastrectomy, Roux-En-Y Gastric Bypass, and Ileal Transposition on Myocardial Ischaemia-Reperfusion Injury in Non-Obese Non-Diabetic Rats. Sci. Rep. 2021, 11, 23888. [Google Scholar] [CrossRef]
  41. Skuratovskaia, D.; Vulf, M.; Chasovskikh, N.; Komar, A.; Kirienkova, E.; Shunkin, E.; Zatolokin, P.; Litvinova, L. The Links of Ghrelin to Incretins, Insulin, Glucagon, and Leptin After Bariatric Surgery. Front. Genet. 2021, 12, 612501. [Google Scholar] [CrossRef] [PubMed]
  42. Nielsen, S.; Svane, M.S.; Kuhre, R.E.; Clausen, T.R.; Kristiansen, V.B.; Rehfeld, J.F.; Holst, J.J.; Madsbad, S.; Bojsen-Moller, K.N. Chenodeoxycholic Acid Stimulates Glucagon-like Peptide-1 Secretion in Patients after Roux-En-Y Gastric Bypass. Physiol. Rep. 2017, 5, e13140. [Google Scholar] [CrossRef] [PubMed]
  43. Hindsø, M.; Svane, M.S.; Hedbäck, N.; Holst, J.J.; Madsbad, S.; Bojsen-Møller, K.N. The Role of GLP-1 in Postprandial Glucose Metabolism after Bariatric Surgery: A Narrative Review of Human GLP-1 Receptor Antagonist Studies. Surg. Obes. Relat. Dis. 2021, 17, 1383–1391. [Google Scholar] [CrossRef] [PubMed]
  44. Larraufie, P.; Roberts, G.P.; McGavigan, A.K.; Kay, R.G.; Li, J.; Leiter, A.; Melvin, A.; Biggs, E.K.; Ravn, P.; Davy, K.; et al. Important Role of the GLP-1 Axis for Glucose Homeostasis after Bariatric Surgery. Cell Rep. 2019, 26, 1399–1408.e6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Tremaroli, V.; Karlsson, F.; Werling, M.; Ståhlman, M.; Kovatcheva-Datchary, P.; Olbers, T.; Fändriks, L.; le Roux, C.W.; Nielsen, J.; Bäckhed, F. Roux-En-Y Gastric Bypass and Vertical Banded Gastroplasty Induce Long-Term Changes on the Human Gut Microbiome Contributing to Fat Mass Regulation. Cell Metab. 2015, 22, 228–238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Kimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Fukuda, T.; Bouchi, R.; Takeuchi, T.; Amo-Shiinoki, K.; Kudo, A.; Tanaka, S.; Tanabe, M.; Akashi, T.; Hirayama, K.; Odamaki, T.; et al. Importance of Intestinal Environment and Cellular Plasticity of Islets in the Development of Postpancreatectomy Diabetes. Diabetes Care 2021, 44, 1002–1011. [Google Scholar] [CrossRef]
  48. López-Montoya, P.; Cerqueda-García, D.; Rodríguez-Flores, M.; López-Contreras, B.; Villamil-Ramírez, H.; Morán-Ramos, S.; Molina-Cruz, S.; Rivera-Paredez, B.; Antuna-Puente, B.; Velázquez-Cruz, R.; et al. Association of Gut Microbiota with Atherogenic Dyslipidemia, and Its Impact on Serum Lipid Levels after Bariatric Surgery. Nutrients 2022, 14, 3545. [Google Scholar] [CrossRef]
  49. Landecho, M.F.; Tuero, C.; Valentí, V.; Bilbao, I.; de la Higuera, M.; Frühbeck, G. Relevance of Leptin and Other Adipokines in Obesity-Associated Cardiovascular Risk. Nutrients 2019, 11, 2664. [Google Scholar] [CrossRef] [Green Version]
  50. Hunt, S.C.; Davidson, L.E.; Adams, T.D.; Ranson, L.; McKinlay, R.D.; Simper, S.C.; Litwin, S.E. Associations of Visceral, Subcutaneous, Epicardial, and Liver Fat with Metabolic Disorders up to 14 Years After Weight Loss Surgery. Metab. Syndr. Relat. Disord. 2021, 19, 83–92. [Google Scholar] [CrossRef]
  51. de Oliveira Dos Santos, A.R.; de Oliveira Zanuso, B.; Miola, V.F.B.; Barbalho, S.M.; Santos Bueno, P.C.; Flato, U.A.P.; Detregiachi, C.R.P.; Buchaim, D.V.; Buchaim, R.L.; Tofano, R.J.; et al. Adipokines, Myokines, and Hepatokines: Crosstalk and Metabolic Repercussions. Int. J. Mol. Sci. 2021, 22, 2639. [Google Scholar] [CrossRef] [PubMed]
  52. Muruzábal, F.J.; Frühbeck, G.; Gómez-Ambrosi, J.; Archanco, M.; Burrell, M.A. Immunocytochemical Detection of Leptin in Non-Mammalian Vertebrate Stomach. Gen. Comp. Endocrinol. 2002, 128, 149–152. [Google Scholar] [CrossRef]
  53. Gómez-Ambrosi, J.; Catalán, V.; Rodríguez, A.; Andrada, P.; Ramírez, B.; Ibáñez, P.; Vila, N.; Romero, S.; Margall, M.A.; Gil, M.J.; et al. Increased Cardiometabolic Risk Factors and Inflammation in Adipose Tissue in Obese Subjects Classified as Metabolically Healthy. Diabetes Care 2014, 37, 2813–2821. [Google Scholar] [CrossRef] [Green Version]
  54. Sweeney, G. Cardiovascular Effects of Leptin. Nat. Rev. Cardiol. 2010, 7, 22–29. [Google Scholar] [CrossRef] [PubMed]
  55. Šebunova, N.; Štšepetova, J.; Kullisaar, T.; Suija, K.; Rätsep, A.; Junkin, I.; Soeorg, H.; Lember, M.; Sillakivi, T.; Mändar, R. Changes in Adipokine Levels and Metabolic Profiles Following Bariatric Surgery. BMC Endocr. Disord. 2022, 22, 33. [Google Scholar] [CrossRef] [PubMed]
  56. Huypens, P.R. Leptin and Adiponectin Regulate Compensatory Beta Cell Growth in Accordance to Overweight. Med. Hypotheses 2007, 68, 1134–1137. [Google Scholar] [CrossRef] [PubMed]
  57. Yamauchi, T.; Kadowaki, T. Adiponectin Receptor as a Key Player in Healthy Longevity and Obesity-Related Diseases. Cell Metab. 2013, 17, 185–196. [Google Scholar] [CrossRef] [Green Version]
  58. Frühbeck, G.; Catalán, V.; Rodríguez, A.; Ramírez, B.; Becerril, S.; Salvador, J.; Colina, I.; Gómez-Ambrosi, J. Adiponectin-Leptin Ratio Is a Functional Biomarker of Adipose Tissue Inflammation. Nutrients 2019, 11, 454. [Google Scholar] [CrossRef] [Green Version]
  59. Unamuno, X.; Izaguirre, M.; Gómez-Ambrosi, J.; Rodríguez, A.; Ramírez, B.; Becerril, S.; Valentí, V.; Moncada, R.; Silva, C.; Salvador, J.; et al. Increase of the Adiponectin/Leptin Ratio in Patients with Obesity and Type 2 Diabetes after Roux-En-Y Gastric Bypass. Nutrients 2019, 11, 2069. [Google Scholar] [CrossRef] [Green Version]
  60. Park, H.K.; Kwak, M.K.; Kim, H.J.; Ahima, R.S. Linking Resistin, Inflammation, and Cardiometabolic Diseases. Korean J. Intern. Med. 2017, 32, 239–247. [Google Scholar] [CrossRef]
  61. Doulamis, I.P.; Konstantopoulos, P.; Tzani, A.; Antoranz, A.; Minia, A.; Daskalopoulou, A.; Charalampopoulos, A.; Alexopoulos, L.; Perrea, D.N.; Menenakos, E. Visceral White Adipose Tissue and Serum Proteomic Alternations in Metabolically Healthy Obese Patients Undergoing Bariatric Surgery. Cytokine 2019, 115, 76–83. [Google Scholar] [CrossRef] [PubMed]
  62. Arica, P.C.; Aydin, S.; Zengin, U.; Kocael, A.; Orhan, A.; Zengin, K.; Gelisgen, R.; Taskin, M.; Uzun, H. The Effects on Obesity Related Peptides of Laparoscopic Gastric Band Applications in Morbidly Obese Patients. J. Investig. Surg. Off. J. Acad. Surg. Res. 2018, 31, 89–95. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, Y.; Xiong, J.; He, H.; Zhu, S.; Mo, Z. Visfatin level after laparoscopic Roux-en-Y gastric bypass surgery in patients with Type 2 diabetes. Zhong Nan Da Xue Xue Bao Yi Xue Ban 2013, 38, 258–261. [Google Scholar] [CrossRef] [PubMed]
  64. Tok, A.; Ozer, A.; Kanat-Pektas, M.; Aral, M.; Sakalli, H.; Aydogdu, S.; Yutan-Kaya, E.; Sager, H. The Role of Omentin in Early Pregnancy Losses. J. Obstet. Gynaecol. J. Inst. Obstet. Gynaecol. 2020, 40, 107–110. [Google Scholar] [CrossRef] [PubMed]
  65. Watanabe, T.; Watanabe-Kominato, K.; Takahashi, Y.; Kojima, M.; Watanabe, R. Adipose Tissue-Derived Omentin-1 Function and Regulation. Compr. Physiol. 2017, 7, 765–781. [Google Scholar] [CrossRef]
  66. Tan, Y.-L.; Zheng, X.-L.; Tang, C.-K. The Protective Functions of Omentin in Cardiovascular Diseases. Clin. Chim. Acta Int. J. Clin. Chem. 2015, 448, 98–106. [Google Scholar] [CrossRef]
  67. Yang, R.-Z.; Lee, M.-J.; Hu, H.; Pray, J.; Wu, H.-B.; Hansen, B.C.; Shuldiner, A.R.; Fried, S.K.; McLenithan, J.C.; Gong, D.-W. Identification of Omentin as a Novel Depot-Specific Adipokine in Human Adipose Tissue: Possible Role in Modulating Insulin Action. Am. J. Physiol. Endocrinol. Metab. 2006, 290, E1253–E1261. [Google Scholar] [CrossRef]
  68. Goodarzi, G.; Shirgir, A.; Alavi, S.; Khoshi, A. Effect of Insulin–Glucose Metabolism Compared with Obesity on Adipose Omentin Gene Expression in Different Models of Diabetic C57BL/6 Mice. Diabetol. Metab. Syndr. 2019, 11, 65. [Google Scholar] [CrossRef] [Green Version]
  69. Zhou, H.; Zhang, Z.; Qian, G.; Zhou, J. Omentin-1 Attenuates Adipose Tissue Inflammation via Restoration of TXNIP/NLRP3 Signaling in High-Fat Diet-Induced Obese Mice. Fundam. Clin. Pharmacol. 2020, 34, 721–735. [Google Scholar] [CrossRef]
  70. Moreno-Navarrete, J.M.; Catalán, V.; Ortega, F.; Gómez-Ambrosi, J.; Ricart, W.; Frühbeck, G.; Fernández-Real, J.M. Circulating Omentin Concentration Increases after Weight Loss. Nutr. Metab. 2010, 7, 27. [Google Scholar] [CrossRef]
  71. Lis, I.; Pelczyńska, M.; Miller-Kasprzak, E.; Kujawska-Łuczak, M.; Mądry, E.; Bogdański, P. Association of Serum Omentin Concentration with Anthropometric, Physiological, and Biochemical Parameters in Obese Individuals. Nutrition 2020, 79–80, 110866. [Google Scholar] [CrossRef]
  72. Leandro, A.; Queiroz, M.; Azul, L.; Seiça, R.; Sena, C.M. Omentin: A Novel Therapeutic Approach for the Treatment of Endothelial Dysfunction in Type 2 Diabetes. Free Radic. Biol. Med. 2021, 162, 233–242. [Google Scholar] [CrossRef] [PubMed]
  73. As´habi, A.; Sadeghi, M.; Arab, A.; Hajianfar, H. The Association between Omentin and Diabetes: A Systematic Review and Meta-Analysis of Observational Studies. Diabetes Metab. Syndr. Obes. Targets Ther. 2019, 12, 1277–1286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Lapointe, M.; Poirier, P.; Martin, J.; Bastien, M.; Auclair, A.; Cianflone, K. Omentin Changes Following Bariatric Surgery and Predictive Links with Biomarkers for Risk of Cardiovascular Disease. Cardiovasc. Diabetol. 2014, 13, 124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Alipour, F.G.; Ashoori, M.R.; Pilehvar-Soltanahmadi, Y.; Zarghami, N. An Overview on Biological Functions and Emerging Therapeutic Roles of Apelin in Diabetes Mellitus. Diabetes Metab. Syndr. 2017, 11 (Suppl. S2), S919–S923. [Google Scholar] [CrossRef]
  76. Elsehmawy, A.A.E.W.; El-Toukhy, S.E.; Seliem, N.M.A.; Moustafa, R.S.; Mohammed, D.S. Apelin and Chemerin as Promising Adipokines in Children with Type 1 Diabetes Mellitus. Diabetes Metab. Syndr. Obes. Targets Ther. 2019, 12, 383–389. [Google Scholar] [CrossRef] [Green Version]
  77. Cavallo, M.G.; Sentinelli, F.; Barchetta, I.; Costantino, C.; Incani, M.; Perra, L.; Capoccia, D.; Romeo, S.; Cossu, E.; Leonetti, F.; et al. Altered Glucose Homeostasis Is Associated with Increased Serum Apelin Levels in Type 2 Diabetes Mellitus. PLoS ONE 2012, 7, e51236. [Google Scholar] [CrossRef] [Green Version]
  78. Yu, Y.; Fernandez, I.D.; Meng, Y.; Zhao, W.; Groth, S.W. Gut Hormones, Adipokines, and pro- and Anti-Inflammatory Cytokines/Markers in Loss of Control Eating: A Scoping Review. Appetite 2021, 166, 105442. [Google Scholar] [CrossRef]
  79. Bertrand, C.; Valet, P.; Castan-Laurell, I. Apelin and Energy Metabolism. Front. Physiol. 2015, 6, 115. [Google Scholar] [CrossRef]
  80. Gourdy, P.; Cazals, L.; Thalamas, C.; Sommet, A.; Calvas, F.; Galitzky, M.; Vinel, C.; Dray, C.; Hanaire, H.; Castan-Laurell, I.; et al. Apelin Administration Improves Insulin Sensitivity in Overweight Men during Hyperinsulinaemic-Euglycaemic Clamp. Diabetes Obes. Metab. 2018, 20, 157–164. [Google Scholar] [CrossRef]
  81. Tan, C.; Zheng, Z.; Wan, X.; Cao, J.; Wei, R.; Duan, J. The Role of Gut Microbiota and Amino Metabolism in the Effects of Improvement of Islet β-Cell Function after Modified Jejunoileal Bypass. Sci. Rep. 2021, 11, 4809. [Google Scholar] [CrossRef] [PubMed]
  82. Alejandro, E.U.; Gregg, B.; Blandino-Rosano, M.; Cras-Méneur, C.; Bernal-Mizrachi, E. Natural History of β-Cell Adaptation and Failure in Type 2 Diabetes. Mol. Aspects Med. 2015, 42, 19–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Liu, H.; Hu, C.; Zhang, X.; Jia, W. Role of Gut Microbiota, Bile Acids and Their Cross-Talk in the Effects of Bariatric Surgery on Obesity and Type 2 Diabetes. J. Diabetes Investig. 2018, 9, 13–20. [Google Scholar] [CrossRef] [PubMed]
  84. 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, 917–925. [Google Scholar] [CrossRef] [PubMed]
  85. Liou, A.P.; Paziuk, M.; Luevano, J.-M.; Machineni, S.; Turnbaugh, P.J.; Kaplan, L.M. Conserved Shifts in the Gut Microbiota Due to Gastric Bypass Reduce Host Weight and Adiposity. Sci. Transl. Med. 2013, 5, 178ra41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Martínez-López, Y.E.; Esquivel-Hernández, D.A.; Sánchez-Castañeda, J.P.; Neri-Rosario, D.; Guardado-Mendoza, R.; Resendis-Antonio, O. Type 2 Diabetes, Gut Microbiome, and Systems Biology: A Novel Perspective for a New Era. Gut Microbes 2022, 14, 2111952. [Google Scholar] [CrossRef] [PubMed]
  87. Islam, K.B.M.S.; Fukiya, S.; Hagio, M.; Fujii, N.; Ishizuka, S.; Ooka, T.; Ogura, Y.; Hayashi, T.; Yokota, A. Bile Acid Is a Host Factor That Regulates the Composition of the Cecal Microbiota in Rats. Gastroenterology 2011, 141, 1773–1781. [Google Scholar] [CrossRef] [PubMed]
  88. Patti, M.-E.; Houten, S.M.; Bianco, A.C.; Bernier, R.; Larsen, P.R.; Holst, J.J.; Badman, M.K.; Maratos-Flier, E.; Mun, E.C.; Pihlajamaki, J.; et al. Serum Bile Acids Are Higher in Humans With Prior Gastric Bypass: Potential Contribution to Improved Glucose and Lipid Metabolism. Obes. Silver Spring Md 2009, 17, 1671–1677. [Google Scholar] [CrossRef] [Green Version]
  89. Ryan, K.K.; Tremaroli, V.; Clemmensen, C.; Kovatcheva-Datchary, P.; Myronovych, A.; Karns, R.; Wilson-Pérez, H.E.; Sandoval, D.A.; Kohli, R.; Bäckhed, F.; et al. FXR Is a Molecular Target for the Effects of Vertical Sleeve Gastrectomy. Nature 2014, 509, 183–188. [Google Scholar] [CrossRef] [Green Version]
  90. Liu, J.; Yang, D.; Wang, B.; Zeng, Y.; Li, W. The Value of MiRNAs in the Prognosis of Obese Patients Receiving Bariatric Surgery. Am. J. Transl. Res. 2021, 13, 1905–1914. [Google Scholar]
  91. Flynt, A.S.; Lai, E.C. Biological Principles of MicroRNA-Mediated Regulation: Shared Themes amid Diversity. Nat. Rev. Genet. 2008, 9, 831–842. [Google Scholar] [CrossRef] [PubMed]
  92. de Planell-Saguer, M.; Rodicio, M.C. Detection Methods for MicroRNAs in Clinic Practice. Clin. Biochem. 2013, 46, 869–878. [Google Scholar] [CrossRef] [PubMed]
  93. Catanzaro, G.; Filardi, T.; Sabato, C.; Vacca, A.; Migliaccio, S.; Morano, S.; Ferretti, E. Tissue and Circulating MicroRNAs as Biomarkers of Response to Obesity Treatment Strategies. J. Endocrinol. Investig. 2021, 44, 1159–1174. [Google Scholar] [CrossRef] [PubMed]
  94. Liao, C.-H.; Wang, C.-Y.; Liu, K.-H.; Liu, Y.-Y.; Wen, M.-S.; Yeh, T.-S. MiR-122 Marks the Differences between Subcutaneous and Visceral Adipose Tissues and Associates with the Outcome of Bariatric Surgery. Obes. Res. Clin. Pract. 2018, 12, 570–577. [Google Scholar] [CrossRef]
  95. Doyon, L.; Das, S.; Sullivan, T.; Rieger-Christ, K.; Sherman, J.; Roque, S.; Nepomnayshy, D. Can Genetics Help Predict Efficacy of Bariatric Surgery? An Analysis of MicroRNA Profiles. Surg. Obes. Relat. Dis. Off. J. Am. Soc. Bariatr. Surg. 2020, 16, 1802–1807. [Google Scholar] [CrossRef]
  96. Guay, C.; Regazzi, R. MicroRNAs and the Functional β Cell Mass: For Better or Worse. Diabetes Metab. 2015, 41, 369–377. [Google Scholar] [CrossRef] [Green Version]
  97. Atkin, S.L.; Ramachandran, V.; Yousri, N.A.; Benurwar, M.; Simper, S.C.; McKinlay, R.; Adams, T.D.; Najafi-Shoushtari, S.H.; Hunt, S.C. Changes in Blood MicroRNA Expression and Early Metabolic Responsiveness 21 Days Following Bariatric Surgery. Front. Endocrinol. 2019, 9, 773. [Google Scholar] [CrossRef]
  98. Sánchez-Ceinos, J.; Rangel-Zuñiga, O.A.; Clemente-Postigo, M.; Podadera-Herreros, A.; Camargo, A.; Alcalá-Diaz, J.F.; Guzmán-Ruiz, R.; López-Miranda, J.; Malagón, M.M. MiR-223-3p as a Potential Biomarker and Player for Adipose Tissue Dysfunction Preceding Type 2 Diabetes Onset. Mol. Ther. Nucleic Acids 2021, 23, 1035–1052. [Google Scholar] [CrossRef]
  99. Yang, Z.; Chen, H.; Si, H.; Li, X.; Ding, X.; Sheng, Q.; Chen, P.; Zhang, H. Serum MiR-23a, a Potential Biomarker for Diagnosis of Pre-Diabetes and Type 2 Diabetes. Acta Diabetol. 2014, 51, 823–831. [Google Scholar] [CrossRef]
  100. Vasu, S.; Kumano, K.; Darden, C.M.; Rahman, I.; Lawrence, M.C.; Naziruddin, B. MicroRNA Signatures as Future Biomarkers for Diagnosis of Diabetes States. Cells 2019, 8, 1533. [Google Scholar] [CrossRef] [Green Version]
  101. Karolina, D.S.; Tavintharan, S.; Armugam, A.; Sepramaniam, S.; Pek, S.L.T.; Wong, M.T.K.; Lim, S.C.; Sum, C.F.; Jeyaseelan, K. Circulating MiRNA Profiles in Patients with Metabolic Syndrome. J. Clin. Endocrinol. Metab. 2012, 97, E2271–E2276. [Google Scholar] [CrossRef] [PubMed]
  102. Kong, L.; Zhu, J.; Han, W.; Jiang, X.; Xu, M.; Zhao, Y.; Dong, Q.; Pang, Z.; Guan, Q.; Gao, L.; et al. Significance of Serum MicroRNAs in Pre-Diabetes and Newly Diagnosed Type 2 Diabetes: A Clinical Study. Acta Diabetol. 2011, 48, 61–69. [Google Scholar] [CrossRef] [PubMed]
  103. Jiménez-Lucena, R.; Camargo, A.; Alcalá-Diaz, J.F.; Romero-Baldonado, C.; Luque, R.M.; van Ommen, B.; Delgado-Lista, J.; Ordovás, J.M.; Pérez-Martínez, P.; Rangel-Zúñiga, O.A.; et al. A Plasma Circulating MiRNAs Profile Predicts Type 2 Diabetes Mellitus and Prediabetes: From the CORDIOPREV Study. Exp. Mol. Med. 2018, 50, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Eikelis, N.; Dixon, J.B.; Lambert, E.A.; Hanin, G.; Tzur, Y.; Greenberg, D.S.; Soreq, H.; Marques, F.Z.; Fahey, M.T.; Head, G.A.; et al. MicroRNA-132 May Be Associated with Blood Pressure and Liver Steatosis-Preliminary Observations in Obese Individuals. J. Hum. Hypertens. 2021, 36, 911–916. [Google Scholar] [CrossRef] [PubMed]
  105. Amouyal, C.; Castel, J.; Guay, C.; Lacombe, A.; Denom, J.; Migrenne-Li, S.; Rouault, C.; Marquet, F.; Georgiadou, E.; Stylianides, T.; et al. A Surrogate of Roux-En-Y Gastric Bypass (the Enterogastro Anastomosis Surgery) Regulates Multiple Beta-Cell Pathways during Resolution of Diabetes in Ob/Ob Mice. EBioMedicine 2020, 58, 102895. [Google Scholar] [CrossRef]
  106. Sangiao-Alvarellos, S.; Theofilatos, K.; Barwari, T.; Gutmann, C.; Takov, K.; Singh, B.; Juiz-Valiña, P.; Varela-Rodríguez, B.M.; Outeiriño-Blanco, E.; Duregotti, E.; et al. Metabolic Recovery after Weight Loss Surgery Is Reflected in Serum MicroRNAs. BMJ Open Diabetes Res. Care 2020, 8, e001441. [Google Scholar] [CrossRef]
  107. Cereijo, R.; Taxerås, S.D.; Piquer-Garcia, I.; Pellitero, S.; Martínez, E.; Tarascó, J.; Moreno, P.; Balibrea, J.; Puig-Domingo, M.; Jiménez-Pavón, D.; et al. Elevated Levels of Circulating MiR-92a Are Associated with Impaired Glucose Homeostasis in Patients with Obesity and Correlate with Metabolic Status After Bariatric Surgery. Obes. Surg. 2020, 30, 174–179. [Google Scholar] [CrossRef]
  108. Lopez, Y.N.; Coen, P.; Goodpaster, B.; Seyhan, A. Gastric Bypass Surgery with Exercise Alters Plasma MicroRNAs That Predict Improvements in Cardiometabolic Risk. Int. J. Obes. 2005 2017, 41, 1121–1130. [Google Scholar] [CrossRef] [Green Version]
  109. Hubal, M.J.; Nadler, E.P.; Ferrante, S.C.; Barberio, M.D.; Suh, J.-H.; Wang, J.; Dohm, G.L.; Pories, W.J.; Mietus-Snyder, M.; Freishtat, R.J. Circulating adipocyte-derived exosomal micrornas associated with decreased insulin resistance after gastric bypass. Obes. Silver Spring Md. 2017, 25, 102–110. [Google Scholar] [CrossRef] [Green Version]
  110. Ortega, F.J.; Mercader, J.M.; Moreno-Navarrete, J.M.; Nonell, L.; Puigdecanet, E.; Rodriquez-Hermosa, J.I.; Rovira, O.; Xifra, G.; Guerra, E.; Moreno, M.; et al. Surgery-Induced Weight Loss Is Associated With the Downregulation of Genes Targeted by MicroRNAs in Adipose Tissue. J. Clin. Endocrinol. Metab. 2015, 100, E1467–E1476. [Google Scholar] [CrossRef] [Green Version]
  111. Kuryłowicz, A.; Wicik, Z.; Owczarz, M.; Jonas, M.I.; Kotlarek, M.; Świerniak, M.; Lisik, W.; Jonas, M.; Noszczyk, B.; Puzianowska-Kuźnicka, M. NGS Reveals Molecular Pathways Affected by Obesity and Weight Loss-Related Changes in MiRNA Levels in Adipose Tissue. Int. J. Mol. Sci. 2017, 19, 66. [Google Scholar] [CrossRef] [PubMed]
  112. Ortega, F.J.; Moreno, M.; Mercader, J.M.; Moreno-Navarrete, J.M.; Fuentes-Batllevell, N.; Sabater, M.; Ricart, W.; Fernández-Real, J.M. Inflammation Triggers Specific MicroRNA Profiles in Human Adipocytes and Macrophages and in Their Supernatants. Clin. Epigenetics 2015, 7, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ortega, F.J.; Mercader, J.M.; Catalán, V.; Moreno-Navarrete, J.M.; Pueyo, N.; Sabater, M.; Gómez-Ambrosi, J.; Anglada, R.; Fernández-Formoso, J.A.; Ricart, W.; et al. Targeting the Circulating MicroRNA Signature of Obesity. Clin. Chem. 2013, 59, 781–792. [Google Scholar] [CrossRef] [Green Version]
  114. Hulsmans, M.; Sinnaeve, P.; Van der Schueren, B.; Mathieu, C.; Janssens, S.; Holvoet, P. Decreased MiR-181a Expression in Monocytes of Obese Patients Is Associated with the Occurrence of Metabolic Syndrome and Coronary Artery Disease. J. Clin. Endocrinol. Metab. 2012, 97, E1213–E1218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Zhang, J.; Bowers, J.; Liu, L.; Wei, S.; Gowda, G.A.N.; Hammoud, Z.; Raftery, D. Esophageal Cancer Metabolite Biomarkers Detected by LC-MS and NMR Methods. PLoS ONE 2012, 7, e30181. [Google Scholar] [CrossRef] [PubMed]
  116. Lei, Z.; Huhman, D.V.; Sumner, L.W. Mass Spectrometry Strategies in Metabolomics. J. Biol. Chem. 2011, 286, 25435–25442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Vizioli, C.; Jaime-Lara, R.B.; Franks, A.T.; Ortiz, R.; Joseph, P.V. Untargeted Metabolomic Approach Shows No Differences in Subcutaneous Adipose Tissue of Diabetic and Non-Diabetic Subjects Undergoing Bariatric Surgery: An Exploratory Study. Biol. Res. Nurs. 2021, 23, 109–118. [Google Scholar] [CrossRef] [PubMed]
  118. Samczuk, P.; Hady, H.R.; Adamska-Patruno, E.; Citko, A.; Dadan, J.; Barbas, C.; Kretowski, A.; Ciborowski, M. In-and-Out Molecular Changes Linked to the Type 2 Diabetes Remission after Bariatric Surgery: An Influence of Gut Microbes on Mitochondria Metabolism. Int. J. Mol. Sci. 2018, 19, 3744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  119. Lopes, T.I.B.; Geloneze, B.; Pareja, J.C.; Calixto, A.R.; Ferreira, M.M.C.; Marsaioli, A.J. Blood Metabolome Changes Before and After Bariatric Surgery: A (1)H NMR-Based Clinical Investigation. Omics J. Integr. Biol. 2015, 19, 318–327. [Google Scholar] [CrossRef] [Green Version]
  120. Vaishya, S.; Sarwade, R.D.; Seshadri, V. MicroRNA, Proteins, and Metabolites as Novel Biomarkers for Prediabetes, Diabetes, and Related Complications. Front. Endocrinol. 2018, 9, 180. [Google Scholar] [CrossRef] [Green Version]
  121. Kwee, L.C.; Ilkayeva, O.; Muehlbauer, M.J.; Bihlmeyer, N.; Wolfe, B.; Purnell, J.Q.; Xavier Pi-Sunyer, F.; Chen, H.; Bahnson, J.; Newgard, C.B.; et al. Metabolites and Diabetes Remission after Weight Loss. Nutr. Diabetes 2021, 11, 10. [Google Scholar] [CrossRef] [PubMed]
  122. Luo, P.; Yu, H.; Zhao, X.; Bao, Y.; Hong, C.S.; Zhang, P.; Tu, Y.; Yin, P.; Gao, P.; Wei, L.; et al. Metabolomics Study of Roux-En-Y Gastric Bypass Surgery (RYGB) to Treat Type 2 Diabetes Patients Based on Ultraperformance Liquid Chromatography–Mass Spectrometry. Available online: https://pubs.acs.org/doi/pdf/10.1021/acs.jproteome.6b00022 (accessed on 31 January 2022).
  123. Zheng, X.; Chen, T.; Zhao, A.; Ning, Z.; Kuang, J.; Wang, S.; You, Y.; Bao, Y.; Ma, X.; Yu, H.; et al. Hyocholic Acid Species as Novel Biomarkers for Metabolic Disorders. Nat. Commun. 2021, 12, 1487. [Google Scholar] [CrossRef] [PubMed]
  124. Ha, J.; Jang, M.; Kwon, Y.; Park, Y.S.; Park, D.J.; Lee, J.-H.; Lee, H.-J.; Ha, T.K.; Kim, Y.-J.; Han, S.-M.; et al. Metabolomic Profiles Predict Diabetes Remission after Bariatric Surgery. J. Clin. Med. 2020, 9, 3897. [Google Scholar] [CrossRef] [PubMed]
  125. Narath, S.H.; Mautner, S.I.; Svehlikova, E.; Schultes, B.; Pieber, T.R.; Sinner, F.M.; Gander, E.; Libiseller, G.; Schimek, M.G.; Sourij, H.; et al. An Untargeted Metabolomics Approach to Characterize Short-Term and Long-Term Metabolic Changes after Bariatric Surgery. PLoS ONE 2016, 11, e0161425. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Sarosiek, K.; Pappan, K.L.; Gandhi, A.V.; Saxena, S.; Kang, C.Y.; McMahon, H.; Chipitsyna, G.I.; Tichansky, D.S.; Arafat, H.A. Conserved Metabolic Changes in Nondiabetic and Type 2 Diabetic Bariatric Surgery Patients: Global Metabolomic Pilot Study. J. Diabetes Res. 2016, 2016, 3467403. [Google Scholar] [CrossRef] [Green Version]
  127. Nemati, R.; Lu, J.; Tura, A.; Smith, G.; Murphy, R. Acute Changes in Non-Esterified Fatty Acids in Patients with Type 2 Diabetes Receiving Bariatric Surgery. Obes. Surg. 2017, 27, 649–656. [Google Scholar] [CrossRef]
Table 2. Differentially expressed miRNAs with their roles/targets after surgery.
Table 2. Differentially expressed miRNAs with their roles/targets after surgery.
StudyInterventionPopulationSourceRegulated miRNAsRole/Target
Sangiao-Alvarellos et al. (2020) [106]Bariatric surgery155 obese patients
47 CTRL
Serum/plasmamiR-122
miR-885-5 p
miR-192
Regulation of hepatic biochemical processes
Cereijo et al. (2020) [107]Bariatric surgery26 obese patientsSerummiR-92aGlucose homeostasis
Macartney-Coxson et al. (2020) [95]RYGB15 obese womenSAT
VAT
SAT:
miR-23a-5p, miR-27a-5p, miR-200c-3p, miR-223-3p, miR-1246, miR-24-2-5p, miR-128, miR-421, miR-3178, miR-1224-5p, miR-221, miR-22, miR-762 (↓)
VAT:
miR-223-3p (↓)
Inflammation, glucose uptake
Doyon et al. (2020) [95]LSG
RYGP
20 obese patientsSerumhas-miR-375
hsa-miR-126-3 p
hsa-miR-663 a
hsa-miR-30 c-5 p
hsa-miR-100-5 p
hsa-miR-27 a-3 p
hsa-miR-590-5 p
Fatty acid biosynthesis,
obesity,
adipocyte proliferation,
T2D
Bae et al. (2019) [94]LSG (n = 2) RYGB (n = 14)16 obese patients
18 CTRL
Serum exosomesmiR-424-5pBiomarker of weight loss
Liao et al. (2018) [94]LSG20 obese patients
8 CTRL
SAT
VAT
VAT:
miR-122 (↑)
PPAR-γ
Atkin et al. (2018) [97]RYGB29 T2D patientsPlasmamiR-7-5p, let-7f-5p, miR-15b-5p, miR-320c, miR-205-5p, miR-335-5p (↑)
let-7i-5p (↓)
Inflammation, adipocyte proliferation, ß-cell function, thyroid and pituitary function
Nunez-Lopez et al. (2017) [108]RYGB22 obese patientsPlasmamiR-15a (↑)
miR-34a, miR-122 (↓)
Biomarkers of weight loss/glucose metabolism
Hubal et al. (2017) [109]RYGB6 obese womenPlasma and serum adipocyte-derived exosomeslet-7a-5p, miR-16-5pInsulin signaling
Nardelli et al. (2017) [110]LAGB3 obese patients
2 CTRL
SATmiR-519d, miR-299-5p, miR-212, miR-671-3p (↓)
miR-370, miR-487a (↑)
PPAR-α (miR-519d)
Kurylowicz et al. (2016) [111]Bariatric surgery20 obese patients
7 CTRL
SATmiR-146b-3p, miR-146b-5p, miR-223-3p, miR-223-5p, miR-941 (↑)BMPR2, FOXP1, IGF1R
Ortega et al. (2015) [110]RYGB16 obese patientsSATmiR-155, miR-221, miR-130b (↓)Inflammation
Ortega et al. (2015) [112]RYGB9 obese womenSATmiR-19a/b, miR-146a/b, miR-155, miR-193b, miR-221, miR-222, miR-223, miR-376c, miR-411 (↓)Glucose uptake, lipid metabolism, energy homeostasis
Ortega et al. (2013) [113]RYGB6 obese patientsPlasmamiR-221 and miR-199a-3p (↑)
miR-16-1, miR-122, miR-140-5p, miR-193a-5p (↓)
Hulsmans et al. (2012) [114]RYGB9 obese patients
6 CTRL
PBMCmiR-181 (↑)TLR-NFkB pathway
CTRL, control subjects; RYGP, Roux-en-Y gastric bypass; PBMC, peripheral blood mononuclear cells; T2D, type 2 diabetes; SAT, subcutaneous adipose tissue; VAT, visceral adipose tissue; LSG, laparoscopic sleeve gastrectomy; BMPR2, bone morphogenic protein receptor 2; FOXP, forkhead box protein P1; IGF1R, insulin-like growth factor receptor 1; LAGB, laparoscopic adjustable gastric banding; ↑ indicates increased expression level; ↓ indicates reduced expression level.
Table 3. Metabolomic profiles of metabolic surgery.
Table 3. Metabolomic profiles of metabolic surgery.
StudyIntervention/Surgery TypePopulationFollow-Up
Period
Role/Target MetabolitesOutcomes
Zheng et al. (2021) [123]RYGB38 individuals with T2D12 monthsHyocholic acid (HCA)Serum HCA levels increased in the patients after RYGB (p < 0.05).
HCA species play critical roles in regulating glucose homeostasis and are protective against the development of T2DM in humans.
Ha et al. (2020) [124]SG
RYGB
24 individuals with T2D3 monthsAmino acid metabolites (AAMs):
L-DOPA and 3-HAA
The prognostic performances of L-DOPA (AUROC = 0.97; 95% CI, 0.91 to 1.00) and 3-HAA (AUROC = 0.86; 95% CI, 0.63 to 1.00).
AAMs are superior for predicting T2D remission postoperatively compared with existing prediction models.
Zhao et al. (2017) [125]RYGB419 individuals
38 obese individuals with T2D after RYGB
381 T2D and CTRL with overweight or obesity
12 months after RYGBTargeted: branched-chain amino acids (BCAAs), aromatic amino acids (AAAs), and acylcarnitinesHigher baseline stearic acid/palmitic acid ratio (S/P) was associated with a greater probability of diabetes remission after RYGB (odds ratio, 2.16 (95% CI 1.10–4.26))
and may serve as a diagnostic marker in preoperative patient assessment.
Narath et al. (2016) [125]RYGB44 obese, including 24 patients with T2D at baseline and 9 with diabetes remission12 monthsUntargeted: 36 metabolites that are known markers for cardiovascular riskTrimethylamine-N-oxide, alanine, phenylalanine, and indoxyl-sulfate
are four metabolites that significantly decline in patients with diabetes remission compared to patients without diabetes remission
(sarcosine: p = 0.031, pyroglutamic acid: p = 0.044, alanine: p = 0.005, and leucyl-proline: p = 0.049).
Lopes et al. (2016) [126]RYGB10 obese individuals with T2DM12 monthsUntargeted: metabolic and lipoprotein profiles and fatty acid profileGlucose levels decreased significantly after RYGB (from 159.8 ± 61.4 to 100.0 ± 22.9 mg/dL), demonstrating T2D remission (p < 0.05).
Lower levels of metabolic profile: lactate, alanine, and branched-chain amino acids
The VLDL, LDL, N-acetyl-glycoproteins, and unsaturated lipid levels decreased, but phosphatidylcholine and high-density lipoprotein increased after RYGB.
Sarosiek et al. (2016) [126]SG or full GB15 patients:
gastric sleeve with T2D (5), gastric sleeve
without T2D (5), and gastric bypass
with T2D (5)
28 daysNontargeted global metabolomics: glucose and lipid metabolism, histidine, and its metabolitesA total of 62 compounds were significantly different postsurgery compared to baseline (p < 0.05).
Significant improvement in fat mobilization and oxidation (p < 0.05)
and liver function (p < 0.05) after surgery.
Nemati et al. (2016) [127]LGB (Roux), LSG38 obese individuals with T2D:
GBP (11)
SG (14)
VLCD (13)
3 daysTargeted: NEFAs,
palmitic acid,
monounsaturated/polyunsaturated ratio (MUFA/PUFA),
linoleic acid, and
unsaturated/saturated fat
Linoleic acid was positively correlated with total insulin secretion (p = 0.03).
Glucose sensitivity correlated with palmitic acid (p = 0.01).
GBP, SG, and VLCD had similar acute effects that decreased palmitic acid (p < 0.05).
Several NEFAs correlated with beta-cell function parameters and HOMA-IR (p < 0.05).
Luo et al. (2016) [56]RYGB35 individuals with T2D, 23 remission and 12 nonremission patients with T2D, were measured at baseline and 6 and 12 months after RYGB.6 and 12 monthsUntargeted: free fatty acids (FFAs), acylcarnitines, amino acids, bile acids, and lipid speciesInsulin sensitivity, energy metabolism, and inflammation were related to metabolic alterations in free fatty acids (FFAs), acylcarnitines, amino acids, bile acids, and lipids species (p < 0.05). Baseline levels of tryptophan, bilirubin, and indoxyl sulfate measured before surgery as well as levels of FFA 16:0, FFA 18:3, FFA 17:2, and hippuric acid measured 6 months after surgery best predicted the suitability and efficacy of RYGB for patients with T2DM.
CTRL, control subjects; RYGP, Roux-en-Y gastric bypass; T2D, type 2 diabetes; SAT, subcutaneous adipose tissue; VAT, visceral adipose tissue; LSG, laparoscopic sleeve gastrectomy; LAGB, laparoscopic adjustable gastric banding; BPD, biliopancreatic diversion with duodenal switch; DJBL, duodenojejunal bypass liners; GB, gastric bypass; VLCD, very-low-calorie diet; HILIC, hydrophilic interaction liquid chromatography; UPLC–MS, ultra-performance liquid chromatography–mass spectrometry; UHPLC-MS/MS, ultrahigh-performance liquid chromatography–tandem mass spectrometry; 1H NMR, proton nuclear magnetic resonance; LC-MS/MS, Liquid Chromatography with tandem mass spectrometry; VLDL, very low density lipoprotein; LDL, low-density lipoprotein.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Fazliana, M.; Nor Hanipah, Z. Mechanisms and Outcomes of Metabolic Surgery in Type 2 Diabetes. Metabolites 2022, 12, 1134. https://doi.org/10.3390/metabo12111134

AMA Style

Fazliana M, Nor Hanipah Z. Mechanisms and Outcomes of Metabolic Surgery in Type 2 Diabetes. Metabolites. 2022; 12(11):1134. https://doi.org/10.3390/metabo12111134

Chicago/Turabian Style

Fazliana, Mansor, and Zubaidah Nor Hanipah. 2022. "Mechanisms and Outcomes of Metabolic Surgery in Type 2 Diabetes" Metabolites 12, no. 11: 1134. https://doi.org/10.3390/metabo12111134

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

Article Metrics

Back to TopTop