Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Aug 15, 2021; 12(8): 1187-1199
Published online Aug 15, 2021. doi: 10.4239/wjd.v12.i8.1187
Progress in treatment of type 2 diabetes by bariatric surgery
Zhang-Liu Jin, Wei Liu, Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan Province, China
ORCID number: Zhang-Liu Jin (0000-0001-7651-1454); Wei Liu (0000-0002-7035-108X).
Author contributions: Jin ZL and Liu W contributed to the conception and design of the study, preparation of the content, and writing of the paper.
Supported by National Natural Science Foundation of China, No. 81670481.
Conflict-of-interest statement: The authors declare having no conflict of interests for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Wei Liu, FACS, MD, Professor, Department of General Surgery & Department of Biliopancreatic and Metabolic Surgery, The Second Xiangya Hospital, Central South University, No. 139 Renmin Road, Changsha 410011, Hunan Province, China. liuweixy@csu.edu.cn
Received: February 17, 2021
Peer-review started: February 17, 2021
First decision: March 16, 2021
Revised: March 29, 2021
Accepted: July 6, 2021
Article in press: July 6, 2021
Published online: August 15, 2021

Abstract

The incidence of type 2 diabetes (T2D) is increasing at an alarming rate worldwide. Bariatric surgical procedures, such as the vertical sleeve gastrectomy and Roux-en-Y gastric bypass, are the most efficient approaches to obtain substantial and durable remission of T2D. The benefits of bariatric surgery are realized through the consequent increased satiety and alterations in gastrointestinal hormones, bile acids, and the intestinal microbiota. A comprehensive understanding of the mechanisms by which various bariatric surgical procedures exert their benefits on T2D could contribute to the design of better non-surgical treatments for T2D. In this review, we describe the classification and evolution of bariatric surgery and explore the multiple mechanisms underlying the effect of bariatric surgery on insulin resistance. Based upon our summarization of the current knowledge on the underlying mechanisms, we speculate that the gut might act as a new target for improving T2D. Our ultimate goal with this review is to provide a better understanding of T2D pathophysiology in order to support development of T2D treatments that are less invasive and more scalable.

Key Words: Obesity, Bariatric surgery, Type 2 diabetes, Insulin resistance, Bile acids, Microbiota

Core Tip: Bariatric surgery is an effective treatment for type 2 diabetes (T2D), providing long-term remission. Among these types of weight loss procedures, the vertical sleeve gastrectomy and Roux-en-Y gastric bypass are extensively performed worldwide, but in the United States especially. Through establishment of reduced caloric intake and alterations in gut hormones, bile acids, and intestinal microbes, these procedures also contribute to the resolution of T2D. Understanding the mechanisms underlying the effects of bariatric surgery on T2D might provide new targets for more effective non-surgical treatments, such as medications, for T2D.



INTRODUCTION

Obesity is a chronic disease, affecting individuals throughout the world and steadily escalating[1,2]. Indeed, the incidence of obesity has more than doubled from 1975 (at 5%) to 2014 (at 13%)[3,4]. According to this trend, the number of obese people may account for as many as one-fifth of the world’s population in the recent upcoming years. Obesity is an important risk for type 2 diabetes (T2D)[3], and as such the alarming rise in obesity has been accompanied by an expanding burden of T2D. At present, the prevalence of T2D stands at 9% worldwide, but it is predicted to reach approximately 12% by 2025 if trends continue[5], making it imperative to address the problem of obesity and T2D.

Although nonsurgical intervention can lead to weight reduction and concomitant improvement of T2D, the magnitude is modest and the benefits are not durable[6,7]. Bariatric surgeries, such as the vertical sleeve gastrectomy (VSG) and Roux-en-Y gastric bypass (RYGB) procedures, have proven to be the most efficient treatment for obesity and T2D[8-11]. Moreover, compared to the currently available nonsurgical interventions, bariatric surgery yields better outcomes for glycemic control and remission of T2D[7,8,12,13]. Yet, weight loss alone is not the key mechanism by which these surgical procedures imperatively improve T2D. Understanding the molecular underpinnings of these procedures is paramount, as they are now heavily employed in the treatment for diabetes.

The purpose of this review is to summarize the recent advances in this field and highlight the mechanisms by which bariatric surgeries benefit diabetic patients. Here, we describe contemporary bariatric surgery procedures and their beneficial effects on T2D, and discuss the implication of each on future research to improve the treatment of T2D, particularly for future nonsurgical approaches.

EVOLUTION OF BARIATRIC SURGERY

Despite bariatric surgery having been originally developed in the 1950s, the annual number of bariatric surgeries performed worldwide remains relatively low. In 2019, 833678 operations were reported (according to the International Federation for Surgery of Obesity Global Registry data)[14,15]. It is worth noting that this global number represents less than 1% of the overall eligible population with morbid obesity; as such, the potential for greater application of bariatric surgery is very large. During the period from 2010 to 2018, the proportion of RYGB procedures actually decreased (from 55% to 17%), as did that of the adjustable gastric banding (AGB) procedure (from 40% to 5%-10%)[14]. By comparison, the proportion of VSG procedures rose substantially (from 2% to 61%)[14]. The biliopancreatic diversion (BPD) procedure currently accounts for approximately 1% of the overall bariatric surgeries performed[16]. Thus, the most commonly performed bariatric surgery worldwide is VSG, followed by RYGB. In terms of the T2D remission outcome, it remains unknown whether any difference exists between the two most prevalent procedures and the underlying mechanisms of both procedures remain to be fully elucidated.

CLASSIFICATION OF BARIATRIC SURGERY PROCEDURES

In line with the direct surgical effects on food intake and/or nutrient absorption, bariatric surgical procedures are traditionally classified as restrictive, malabsorptive, or mixed operations. The restrictive-type techniques, including AGB, VSG, and vertical banded gastroplasty, physically decrease the size of the stomach in order to trigger earlier satiety during meals. The malabsorptive-type techniques, such as BPD, establish a bypass of the partial small bowel in order to induce bile acids (BAs) and food to be mixed in the distal 50-100 cm of the ileum, thereby prompting macronutrient malabsorption. The mixed-type procedures, such as RYGB, combine physical reduction of the stomach volume with a bowel bypass[17]. Due to the overall advancements in surgical techniques and greater knowledge gained through related clinical research, several novel bariatric surgical procedures have been introduced; these include the ileal interposition and duodenal-jejunal bypass. However, VSG and RYGB still account for the majority of weight-loss surgeries performed internationally. Given that any reconfiguration of the gastrointestinal tract involves a complex operation, classifying the modalities of such procedures into restrictive, malabsorption, or mixed is too simplistic; gaining a definitive understanding of the outcomes of the different bariatric operations will facilitate the most accurate application of each to achieve maximal benefit.

SURGICAL PROCEDURES
AGB

In AGB, a silicone ring is placed to encircle the upper region of the stomach and form a high-pressure zone above the gastric band, creating a small gastric pouch. The size of the gastric band itself can be adjusted by injection of sterile saline or air in a subcutaneous port. The goal of this approach is to decrease hunger and consequent caloric consumption[18,19].

Unfortunately, AGB has several risks and undesirable side effects; for example, it increases the risk of gastroesophageal reflux and is associated with a risk of band erosion[20]. Its benefits on weight loss are also relatively short-term. Thus, the prevalence of this technique has declined, both in the United States (where it enjoyed a particular popularity) and worldwide[21-24]. The rates of AGB impacts on weight loss and subsequent resolution of T2D remain appreciably below 50%, with 34% of patients experiencing excessive weight loss and 33% of patients achieving remission of T2D at 1 year[25] (Table 1).

Table 1 Randomized controlled trials of bariatric surgery vs medical treatment for type 2 diabetes.
Ref.
Intervention
Control
Follow-up in mo
Diabetes remission, surgery vs control (%)
Parikh et al[30], 2014VSGMedication665 vs 0
Simonson et al[25], 2019AGBMedication1233 vs 23
Cummings et al[28], 2016RYGBMedication1260 vs 6
Mingrone et al[29], 2015BPD; RYGBMedication2495 vs 0; 75 vs 0
RYGB

In RYGB, the stomach is transected along the lesser curvature to create a small gastric pouch (10-30 mL volume), which is anastomosed to the segment of the intestinal division to create an alimentary limb (75-100 cm length) following transection of the jejunum, without exposure to biliopancreatic secretion[26]. The stomach remnant is left in situ and in continuity with the duodenal and proximal jejunum, forming a biliopancreatic limb that contains only digestive enzymes and preventing direct contact with chyme. Following transection of the jejunum, the restoration of intestinal continuity occurs via a structuring of the proximal stump of the small bowel that is anastomosed to the alimentary limb to create a common limb, where the chyme is then allowed to contact the digestive enzymes and go through the processes of digestion and absorption.

The surgical realignment of the gastrointestinal tract represents not only a profound anatomic alteration but a physiological one as well, changing the profiles of BAs, gut hormones, and even the gut microbiota. Contingent upon the patient’s body mass index (BMI) and/or severity of T2D, the extension of the alimentary limb length can contribute to a better weight reduction and more notable remission of T2D[27], although it is also accompanied by an increased risk of nutrient deficiency and other complications, like urolithiasis. The effect of RYGB on T2D has been reported to have remission rates of 60%[28] and 75%[29] after 1 and 2 years, respectively (Table 1), which are similar to those of VSG[30].

VSG

In VSG, along the great curvature transecting 70%-80% of stomach, the remnant stomach remains as a tubular structure. During the meal, then, the tubular stomach is short of accommodative ability and enhances gastric emptying[31-34].

Over the last decade, VSG has been performed as a single-stage procedure. Given the maintenance of the native food passage and the reduction of gastric volume, VSG markedly diminishes the risk of nutrient deficiency. Its relative simplicity and good clinical outcomes have allowed VSG to surpass RYGB in recent years as the most prevalent weight-loss surgery in the United States and worldwide[35]. On account of the mechanical removal of the great curvature, gastric hormone levels become markedly altered, the most obvious of which being the secretion of ghrelin, a hunger hormone produced by the X/A-like cells in the fundus of the stomach. However, the levels of secreted peptide-YY (PYY), which controls the blood glucose concentration, become increased. The rate of T2D resolution after VSG has been reported as 65%[30] (Table 1).

BPD

BPD consists of two distinct stages, namely, creation of a tubular gastric pouch and an intestinal bypass. The VSG is conducted via removal of approximately 80% of the stomach, after which most of the small bowel is bypassed, leading to malabsorption. The duodenum is divided at the first portion, followed by transection of a segment of the distal ileum (at 250 cm proximal to the ileocecal valve) and anastomosis to the proximal end of the divided duodenum. Intestinal continuity is restored by the ileoileostomy, at 100 cm proximal to the ileocecal valve.

Unlike other procedures, BPD not only decreases caloric consumption but also leads to malabsorption of some nutrients and vitamins. Owing to the malabsorption resulting from the bypass of the major portion of the bowel, BPD is considered the most effective bariatric surgery for severe obesity and T2D. A randomized trial showed that BPD leads to a 70% excessive weight loss by the 2-year follow-up and more than 90% resolution rate of T2D compared to conventional medical therapy[29]. Nonetheless, because of the technical complexity and associated complications, such as nutritional deficiency, compared with RYGB and VSG, the use of BPD has been declining year by year[36]. At present, BPD is mainly applied to treat patients whose BMI is greater than 50 kg/m2 or who have refractory T2D[12,13,29]. The 2-year diabetes remission rate after BPD is 95%, representing the highest remission rate of all bariatric surgeries[29] (Table 1).

Control of T2D by bariatric surgery

Although bariatric surgery confers the potent ability to the remission of T2D, it is only indicated for obese diabetic patients (BMI > 35 kg/m2). The pathogenesis of T2D is mainly attributable to insulin resistance and impairment of β-cell function[37]. Plenty of studies have investigated the mechanisms by which bariatric procedures might result in T2D remission via increase of insulin sensitivity and/or β cell function[38-40]. The resolution of T2D after bariatric procedures was traditionally thought to be the result of decreased caloric consumption, weight loss, and nutritional malabsorption; however, the remission of diabetes occurs sooner than the surgery-induced weight loss[41,42]. Emerging evidence supports the hypothesis that bariatric procedures remit T2D via mechanisms that are independent of weight reduction[11,43-45]. Thus, investigations of the alterations in the gastrointestinal tract, either anatomical or physiological, will help to provide a better understanding of the effect of bariatric surgery on T2D[46-48].

Lipid metabolism

Multiple mechanisms result in defective insulin secretion and response in T2D, such as lipotoxicity, oxidative stress, and endoplasmic reticulum (ER) stress[49]. The majority of patients with severe obesity present some dyslipidemia, such as hyperlipemia and lipoprotein abnormality, which cause excessive fat deposition in important tissues and/or organs, including adipose tissue and the liver, muscle, and pancreas. The excess accumulation of fat in the body induces chronic tissue inflammation and consequent tissue insulin insensitivity, which is a well-described feature of obese diabetic patients[50]. Thereby, the mechanism that accelerates the improvement of hyperlipemia may improve tissues and/or organs functions and insulin sensitivity, and eventually leads to remission of T2D. Evidence is expanding that bariatric surgery produces marked improvement in dyslipidemia[51,52]. However, there are some differences in clinical effectiveness on dyslipidemia, possibly due to variance in each surgical anatomy. Taken together, the improvement of dyslipidemia metabolism after bariatric surgery may contribute to the attenuated insulin resistance and resolution of T2D, but the molecular mechanism warrants further investigation.

POTENTIAL MECHANISMS OF IMPROVEMENT OF T2D
Gastrointestinal hormones

Ghrelin: Ghrelin, an appetite-stimulating hormone mainly secreted from gastric X/A-like cells (PD/1 cells in human), regulates peripheral glucose homeostasis in a pattern that decreases glucose-stimulated insulin release[53,54] and promotes insulin resistance in muscle[55], in addition to increased food intake[56,57]. In particular for VSG, the removal of the gastric fundus markedly blocks the major source of ghrelin. Thus, inhibition of ghrelin production seems to be a plausible explanation for the observed improved glycemia. Accumulating evidence shows that circulating ghrelin is decreased after VSG, but decreased or not changed at all after RYGB[58-60] (Table 2). Nevertheless, in the VSG mouse model, the glycemic control outcome is similar between ghrelin-deficient and wild-type mice[58]. Altogether, the data suggest that decreased ghrelin cannot completely explain the observed improved glycemic homeostasis after VSG.

Table 2 Several factors contributing to improved type 2 diabetes after bariatric surgery.
Target
Major site of secretion (anatomical location)
VSG
RYGB
GhrelinX/A-like cells (stomach)DecreaseDecrease or no change
GLP1L cells (distal gut)IncreaseIncrease
PYYL cells (distal gut)IncreaseIncrease
Bile acidsHepatocytesIncreaseIncrease
FGF-15/19IleumIncreaseIncrease
MicrobiotaGutChangeChange
EnteroplasticityGutChangeChange

Glucagon-like peptide: Glucagon-like peptide (GLP-1), produced from intestinal L cells, activates insulin secretion and reduces glucagon release in a glucose-dependent manner in response to nutrient uptake in the gut[61]. Despite administration at a superphysiological dose, GLP-1 analog only partially improves the incretin effect in patients with T2D[62]. Following both VSG and RYGB, the postprandial level of GLP-1 is markedly increased, implying that GLP-1 acts as an incretin signal contributing to glycemic homeostasis[63,64] (Table 2). Mouse model studies comparing pharmacologic blockade of the GLP-1 receptor and bariatric surgeries, including both VSG and RYGB[65-68], have found similar responses to glycemic control in wild-type mice, suggesting that the action of endogenous GLP-1 does not account for the benefit of those bariatric procedures on T2D.

PYY: PYY, a 36-amino acid peptide, is produced by L cells and expressed in the pancreas and neurons in the central nervous system[69,70]. PYY was first reported in the early 1980s, when it was characterized as playing an important role in promoting gastric and pancreatic secretions and modulating the gastrointestinal tract function. In recent years, expanding evidence has signified that PYY can act on the Y2 receptor to regulate insulin sensitivity and glucose uptake. Moreover, PYY has also been shown to act on pancreatic islets to regulate insulin release. A lack of PYY in the gut and pancreas with reduced β cell mass resulted in insulin secretion disorder[71]. In contrast, overexpression of PYY in islets improved insulin secretion in response to glucose and increased β cell mass[72]. Of note, a large amount of evidence has emerged to indicate that the serum level of PYY is elevated following both VSG and RYGB[73] (Table 2). Therefore, PYY is likely to play a vital role in the bariatric surgery-induced remission of T2D.

BAs

In response to a meal, BAs are secreted by hepatocytes and released into the duodenum. Although it was shown over that past decade that BAs enable micelle formation and stimulate nutrient absorption and emulsification, it is only now becoming clear that BAs serve as signaling molecules in multiple biological responses, including glucose metabolism. Circulating BA levels become increased after bariatric procedures, including both VSG and RYGB, and have been implicated in the regulation of glucose homeostasis (as observed in rodent models and human patients)[74-77] (Table 2). These findings also represent a plausible explanation for the increase in BAs that occurs upon realignment of the gastrointestinal tract by the RYGB technique’s exposure of the ileum to chyme that had avoided the digestion process thus far. In line with this notion, when high-fat diet-induced obese rodents were subjected to exposure of the ileum to BAs, they achieved a level of glucose improvement that was identical to that observed in T2D patients after RYGB, suggesting that BAs may play a pivotal role in the effect of RYGB on glycemic control[78,79]. Intriguingly, an increase in circulating BAs has been found in rodents and humans following VSG, further suggesting that BA profile changes likely represent a physiological modality for T2D remission via bariatric surgery.

The increased serum BAs contribute to the improvement of impaired glucose homeostasis mainly through two corresponding signaling pathways, namely, those involving the farnesoid-X receptor (FXR) and the transmembrane G protein-coupled receptor 5 (TGR5)[77,80-83]. Overexpression of FXR in db/db mice improved metabolic disorders, indicating that FXR signaling may serve as a therapeutic target for maintaining metabolic homeostasis[84,85]. BAs function as a ligand for FXR, which can underlie the observed improvement of glucose metabolism via the FXR-related pathway. Compared with wild-type mice, mice that are deficient in FXR forfeit the ability to maintain glucose equilibrium following VSG[86]. Furthermore, it was shown that the increase in fibroblast growth factor-15 (FGF19 in human), a downstream effector of the BAs-FXR pathway, after bariatric surgery contributes to hepatic glycogen synthesis and reduces glycemia[87-89].

In contrast to FXR, TGR5, a G protein-coupled receptor, is expressed in multiple tissues, including the intestine, skeletal muscle, liver, and adipose tissue. The increased BAs after VSG confer the ability to remit insulin resistance in a TGR5-activation manner[90,91]. Compared with results from studies in wild-type mice, the improvement of T2D in TGR5-/- mice was severely blunted, suggesting that TGR5 might be essential for glycemic control after VSG[90].

Gut microbiota

Over the past years, the association between the gut microbiota and altered metabolic processes has been recognized in both rodents and humans[92-96]. In addition, a large bacterial population shift has been observed following the bariatric procedures, including VSG and RYGB[97-100] (Table 2). Compared with results in sham operation models, the relative abundance of Gammaproteobacteria (Escherichia) and Verrucomicrobia (Akkermansia) is rapidly and sustainedly enhanced after RYGB[101]. In concert with this, the shift of the gut microbiota from the RYGB group to germ-free mice leads to a weight reduction, implying that gut microbiota contributes to weight loss[101]. Moreover, allogenic fecal microbiota transplantation using metabolic syndrome donors led to impairment in insulin sensitivity for the metabolic syndrome recipients compared with using post-RYGB donors[102]; this finding indicates that the alteration of intestinal microbes after RYGB can exert a positive effect by improving insulin resistance.

Enteroplasticity

In response to internal and external environmental stimuli, the processes of proliferation, migration, death, and differentiation of epithelial cells take place in the human small intestine[103]. Thereby, enteroplasticity or intestinal adaptation, including morphological and nervous system alterations, refers to the capacity to adapt functionally, as occurs in diabetes, aging, and so forth[104]. Western diet might contribute to alterations of enteroplasticity that result in metabolic derangement; hence, it is worth exploring whether bariatric surgery might lead to changes in enteroplasticity. Increasing evidence suggests that several bariatric surgical procedures trigger changes of enteroplasticity[105-107] (Table 2). The intestinal morphology, including width and cellular proliferation, was found to be enhanced in the alimentary and common limbs in an RYGB rat model[108,109], and the intestinal villus height and surface area were found to be reduced in mice after VSG[110]. Additionally, some studies indicated that the hepatoportal sensor pathway plays an important role in glycemic control after RYGB, unlike findings after AGB[111]. Altogether, these data signify marked changes in enteroplasticity occurring after bariatric surgery.

CONCLUSION

The escalating pandemic of T2D continues to be a worldwide problem. Through its impressive efficacy, bariatric procedures are still the most effective and efficient durable therapy for the improvement of T2D in severe obesity. Moreover, the outcomes of weight-loss surgery provide novel scientific clues and a theoretical foundation for the gut’s potential to act as a therapeutic target for remission and countering of insulin resistance. Taken together, although great progress has been made in our understanding of the mechanisms by which bariatric surgery may improve T2D, the discrepancy of certain evidence is undetermined and requires further research efforts.

ACKNOWLEDGEMENTS

Dr. Jin ZL wants to thank his wife, Mrs. Gao M, for her support.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Endocrinology and metabolism

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Vazquez-Jimenez JG S-Editor: Gao CC L-Editor: Wang TQ P-Editor: Wang LYT

References
1.  Cho NH, Shaw JE, Karuranga S, Huang Y, da Rocha Fernandes JD, Ohlrogge AW, Malanda B. IDF Diabetes Atlas: Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract. 2018;138:271-281.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3709]  [Cited by in F6Publishing: 3757]  [Article Influence: 626.2]  [Reference Citation Analysis (0)]
2.  Yang W, Lu J, Weng J, Jia W, Ji L, Xiao J, Shan Z, Liu J, Tian H, Ji Q, Zhu D, Ge J, Lin L, Chen L, Guo X, Zhao Z, Li Q, Zhou Z, Shan G, He J;  China National Diabetes and Metabolic Disorders Study Group. Prevalence of diabetes among men and women in China. N Engl J Med. 2010;362:1090-1101.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2186]  [Cited by in F6Publishing: 2231]  [Article Influence: 159.4]  [Reference Citation Analysis (2)]
3.  GBD 2015 Obesity Collaborators, Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, Marczak L, Mokdad AH, Moradi-Lakeh M, Naghavi M, Salama JS, Vos T, Abate KH, Abbafati C, Ahmed MB, Al-Aly Z, Alkerwi A, Al-Raddadi R, Amare AT, Amberbir A, Amegah AK, Amini E, Amrock SM, Anjana RM, Ärnlöv J, Asayesh H, Banerjee A, Barac A, Baye E, Bennett DA, Beyene AS, Biadgilign S, Biryukov S, Bjertness E, Boneya DJ, Campos-Nonato I, Carrero JJ, Cecilio P, Cercy K, Ciobanu LG, Cornaby L, Damtew SA, Dandona L, Dandona R, Dharmaratne SD, Duncan BB, Eshrati B, Esteghamati A, Feigin VL, Fernandes JC, Fürst T, Gebrehiwot TT, Gold A, Gona PN, Goto A, Habtewold TD, Hadush KT, Hafezi-Nejad N, Hay SI, Horino M, Islami F, Kamal R, Kasaeian A, Katikireddi SV, Kengne AP, Kesavachandran CN, Khader YS, Khang YH, Khubchandani J, Kim D, Kim YJ, Kinfu Y, Kosen S, Ku T, Defo BK, Kumar GA, Larson HJ, Leinsalu M, Liang X, Lim SS, Liu P, Lopez AD, Lozano R, Majeed A, Malekzadeh R, Malta DC, Mazidi M, McAlinden C, McGarvey ST, Mengistu DT, Mensah GA, Mensink GBM, Mezgebe HB, Mirrakhimov EM, Mueller UO, Noubiap JJ, Obermeyer CM, Ogbo FA, Owolabi MO, Patton GC, Pourmalek F, Qorbani M, Rafay A, Rai RK, Ranabhat CL, Reinig N, Safiri S, Salomon JA, Sanabria JR, Santos IS, Sartorius B, Sawhney M, Schmidhuber J, Schutte AE, Schmidt MI, Sepanlou SG, Shamsizadeh M, Sheikhbahaei S, Shin MJ, Shiri R, Shiue I, Roba HS, Silva DAS, Silverberg JI, Singh JA, Stranges S, Swaminathan S, Tabarés-Seisdedos R, Tadese F, Tedla BA, Tegegne BS, Terkawi AS, Thakur JS, Tonelli M, Topor-Madry R, Tyrovolas S, Ukwaja KN, Uthman OA, Vaezghasemi M, Vasankari T, Vlassov VV, Vollset SE, Weiderpass E, Werdecker A, Wesana J, Westerman R, Yano Y, Yonemoto N, Yonga G, Zaidi Z, Zenebe ZM, Zipkin B, Murray CJL. Health Effects of Overweight and Obesity in 195 Countries over 25 Years. N Engl J Med. 2017;377:13-27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3869]  [Cited by in F6Publishing: 4181]  [Article Influence: 597.3]  [Reference Citation Analysis (2)]
4.  NCD Risk Factor Collaboration (NCD-RisC). Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19·2 million participants. Lancet. 2016;387:1377-1396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3351]  [Cited by in F6Publishing: 3152]  [Article Influence: 394.0]  [Reference Citation Analysis (0)]
5.  NCD Risk Factor Collaboration (NCD-RisC). Worldwide trends in diabetes since 1980: a pooled analysis of 751 population-based studies with 4.4 million participants. Lancet. 2016;387:1513-1530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2146]  [Cited by in F6Publishing: 2290]  [Article Influence: 286.3]  [Reference Citation Analysis (0)]
6.  Tuah NA, Amiel C, Qureshi S, Car J, Kaur B, Majeed A. Transtheoretical model for dietary and physical exercise modification in weight loss management for overweight and obese adults. Cochrane Database Syst Rev. 2011;CD008066.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
7.  Gloy VL, Briel M, Bhatt DL, Kashyap SR, Schauer PR, Mingrone G, Bucher HC, Nordmann AJ. Bariatric surgery vs non-surgical treatment for obesity: a systematic review and meta-analysis of randomised controlled trials. BMJ. 2013;347:f5934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 811]  [Cited by in F6Publishing: 846]  [Article Influence: 76.9]  [Reference Citation Analysis (0)]
8.  Yu J, Zhou X, Li L, Li S, Tan J, Li Y, Sun X. The long-term effects of bariatric surgery for type 2 diabetes: systematic review and meta-analysis of randomized and non-randomized evidence. Obes Surg. 2015;25:143-158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 116]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
9.  Sha Y, Huang X, Ke P, Wang B, Yuan H, Yuan W, Wang Y, Zhu X, Yan Y. Laparoscopic Roux-en-Y Gastric Bypass Versus Sleeve Gastrectomy for Type 2 Diabetes Mellitus in Nonseverely Obese Patients: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Obes Surg. 2020;30:1660-1670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 12]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
10.  Capoccia D, Coccia F, Guarisco G, Testa M, Rendina R, Abbatini F, Silecchia G, Leonetti F. Long-term Metabolic Effects of Laparoscopic Sleeve Gastrectomy. Obes Surg. 2018;28:2289-2296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
11.  Rubino F, Nathan DM, Eckel RH, Schauer PR, Alberti KG, Zimmet PZ, Del Prato S, Ji L, Sadikot SM, Herman WH, Amiel SA, Kaplan LM, Taroncher-Oldenburg G, Cummings DE;  Delegates of the 2nd Diabetes Surgery Summit. Metabolic Surgery in the Treatment Algorithm for Type 2 Diabetes: A Joint Statement by International Diabetes Organizations. Diabetes Care. 2016;39:861-877.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 603]  [Cited by in F6Publishing: 539]  [Article Influence: 67.4]  [Reference Citation Analysis (0)]
12.  Buchwald H, Avidor Y, Braunwald E, Jensen MD, Pories W, Fahrbach K, Schoelles K. Bariatric surgery: a systematic review and meta-analysis. JAMA. 2004;292:1724-1737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5073]  [Cited by in F6Publishing: 4544]  [Article Influence: 227.2]  [Reference Citation Analysis (0)]
13.  Chang SH, Stoll CR, Song J, Varela JE, Eagon CJ, Colditz GA. The effectiveness and risks of bariatric surgery: an updated systematic review and meta-analysis, 2003-2012. JAMA Surg. 2014;149:275-287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1066]  [Cited by in F6Publishing: 1077]  [Article Influence: 107.7]  [Reference Citation Analysis (1)]
14.  Docherty NG, le Roux CW. Bariatric surgery for the treatment of chronic kidney disease in obesity and type 2 diabetes mellitus. Nat Rev Nephrol. 2020;16:709-720.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 32]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
15.  Welbourn R, Pournaras DJ, Dixon J, Higa K, Kinsman R, Ottosson J, Ramos A, van Wagensveld B, Walton P, Weiner R, Zundel N. Bariatric Surgery Worldwide: Baseline Demographic Description and One-Year Outcomes from the Second IFSO Global Registry Report 2013-2015. Obes Surg. 2018;28:313-322.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 86]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
16.  DeMaria EJ, Pate V, Warthen M, Winegar DA. Baseline data from American Society for Metabolic and Bariatric Surgery-designated Bariatric Surgery Centers of Excellence using the Bariatric Outcomes Longitudinal Database. Surg Obes Relat Dis. 2010;6:347-355.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 277]  [Cited by in F6Publishing: 289]  [Article Influence: 20.6]  [Reference Citation Analysis (0)]
17.  Frühbeck G. Bariatric and metabolic surgery: a shift in eligibility and success criteria. Nat Rev Endocrinol. 2015;11:465-477.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 153]  [Article Influence: 17.0]  [Reference Citation Analysis (0)]
18.  Dixon JB, O'Brien PE, Playfair J, Chapman L, Schachter LM, Skinner S, Proietto J, Bailey M, Anderson M. Adjustable gastric banding and conventional therapy for type 2 diabetes: a randomized controlled trial. JAMA. 2008;299:316-323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 945]  [Cited by in F6Publishing: 900]  [Article Influence: 56.3]  [Reference Citation Analysis (0)]
19.  O'Brien PE, Dixon JB, Laurie C, Skinner S, Proietto J, McNeil J, Strauss B, Marks S, Schachter L, Chapman L, Anderson M. Treatment of mild to moderate obesity with laparoscopic adjustable gastric banding or an intensive medical program: a randomized trial. Ann Intern Med. 2006;144:625-633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 387]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
20.  Aarts EO, Dogan K, Koehestanie P, Aufenacker TJ, Janssen IM, Berends FJ. Long-term results after laparoscopic adjustable gastric banding: a mean fourteen year follow-up study. Surg Obes Relat Dis. 2014;10:633-640.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 65]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
21.  English WJ, DeMaria EJ, Hutter MM, Kothari SN, Mattar SG, Brethauer SA, Morton JM. American Society for Metabolic and Bariatric Surgery 2018 estimate of metabolic and bariatric procedures performed in the United States. Surg Obes Relat Dis. 2020;16:457-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 153]  [Cited by in F6Publishing: 193]  [Article Influence: 48.3]  [Reference Citation Analysis (0)]
22.  Angrisani L, Santonicola A, Iovino P, Formisano G, Buchwald H, Scopinaro N. Bariatric Surgery Worldwide 2013. Obes Surg. 2015;25:1822-1832.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1090]  [Cited by in F6Publishing: 1053]  [Article Influence: 117.0]  [Reference Citation Analysis (1)]
23.  Altieri MS, Yang J, Telem DA, Meng Z, Frenkel C, Halbert C, Talamini M, Pryor AD. Lap band outcomes from 19,221 patients across centers and over a decade within the state of New York. Surg Endosc. 2016;30:1725-1732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 57]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
24.  Kindel T, Martin E, Hungness E, Nagle A. High failure rate of the laparoscopic-adjustable gastric band as a primary bariatric procedure. Surg Obes Relat Dis. 2014;10:1070-1075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
25.  Simonson DC, Vernon A, Foster K, Halperin F, Patti ME, Goldfine AB. Adjustable gastric band surgery or medical management in patients with type 2 diabetes and obesity: three-year results of a randomized trial. Surg Obes Relat Dis. 2019;15:2052-2059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
26.  Pucci A, Batterham RL. Mechanisms underlying the weight loss effects of RYGB and SG: similar, yet different. J Endocrinol Invest. 2019;42:117-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 116]  [Article Influence: 23.2]  [Reference Citation Analysis (0)]
27.  Bal B, Koch TR, Finelli FC, Sarr MG. Managing medical and surgical disorders after divided Roux-en-Y gastric bypass surgery. Nat Rev Gastroenterol Hepatol. 2010;7:320-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
28.  Cummings DE, Arterburn DE, Westbrook EO, Kuzma JN, Stewart SD, Chan CP, Bock SN, Landers JT, Kratz M, Foster-Schubert KE, Flum DR. Gastric bypass surgery vs intensive lifestyle and medical intervention for type 2 diabetes: the CROSSROADS randomised controlled trial. Diabetologia. 2016;59:945-953.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 199]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
29.  Mingrone G, Panunzi S, De Gaetano A, Guidone C, Iaconelli A, Nanni G, Castagneto M, Bornstein S, Rubino F. Bariatric-metabolic surgery vs conventional medical treatment in obese patients with type 2 diabetes: 5 year follow-up of an open-label, single-centre, randomised controlled trial. Lancet. 2015;386:964-973.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 852]  [Cited by in F6Publishing: 803]  [Article Influence: 89.2]  [Reference Citation Analysis (0)]
30.  Parikh M, Chung M, Sheth S, McMacken M, Zahra T, Saunders JK, Ude-Welcome A, Dunn V, Ogedegbe G, Schmidt AM, Pachter HL. Randomized pilot trial of bariatric surgery vs intensive medical weight management on diabetes remission in type 2 diabetic patients who do NOT meet NIH criteria for surgery and the role of soluble RAGE as a novel biomarker of success. Ann Surg. 2014;260:617-22; discussion 622.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 81]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
31.  Hofsø D, Fatima F, Borgeraas H, Birkeland KI, Gulseth HL, Hertel JK, Johnson LK, Lindberg M, Nordstrand N, Cvancarova Småstuen M, Stefanovski D, Svanevik M, Gretland Valderhaug T, Sandbu R, Hjelmesæth J. Gastric bypass vs sleeve gastrectomy in patients with type 2 diabetes (Oseberg): a single-centre, triple-blind, randomised controlled trial. Lancet Diabetes Endocrinol. 2019;7:912-924.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 126]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
32.  Miras AD, le Roux CW. Mechanisms underlying weight loss after bariatric surgery. Nat Rev Gastroenterol Hepatol. 2013;10:575-584.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 215]  [Cited by in F6Publishing: 221]  [Article Influence: 20.1]  [Reference Citation Analysis (0)]
33.  Madsbad S, Dirksen C, Holst JJ. Mechanisms of changes in glucose metabolism and bodyweight after bariatric surgery. Lancet Diabetes Endocrinol. 2014;2:152-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 211]  [Cited by in F6Publishing: 193]  [Article Influence: 19.3]  [Reference Citation Analysis (0)]
34.  Arble DM, Sandoval DA, Seeley RJ. Mechanisms underlying weight loss and metabolic improvements in rodent models of bariatric surgery. Diabetologia. 2015;58:211-220.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
35.  Khorgami Z, Shoar S, Andalib A, Aminian A, Brethauer SA, Schauer PR. Trends in utilization of bariatric surgery, 2010-2014: sleeve gastrectomy dominates. Surg Obes Relat Dis. 2017;13:774-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 150]  [Cited by in F6Publishing: 156]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
36.  Ponce J, Nguyen NT, Hutter M, Sudan R, Morton JM. American Society for Metabolic and Bariatric Surgery estimation of bariatric surgery procedures in the United States, 2011-2014. Surg Obes Relat Dis. 2015;11:1199-1200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 145]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
37.  Accili D. Lilly lecture 2003: the struggle for mastery in insulin action: from triumvirate to republic. Diabetes. 2004;53:1633-1642.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 146]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
38.  Rosen CJ, Ingelfinger JR. Bariatric Surgery and Restoration of Insulin Sensitivity - It's Weight Loss. N Engl J Med. 2020;383:777-778.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
39.  Villarreal-Calderón JR, Cuéllar RX, Ramos-González MR, Rubio-Infante N, Castillo EC, Elizondo-Montemayor L, García-Rivas G. Interplay between the Adaptive Immune System and Insulin Resistance in Weight Loss Induced by Bariatric Surgery. Oxid Med Cell Longev. 2019;2019:3940739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
40.  Mingrone G, Cummings DE. Changes of insulin sensitivity and secretion after bariatric/metabolic surgery. Surg Obes Relat Dis. 2016;12:1199-1205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 37]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
41.  Rubino F, Marescaux J. Effect of duodenal-jejunal exclusion in a non-obese animal model of type 2 diabetes: a new perspective for an old disease. Ann Surg. 2004;239:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 505]  [Cited by in F6Publishing: 448]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
42.  Jackson HT, Anekwe C, Chang J, Haskins IN, Stanford FC. The Role of Bariatric Surgery on Diabetes and Diabetic Care Compliance. Curr Diab Rep. 2019;19:125.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
43.  Strain GW, Gagner M, Inabnet WB, Dakin G, Pomp A. Comparison of effects of gastric bypass and biliopancreatic diversion with duodenal switch on weight loss and body composition 1-2 years after surgery. Surg Obes Relat Dis. 2007;3:31-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 28]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
44.  Camerini G, Marinari GM, Scopinaro N. A new approach to the fashioning of the gastroenteroanastomosis in laparoscopic standard biliopancreatic diversion. Surg Laparosc Endosc Percutan Tech. 2003;13:165-167.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
45.  Lee JH, Jaung R, Beban G, Evennett N, Cundy T. Insulin use and new diabetes after acceptance for bariatric surgery: comparison of outcomes after completion of surgery or withdrawal from the program. BMJ Open Diabetes Res Care. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
46.  Heneghan HM, Nissen S, Schauer PR. Gastrointestinal surgery for obesity and diabetes: weight loss and control of hyperglycemia. Curr Atheroscler Rep. 2012;14:579-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 27]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  le Roux CW, Aylwin SJ, Batterham RL, Borg CM, Coyle F, Prasad V, Shurey S, Ghatei MA, Patel AG, Bloom SR. Gut hormone profiles following bariatric surgery favor an anorectic state, facilitate weight loss, and improve metabolic parameters. Ann Surg. 2006;243:108-114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 742]  [Cited by in F6Publishing: 710]  [Article Influence: 39.4]  [Reference Citation Analysis (0)]
48.  Mabey JG, Chaston JM, Castro DG, Adams TD, Hunt SC, Davidson LE. Gut microbiota differs a decade after bariatric surgery relative to a nonsurgical comparison group. Surg Obes Relat Dis. 2020;16:1304-1311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
49.  Donath MY. Targeting inflammation in the treatment of type 2 diabetes: time to start. Nat Rev Drug Discov. 2014;13:465-476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 478]  [Cited by in F6Publishing: 499]  [Article Influence: 49.9]  [Reference Citation Analysis (0)]
50.  Kojta I, Chacińska M, Błachnio-Zabielska A. Obesity, Bioactive Lipids, and Adipose Tissue Inflammation in Insulin Resistance. Nutrients. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 164]  [Article Influence: 41.0]  [Reference Citation Analysis (0)]
51.  Heffron SP, Parikh A, Volodarskiy A, Ren-Fielding C, Schwartzbard A, Nicholson J, Bangalore S. Changes in Lipid Profile of Obese Patients Following Contemporary Bariatric Surgery: A Meta-Analysis. Am J Med. 2016;129:952-959.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 84]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
52.  Cunha FM, Oliveira J, Preto J, Saavedra A, Costa MM, Magalhães D, Lau E, Bettencourt-Silva R, Freitas P, Varela A, Carvalho D. The Effect of Bariatric Surgery Type on Lipid Profile: An Age, Sex, Body Mass Index and Excess Weight Loss Matched Study. Obes Surg. 2016;26:1041-1047.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 38]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
53.  Reimer MK, Pacini G, Ahrén B. Dose-dependent inhibition by ghrelin of insulin secretion in the mouse. Endocrinology. 2003;144:916-921.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 223]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
54.  Tong J, Prigeon RL, Davis HW, Bidlingmaier M, Kahn SE, Cummings DE, Tschöp MH, D'Alessio D. Ghrelin suppresses glucose-stimulated insulin secretion and deteriorates glucose tolerance in healthy humans. Diabetes. 2010;59:2145-2151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 227]  [Cited by in F6Publishing: 235]  [Article Influence: 16.8]  [Reference Citation Analysis (0)]
55.  Vestergaard ET, Djurhuus CB, Gjedsted J, Nielsen S, Møller N, Holst JJ, Jørgensen JO, Schmitz O. Acute effects of ghrelin administration on glucose and lipid metabolism. J Clin Endocrinol Metab. 2008;93:438-444.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 70]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
56.  Wren AM, Seal LJ, Cohen MA, Brynes AE, Frost GS, Murphy KG, Dhillo WS, Ghatei MA, Bloom SR. Ghrelin enhances appetite and increases food intake in humans. J Clin Endocrinol Metab. 2001;86:5992.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1222]  [Cited by in F6Publishing: 1242]  [Article Influence: 54.0]  [Reference Citation Analysis (0)]
57.  Tschöp M, Smiley DL, Heiman ML. Ghrelin induces adiposity in rodents. Nature. 2000;407:908-913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2834]  [Cited by in F6Publishing: 2708]  [Article Influence: 112.8]  [Reference Citation Analysis (0)]
58.  Chambers AP, Kirchner H, Wilson-Perez HE, Willency JA, Hale JE, Gaylinn BD, Thorner MO, Pfluger PT, Gutierrez JA, Tschöp MH, Sandoval DA, Seeley RJ. The effects of vertical sleeve gastrectomy in rodents are ghrelin independent. Gastroenterology. 2013;144:50-52.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 121]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
59.  Cummings DE, Weigle DS, Frayo RS, Breen PA, Ma MK, Dellinger EP, Purnell JQ. Plasma ghrelin levels after diet-induced weight loss or gastric bypass surgery. N Engl J Med. 2002;346:1623-1630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1709]  [Cited by in F6Publishing: 1493]  [Article Influence: 67.9]  [Reference Citation Analysis (0)]
60.  Holdstock C, Engström BE, Ohrvall M, Lind L, Sundbom M, Karlsson FA. Ghrelin and adipose tissue regulatory peptides: effect of gastric bypass surgery in obese humans. J Clin Endocrinol Metab. 2003;88:3177-3183.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 231]  [Cited by in F6Publishing: 236]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
61.  Drucker DJ. The role of gut hormones in glucose homeostasis. J Clin Invest. 2007;117:24-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 439]  [Cited by in F6Publishing: 424]  [Article Influence: 24.9]  [Reference Citation Analysis (0)]
62.  Vilsbøll T, Christensen M, Junker AE, Knop FK, Gluud LL. Effects of glucagon-like peptide-1 receptor agonists on weight loss: systematic review and meta-analyses of randomised controlled trials. BMJ. 2012;344:d7771.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 594]  [Cited by in F6Publishing: 589]  [Article Influence: 49.1]  [Reference Citation Analysis (0)]
63.  Umeda LM, Silva EA, Carneiro G, Arasaki CH, Geloneze B, Zanella MT. Early improvement in glycemic control after bariatric surgery and its relationships with insulin, GLP-1, and glucagon secretion in type 2 diabetic patients. Obes Surg. 2011;21:896-901.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 91]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
64.  Baggio LL, Drucker DJ. Biology of incretins: GLP-1 and GIP. Gastroenterology. 2007;132:2131-2157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2461]  [Cited by in F6Publishing: 2465]  [Article Influence: 145.0]  [Reference Citation Analysis (0)]
65.  Shah M, Law JH, Micheletto F, Sathananthan M, Dalla Man C, Cobelli C, Rizza RA, Camilleri M, Zinsmeister AR, Vella A. Contribution of endogenous glucagon-like peptide 1 to glucose metabolism after Roux-en-Y gastric bypass. Diabetes. 2014;63:483-493.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 112]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
66.  Salehi M, Gastaldelli A, D'Alessio DA. Blockade of glucagon-like peptide 1 receptor corrects postprandial hypoglycemia after gastric bypass. Gastroenterology. 2014;146:669-680.e2.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 177]  [Article Influence: 17.7]  [Reference Citation Analysis (0)]
67.  Jiménez A, Mari A, Casamitjana R, Lacy A, Ferrannini E, Vidal J. GLP-1 and glucose tolerance after sleeve gastrectomy in morbidly obese subjects with type 2 diabetes. Diabetes. 2014;63:3372-3377.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 69]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
68.  Jiménez A, Casamitjana R, Viaplana-Masclans J, Lacy A, Vidal J. GLP-1 action and glucose tolerance in subjects with remission of type 2 diabetes after gastric bypass surgery. Diabetes Care. 2013;36:2062-2069.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 90]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
69.  Price SL, Bloom SR. Protein PYY and its role in metabolism. Front Horm Res. 2014;42:147-154.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
70.  Zac-Varghese S, De Silva A, Bloom SR. Translational studies on PYY as a novel target in obesity. Curr Opin Pharmacol. 2011;11:582-585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
71.  Sam AH, Gunner DJ, King A, Persaud SJ, Brooks L, Hostomska K, Ford HE, Liu B, Ghatei MA, Bloom SR, Bewick GA. Selective ablation of peptide YY cells in adult mice reveals their role in beta cell survival. Gastroenterology. 2012;143:459-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 55]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
72.  Shi YC, Loh K, Bensellam M, Lee K, Zhai L, Lau J, Cantley J, Luzuriaga J, Laybutt DR, Herzog H. Pancreatic PYY Is Critical in the Control of Insulin Secretion and Glucose Homeostasis in Female Mice. Endocrinology. 2015;156:3122-3136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 33]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
73.  Farey JE, Preda TC, Fisher OM, Levert-Mignon AJ, Stewart RL, Karsten E, Herbert BR, Swarbrick MM, Lord RV. Effect of Laparoscopic Sleeve Gastrectomy on Fasting Gastrointestinal, Pancreatic, and Adipose-Derived Hormones and on Non-Esterified Fatty Acids. Obes Surg. 2017;27:399-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 22]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
74.  Browning MG, Pessoa BM, Khoraki J, Campos GM. Changes in Bile Acid Metabolism, Transport, and Signaling as Central Drivers for Metabolic Improvements After Bariatric Surgery. Curr Obes Rep. 2019;8:175-184.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 30]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
75.  Jahansouz C, Xu H, Hertzel AV, Serrot FJ, Kvalheim N, Cole A, Abraham A, Luthra G, Ewing K, Leslie DB, Bernlohr DA, Ikramuddin S. Bile Acids Increase Independently From Hypocaloric Restriction After Bariatric Surgery. Ann Surg. 2016;264:1022-1028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 59]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
76.  Khan FH, Shaw L, Zhang W, Salazar Gonzalez RM, Mowery S, Oehrle M, Zhao X, Jenkins T, Setchell KD, Inge TH, Kohli R. Fibroblast growth factor 21 correlates with weight loss after vertical sleeve gastrectomy in adolescents. Obesity (Silver Spring). 2016;24:2377-2383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 26]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
77.  Steinert RE, Peterli R, Keller S, Meyer-Gerspach AC, Drewe J, Peters T, Beglinger C. Bile acids and gut peptide secretion after bariatric surgery: a 1-year prospective randomized pilot trial. Obesity (Silver Spring). 2013;21:E660-E668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 145]  [Article Influence: 13.2]  [Reference Citation Analysis (0)]
78.  Albaugh VL, Banan B, Antoun J, Xiong Y, Guo Y, Ping J, Alikhan M, Clements BA, Abumrad NN, Flynn CR. Role of Bile Acids and GLP-1 in Mediating the Metabolic Improvements of Bariatric Surgery. Gastroenterology. 2019;156:1041-1051.e4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 102]  [Article Influence: 20.4]  [Reference Citation Analysis (0)]
79.  Albaugh VL, Banan B, Ajouz H, Abumrad NN, Flynn CR. Bile acids and bariatric surgery. Mol Aspects Med. 2017;56:75-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 86]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
80.  Molinaro A, Wahlström A, Marschall HU. Role of Bile Acids in Metabolic Control. Trends Endocrinol Metab. 2018;29:31-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 247]  [Article Influence: 41.2]  [Reference Citation Analysis (0)]
81.  Lefebvre P, Cariou B, Lien F, Kuipers F, Staels B. Role of bile acids and bile acid receptors in metabolic regulation. Physiol Rev. 2009;89:147-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1046]  [Cited by in F6Publishing: 1114]  [Article Influence: 74.3]  [Reference Citation Analysis (0)]
82.  González-Regueiro JA, Moreno-Castañeda L, Uribe M, Chávez-Tapia NC. The Role of Bile Acids in Glucose Metabolism and Their Relation with Diabetes. Ann Hepatol. 2017;16:16-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 154]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
83.  Fiorucci S, Distrutti E. Bile Acid-Activated Receptors, Intestinal Microbiota, and the Treatment of Metabolic Disorders. Trends Mol Med. 2015;21:702-714.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 273]  [Cited by in F6Publishing: 314]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
84.  Cariou B, van Harmelen K, Duran-Sandoval D, van Dijk TH, Grefhorst A, Abdelkarim M, Caron S, Torpier G, Fruchart JC, Gonzalez FJ, Kuipers F, Staels B. The farnesoid X receptor modulates adiposity and peripheral insulin sensitivity in mice. J Biol Chem. 2006;281:11039-11049.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 391]  [Cited by in F6Publishing: 411]  [Article Influence: 22.8]  [Reference Citation Analysis (0)]
85.  Zhang Y, Lee FY, Barrera G, Lee H, Vales C, Gonzalez FJ, Willson TM, Edwards PA. Activation of the nuclear receptor FXR improves hyperglycemia and hyperlipidemia in diabetic mice. Proc Natl Acad Sci USA. 2006;103:1006-1011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 743]  [Cited by in F6Publishing: 702]  [Article Influence: 39.0]  [Reference Citation Analysis (0)]
86.  Ryan KK, Tremaroli V, Clemmensen C, Kovatcheva-Datchary P, Myronovych A, Karns R, Wilson-Pérez HE, Sandoval DA, Kohli R, Bäckhed F, Seeley RJ. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509:183-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 694]  [Cited by in F6Publishing: 704]  [Article Influence: 70.4]  [Reference Citation Analysis (0)]
87.  Sachdev S, Wang Q, Billington C, Connett J, Ahmed L, Inabnet W, Chua S, Ikramuddin S, Korner J. FGF 19 and Bile Acids Increase Following Roux-en-Y Gastric Bypass but Not After Medical Management in Patients with Type 2 Diabetes. Obes Surg. 2016;26:957-965.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 76]  [Article Influence: 9.5]  [Reference Citation Analysis (0)]
88.  Nemati R, Lu J, Dokpuang D, Booth M, Plank LD, Murphy R. Increased Bile Acids and FGF19 After Sleeve Gastrectomy and Roux-en-Y Gastric Bypass Correlate with Improvement in Type 2 Diabetes in a Randomized Trial. Obes Surg. 2018;28:2672-2686.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 58]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
89.  Bozadjieva N, Heppner KM, Seeley RJ. Targeting FXR and FGF19 to Treat Metabolic Diseases-Lessons Learned From Bariatric Surgery. Diabetes. 2018;67:1720-1728.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 67]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
90.  McGavigan AK, Garibay D, Henseler ZM, Chen J, Bettaieb A, Haj FG, Ley RE, Chouinard ML, Cummings BP. TGR5 contributes to glucoregulatory improvements after vertical sleeve gastrectomy in mice. Gut. 2017;66:226-234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 156]  [Article Influence: 22.3]  [Reference Citation Analysis (0)]
91.  Ding L, Sousa KM, Jin L, Dong B, Kim BW, Ramirez R, Xiao Z, Gu Y, Yang Q, Wang J, Yu D, Pigazzi A, Schones D, Yang L, Moore D, Wang Z, Huang W. Vertical sleeve gastrectomy activates GPBAR-1/TGR5 to sustain weight loss, improve fatty liver, and remit insulin resistance in mice. Hepatology. 2016;64:760-773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 122]  [Cited by in F6Publishing: 133]  [Article Influence: 16.6]  [Reference Citation Analysis (0)]
92.  Sommer F, Bäckhed F. The gut microbiota--masters of host development and physiology. Nat Rev Microbiol. 2013;11:227-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2011]  [Cited by in F6Publishing: 2079]  [Article Influence: 189.0]  [Reference Citation Analysis (0)]
93.  Festi D, Schiumerini R, Eusebi LH, Marasco G, Taddia M, Colecchia A. Gut microbiota and metabolic syndrome. World J Gastroenterol. 2014;20:16079-16094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 342]  [Cited by in F6Publishing: 309]  [Article Influence: 30.9]  [Reference Citation Analysis (2)]
94.  Dabke K, Hendrick G, Devkota S. The gut microbiome and metabolic syndrome. J Clin Invest. 2019;129:4050-4057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 354]  [Article Influence: 88.5]  [Reference Citation Analysis (0)]
95.  Bishehsari F, Voigt RM, Keshavarzian A. Circadian rhythms and the gut microbiota: from the metabolic syndrome to cancer. Nat Rev Endocrinol. 2020;16:731-739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 123]  [Article Influence: 30.8]  [Reference Citation Analysis (0)]
96.  Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci USA. 2005;102:11070-11075.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4639]  [Cited by in F6Publishing: 4164]  [Article Influence: 219.2]  [Reference Citation Analysis (1)]
97.  Debédat J, Clément K, Aron-Wisnewsky J. Gut Microbiota Dysbiosis in Human Obesity: Impact of Bariatric Surgery. Curr Obes Rep. 2019;8:229-242.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 70]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
98.  Ciobârcă D, Cătoi AF, Copăescu C, Miere D, Crișan G. Bariatric Surgery in Obesity: Effects on Gut Microbiota and Micronutrient Status. Nutrients. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 57]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
99.  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;27:1345-1357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 104]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
100.  Cӑtoi AF, Vodnar DC, Corina A, Nikolic D, Citarrella R, Pérez-Martínez P, Rizzo M. Gut Microbiota, Obesity and Bariatric Surgery: Current Knowledge and Future Perspectives. Curr Pharm Des. 2019;25:2038-2050.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
101.  Liou AP, Paziuk M, Luevano JM Jr, Machineni S, Turnbaugh PJ, Kaplan LM. Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity. Sci Transl Med. 2013;5:178ra41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 643]  [Cited by in F6Publishing: 671]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
102.  de Groot P, Scheithauer T, Bakker GJ, Prodan A, Levin E, Khan MT, Herrema H, Ackermans M, Serlie MJM, de Brauw M, Levels JHM, Sales A, Gerdes VE, Ståhlman M, Schimmel AWM, Dallinga-Thie G, Bergman JJ, Holleman F, Hoekstra JBL, Groen A, Bäckhed F, Nieuwdorp M. Donor metabolic characteristics drive effects of faecal microbiota transplantation on recipient insulin sensitivity, energy expenditure and intestinal transit time. Gut. 2020;69:502-512.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 158]  [Article Influence: 39.5]  [Reference Citation Analysis (0)]
103.  Shaw D, Gohil K, Basson MD. Intestinal mucosal atrophy and adaptation. World J Gastroenterol. 2012;18:6357-6375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 94]  [Cited by in F6Publishing: 82]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
104.  Drozdowski LA, Clandinin MT, Thomson AB. Morphological, kinetic, membrane biochemical and genetic aspects of intestinal enteroplasticity. World J Gastroenterol. 2009;15:774-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 19]  [Cited by in F6Publishing: 18]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
105.  Li B, Lu Y, Srikant CB, Gao ZH, Liu JL. Intestinal adaptation and Reg gene expression induced by antidiabetic duodenal-jejunal bypass surgery in Zucker fatty rats. Am J Physiol Gastrointest Liver Physiol. 2013;304:G635-G645.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
106.  Habegger KM, Al-Massadi O, Heppner KM, Myronovych A, Holland J, Berger J, Yi CX, Gao Y, Lehti M, Ottaway N, Amburgy S, Raver C, Müller TD, Pfluger PT, Kohli R, Perez-Tilve D, Seeley RJ, Tschöp MH. Duodenal nutrient exclusion improves metabolic syndrome and stimulates villus hyperplasia. Gut. 2014;63:1238-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 41]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
107.  Kohli R, Kirby M, Setchell KD, Jha P, Klustaitis K, Woollett LA, Pfluger PT, Balistreri WF, Tso P, Jandacek RJ, Woods SC, Heubi JE, Tschoep MH, D'Alessio DA, Shroyer NF, Seeley RJ. Intestinal adaptation after ileal interposition surgery increases bile acid recycling and protects against obesity-related comorbidities. Am J Physiol Gastrointest Liver Physiol. 2010;299:G652-G660.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 122]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
108.  le Roux CW, Borg C, Wallis K, Vincent RP, Bueter M, Goodlad R, Ghatei MA, Patel A, Bloom SR, Aylwin SJ. Gut hypertrophy after gastric bypass is associated with increased glucagon-like peptide 2 and intestinal crypt cell proliferation. Ann Surg. 2010;252:50-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 135]  [Cited by in F6Publishing: 140]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
109.  Taqi E, Wallace LE, de Heuvel E, Chelikani PK, Zheng H, Berthoud HR, Holst JJ, Sigalet DL. The influence of nutrients, biliary-pancreatic secretions, and systemic trophic hormones on intestinal adaptation in a Roux-en-Y bypass model. J Pediatr Surg. 2010;45:987-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
110.  Ren Y, Zhao Z, Zhao G, Liu Q, Wang Z, Liu R. Sleeve Gastrectomy Surgery Improves Glucose Metabolism by Downregulating the Intestinal Expression of Sodium-Glucose Cotransporter-3. J Invest Surg. 2020;1-9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
111.  Troy S, Soty M, Ribeiro L, Laval L, Migrenne S, Fioramonti X, Pillot B, Fauveau V, Aubert R, Viollet B, Foretz M, Leclerc J, Duchampt A, Zitoun C, Thorens B, Magnan C, Mithieux G, Andreelli F. Intestinal gluconeogenesis is a key factor for early metabolic changes after gastric bypass but not after gastric lap-band in mice. Cell Metab. 2008;8:201-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 237]  [Cited by in F6Publishing: 197]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]