Molecular remodeling of adipose tissue is associated with metabolic recovery after weight loss surgery

Annie Bouchard-Mercier, Juan de Toro-Martín, Mélanie Nadeau, Odette Lescelleur, Stéfane Lebel, Denis Richard, Laurent Biertho, André Tchernof, Marie-Claude Vohl, Annie Bouchard-Mercier, Juan de Toro-Martín, Mélanie Nadeau, Odette Lescelleur, Stéfane Lebel, Denis Richard, Laurent Biertho, André Tchernof, Marie-Claude Vohl

Abstract

Background: Bariatric surgery is an effective therapy for individuals with severe obesity to achieve sustainable weight loss and to reduce comorbidities. Examining the molecular signature of subcutaneous adipose tissue (SAT) following different types of bariatric surgery may help in gaining further insight into their distinct metabolic impact.

Results: Subjects undergoing biliopancreatic diversion with duodenal switch (BPD-DS) showed a significantly higher percentage of total weight loss than those undergoing gastric bypass or sleeve gastrectomy (RYGB + SG) (41.7 ± 4.6 vs 28.2 ± 6.8%; p = 0.00005). Individuals losing more weight were also significantly more prone to achieve both type 2 diabetes and dyslipidemia remission (OR = 0.75; 95%CI = 0.51-0.91; p = 0.03). Whole transcriptome and methylome profiling showed that bariatric surgery induced a profound molecular remodeling of SAT at 12 months postoperative, mainly through gene down-regulation and hypermethylation. The extent of changes observed was greater following BPD-DS, with 61.1% and 49.8% of up- and down-regulated genes, as well as 85.7% and 70.4% of hyper- and hypomethylated genes being exclusive to this procedure, and mostly associated with a marked decrease of immune and inflammatory responses. Weight loss was strongly associated with genes being simultaneously differentially expressed and methylated in BPD-DS, with the strongest association being observed for GPD1L (r2 = 0.83; p = 1.4 × 10-6).

Conclusions: Present findings point to the greater SAT molecular remodeling following BPD-DS as potentially linked with higher metabolic remission rates. These results will contribute to a better understanding of the metabolic pathways involved in the response to bariatric surgery and will eventually lead to the development of gene targets for the treatment of obesity. Trial registration ClinicalTrials.gov NCT02390973.

Keywords: Bariatric surgery; Dyslipidemia; Methylomic; Obesity; Remission; Subcutaneous adipose tissue; Transcriptomic; Type 2 diabetes; Whole genome.

Conflict of interest statement

AT and LB received research funding from Johnson & Johnson for the present study in conjunction with a team grant from the Canadian Institutes of Health Research. They also receive funding from Medtronic and GI Windows for studies unrelated to the present article. AT received consulting fees from Novo Nordisk, Eli Lilly and Bausch Health. The remaining authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Flow diagram of study participants
Fig. 2
Fig. 2
Gene expression changes in subcutaneous adipose tissue were more pronounced following BPD-DS. Panel A shows differentially expressed genes (DEGs) exclusive for BPD-DS (green dots) and common to both surgery groups (red dots). Panel B shows DEGs exclusive for RYGB-DS (blue dots) and common to both surgery groups (purple dots). DEGs were considered significant when false discovery rate (FDR)-corrected p-value < 0.05 and fold change (FC) > 1.5. Panels C and D show density plots of FC distribution among down-regulated and up-regulated DEGs, respectively. Green and blue colors stand for DEGs exclusive for BPD-DS and RYGB + SG, respectively. Red and purple colors stand for DEGs common to both surgery groups but showing the specific FC distribution for each BPD-DS and RYGB + SG surgery, respectively. Dotted lines stand for mean FC values for each surgery group
Fig. 3
Fig. 3
Most of gene methylation changes in subcutaneous adipose tissue occur following BPD-DS. Panel A shows differentially methylated genes (DMGs) exclusive for BPD-DS (green dots) and common to both surgery groups (red dots). Panel B shows DMGs exclusive for RYGB-DS (blue dots) and common to both surgery groups (purple dots). DMGs were defined as loci with at least one differentially methylated CpG site (false discovery rate (FDR)-corrected p-value < 0.05 and fold change (FC) > 1.5. Panels C and D show density plots of FC distribution among hypermethylated and hypomethylated DMGs, respectively. Green and blue colors stand for DEGs exclusive for BPD-DS and RYGB + SG, respectively. Red and purple colors stand for DMGs common to both surgery groups but showing the specific FC distribution for each BPD-DS and RYGB + SG surgery, respectively. Dotted lines stand for mean FC values for each surgery
Fig. 4
Fig. 4
Immune-related pathways were markedly down-regulated following bariatric surgery. Left panel shows top Gene Ontology-Biological Process (GO-BP) terms significantly enriched with up-regulated (red blocks, up) and down-regulated (blue blocks, down) differentially expressed genes (DEGs). Right panel shows top GO-BP terms significantly enriched with hypermethylated (red blocks, up) and hypomethylated (blue blocks, down) differentially methylated genes (DMGs). Each column represents pathways enriched with DEGs specific to BPD-DS, RYGB + SG or common to both surgery groups. Pathways were considered significantly enriched when composed with at least 20 DEGs or DMGs and with FDR-adjusted p-value 

Fig. 5

Genes being simultaneously differentially expressed…

Fig. 5

Genes being simultaneously differentially expressed and methylated largely belonged to the BPD-DS surgery…

Fig. 5
Genes being simultaneously differentially expressed and methylated largely belonged to the BPD-DS surgery group. A and B show respectively the proportion of differentially expressed genes (DEGs) down- and up-regulated that are simultaneously identified as differentially methylated genes (DMGs). The proportion of DEGs common to both surgery groups (COMMON), as well as exclusive to BPD-DS (BPD) and RYGB + SG (GAS) is shown in the inner ring. The proportion of hypermethylated and hypomethylated DMGs is shown in the outer ring. C and D show respectively the proportion of hypermethylated and hypomethylated CpG sites located within body or promoter regions of genes being simultaneously DEGs and DMGs. The proportion of genes being simultaneously DEGs and DMGs is shown in the inner ring, and the proportion of CpG sites for each gene location and surgery group is shown in the outer ring

Fig. 6

Differentially expressed and methylated genes…

Fig. 6

Differentially expressed and methylated genes were associated with weight loss, adipocyte size and…

Fig. 6
Differentially expressed and methylated genes were associated with weight loss, adipocyte size and neck circumference. A to C show the predicted probability (red dots from 0 to 1), obtained by binomial logistic regression, of each participant to have a complete (0) or a partial remission (1), based on %TWL, %adipocyte size and %neck circumference. OR is the odds ratio with 95% confidence intervals (CI) and P is the p value for the linear trend of association. Gray and blue dots refer BPD-DS and RYGB + SG, respectively. D to F respectively show associations between differentially expressed genes (DEGs) in each surgery group with the percentage of total weight loss (%TWL), adipocyte size change (%Adipocyte) and neck circumference change (%Neck). Green, blue and red dots respectively stand for associations at non-adjusted p < 0.05 with DEGs exclusive to BPD-DS, RYGB + SG or common to both surgery groups. Grey dots represent not significant associations. Dot size is proportional to the magnitude (r2) of the association. Results are from multivariate linear regression models adjusted for sex, age and BMI
Fig. 5
Fig. 5
Genes being simultaneously differentially expressed and methylated largely belonged to the BPD-DS surgery group. A and B show respectively the proportion of differentially expressed genes (DEGs) down- and up-regulated that are simultaneously identified as differentially methylated genes (DMGs). The proportion of DEGs common to both surgery groups (COMMON), as well as exclusive to BPD-DS (BPD) and RYGB + SG (GAS) is shown in the inner ring. The proportion of hypermethylated and hypomethylated DMGs is shown in the outer ring. C and D show respectively the proportion of hypermethylated and hypomethylated CpG sites located within body or promoter regions of genes being simultaneously DEGs and DMGs. The proportion of genes being simultaneously DEGs and DMGs is shown in the inner ring, and the proportion of CpG sites for each gene location and surgery group is shown in the outer ring
Fig. 6
Fig. 6
Differentially expressed and methylated genes were associated with weight loss, adipocyte size and neck circumference. A to C show the predicted probability (red dots from 0 to 1), obtained by binomial logistic regression, of each participant to have a complete (0) or a partial remission (1), based on %TWL, %adipocyte size and %neck circumference. OR is the odds ratio with 95% confidence intervals (CI) and P is the p value for the linear trend of association. Gray and blue dots refer BPD-DS and RYGB + SG, respectively. D to F respectively show associations between differentially expressed genes (DEGs) in each surgery group with the percentage of total weight loss (%TWL), adipocyte size change (%Adipocyte) and neck circumference change (%Neck). Green, blue and red dots respectively stand for associations at non-adjusted p < 0.05 with DEGs exclusive to BPD-DS, RYGB + SG or common to both surgery groups. Grey dots represent not significant associations. Dot size is proportional to the magnitude (r2) of the association. Results are from multivariate linear regression models adjusted for sex, age and BMI

References

    1. World Health Organization. Obesity and overweight. 2021. Acessed from:
    1. Center for disease control and prevention. Adults obesity facts. 2021. Acessed from:
    1. Wolfe BM, Kvach E, Eckel RH. Treatment of Obesity. Circ Res. 2016;11:1844.
    1. Wing RR, Phelan S. Long-term weight loss maintenance. Am J Clin Nutr. 2005;82:222s.
    1. O’Brien PE, Hindle A, Brennan L, Skinner S, Burton P, Smith A, et al. Long-term outcomes after bariatric surgery: a systematic review and meta-analysis of weight loss at 10 or more years for all bariatric procedures and a single-centre review of 20-year outcomes after adjustable gastric banding. Obes Surg. 2019;29(1):3.
    1. Vidal J, Corcelles R, Jiménez A, Flores L, Lacy AM. Metabolic and bariatric surgery for obesity. Gastroenterology. 2017;152:1780.
    1. Buchwald H, Buchwald JN. Metabolic (bariatric and nonbariatric) surgery for type 2 diabetes: a personal perspective review. Diabetes Care. 2019;42:331.
    1. Brethauer SA, Kim J, el Chaar M, Papasavas P, Eisenberg D, Rogers A, et al. Standardized outcomes reporting in metabolic and bariatric surgery. Surg Obes Relat Dis. 2015;11:489.
    1. Biertho L, Lebel S, Marceau S, Hould F-SS, Julien F, Biron S. Biliopancreatic diversion with duodenal switch. Surg Clin North Am. 2016;96:815–26.
    1. Metere A, Graves CE. Factors influencing epigenetic mechanisms: is there a role for bariatric surgery? High Throughput. 2020;9(1):6.
    1. Samblas M, Milagro FI, Martínez A. DNA methylation markers in obesity, metabolic syndrome, and weight loss. Epigenetics. 2019;14:421.
    1. Nicoletti CF, Pinhel MS, Noronha NY, Jácome A, Crujeiras AB, Nonino CB. Association of MFSD3 promoter methylation level and weight regain after gastric bypass: assessment for 3 y after surgery. Nutrition. 2020;70:110499.
    1. Assem S, Abdelbaki TN, Mohy-El Dine SH, Ketat AF, Abdelmonsif DA. SERPINE-1 gene methylation and protein as molecular predictors of laparoscopic sleeve gastrectomy outcome. Obes Surg. 2020;30(7):2620.
    1. Barres R, Kirchner H, Rasmussen M, Yan J, Kantor FR, Krook A, et al. Weight loss after gastric bypass surgery in human obesity remodels promoter methylation. Cell Rep. 2013 doi: 10.1016/j.celrep.2013.05.017.
    1. Fraszczyk E, Luijten M, Spijkerman AMW, Snieder H, Wackers PFK, Bloks VW, et al. The effects of bariatric surgery on clinical profile, DNA methylation, and ageing in severely obese patients. Clin Epigenet. 2020 doi: 10.1186/s13148-019-0790-2.
    1. Dahlman I, Sinha I, Gao H, Brodin D, Thorell A, Rydén M, et al. The fat cell epigenetic signature in post-obese women is characterized by global hypomethylation and differential DNA methylation of adipogenesis genes. Int J Obes. 2015 doi: 10.1038/ijo.2015.31.
    1. Benton MC, Johnstone A, Eccles D, Harmon B, Hayes MT, Lea RA, et al. An analysis of DNA methylation in human adipose tissue reveals differential modification of obesity genes before and after gastric bypass and weight loss. Genome Biol. 2015 doi: 10.1186/s13059-014-0569-x.
    1. Garcia LA, Day SE, Coletta RL, Campos B, Benjamin TR, de Filippis E, et al. Weight loss after roux-en-y gastric bypass surgery reveals skeletal muscle DNA methylation changes. Clin Epigent. 2021 doi: 10.1186/s13148-021-01086-6.
    1. Ghosh S, Yates AJ, Frühwald MC, Miecznikowski JC, Plass C, Smiraglia D. Tissue specific DNA methylation of CpG islands in normal human adult somatic tissues distinguishes neural from non-neural tissues. Epigenetics. 2010;5(6):527.
    1. Biertho L, Simon-Hould F, Marceau S, Lebel S, Lescelleur O, Biron S. Current Outcomes of Laparoscopic Duodenal Switch. Ann Surg Innov Res. 2016 doi: 10.1186/s13022-016-0024-7.
    1. Marceau P, Biron S, Marceau S, Hould F-S, Lebel S, Lescelleur O, et al. Long-term metabolic outcomes 5 to 20 years after biliopancreatic diversion. Obes Surg. 2015;25(9):1584.
    1. Bolckmans R, Himpens J. Long-term (>10 Yrs) outcome of the laparoscopic biliopancreatic diversion with duodenal switch. Ann Surg. 2016;264(6):1029.
    1. Skogar ML, Sundbom M. Duodenal switch is superior to gastric bypass in patients with super obesity when evaluated with the bariatric analysis and reporting outcome system (BAROS) Obes Surg. 2017;27(9):2308.
    1. Longo M, Zatterale F, Naderi J, Parrillo L, Formisano P, Raciti GA, et al. Adipose tissue dysfunction as determinant of obesity-associated metabolic complications. Int J Mol Sci. 2019;20(9):3258.
    1. Adami GF, Carbone F, Montecucco F, Camerini G, Cordera R. Adipose tissue composition in obesity and after bariatric surgery. Obes Surg. 2019;29(9):3030.
    1. Grenier-Larouche T, Carreau AM, Geloën A, Frisch F, Biertho L, Marceau S, et al. Fatty acid metabolic remodeling during type 2 diabetes remission after bariatric surgery. Diabetes. 2017;66:2743–55.
    1. de Pinhel MA, Noronha SNY, Nicoletti CF, de Oliveira BAP, Cortes-Oliveira C, Pinhanelli VC, et al. Changes in global transcriptional profiling of women following obesity surgery bypass. Obes Surg. 2018;28:176.
    1. Beisani M, Pappa S, Moreno P, Martínez E, Tarascó J, Granada ML, et al. Laparoscopic sleeve gastrectomy induces molecular changes in peripheral white blood cells. Clin Nutr. 2020;39:592.
    1. ElGendy K, Malcomson FC, Bradburn DM, Mathers JC. Effects of bariatric surgery on DNA methylation in adults: a systematic review and meta-analysis. Surg Obes Relat Dis. 2020;16:128.
    1. Pinhel MAS, Noronha NY, Nicoletti CF, Pereira VA, de Oliveira BA, Cortes-Oliveira C, et al. Changes in DNA methylation and gene expression of insulin and obesity-related gene PIK3R1 after roux-en-Y gastric bypass. Int J Mol Sci. 2020;21:4476.
    1. Gancheva S, Ouni M, Jelenik T, Koliaki C, Szendroedi J, Toledo FGS, et al. Dynamic changes of muscle insulin sensitivity after metabolic surgery. Nat Commun. 2019 doi: 10.1038/s41467-019-12081-0.
    1. Kerr AG, Andersson DP, Rydén M, Arner P, Dahlman I. Long-term changes in adipose tissue gene expression following bariatric surgery. J Int Med. 2020 doi: 10.1111/joim.13066.
    1. Punthakee Z, Goldenberg R, Katz P. Definition, classification and diagnosis of diabetes, prediabetes and metabolic syndrome. Can J Diabetes. 2018;42:10–15.
    1. Grover BT, Morell MC, Kothari SN, Borgert AJ, Kallies KJ, Baker MT. Defining weight loss after bariatric surgery: a call for standardization. Obes Surg. 2019 doi: 10.1007/s11695-019-04022-z.
    1. Corcelles R, Boules M, Froylich D, Hag A, Daigle CR, Aminian A, et al. Total Weight Loss as the Outcome Measure of Choice After Roux-en-Y Gastric Bypass. Obes Surg. 2016 doi: 10.1007/s11695-015-2022-y.
    1. van Dijk SJ, Molloy PL, Varinli H, Morrison JL, Muhlhausler BS. Epigenetics and human obesity. Int J Obes. 2015;39:85.
    1. Mingrone G, Panunzi S, de Gaetano A, Guidone C, Iaconelli A, Nanni G, et al. Bariatric–metabolic surgery versus 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.
    1. Skroubis G, Kouri N, Mead N, Kalfarentzos F. Long-term results of a prospective comparison of roux-en-Y gastric bypass versus a variant of biliopancreatic diversion in a non-superobese population (BMI 35–50 kg/m2) Obes Surg. 2014;24:197.
    1. Sjöström CD, Peltonen M, Sjöström L. Blood pressure and pulse pressure during long-term weight loss in the obese: the swedish obese subjects (SOS) intervention study. Obes Res. 2001;9:188.
    1. Edholm D, Svensson F, Näslund I, Karlsson FA, Rask E, Sundbom M. Long-term results 11 years after primary gastric bypass in 384 patients. Surg Obes Relat Dis. 2013;9:708.
    1. MacHado MB, Velasco IT, Scalabrini-Neto A. Gastric bypass and cardiac autonomic activity: influence of gender and age. Obes Surg. 2009;19:332–8.
    1. Roberts K, Duffy A, Kaufman J, Burrell M, Dziura J, Bell R. Size matters: gastric pouch size correlates with weight loss after laparoscopic Roux-en-Y gastric bypass. Surg Endosc. 2007;21:1397–402.
    1. Melton GB, Steele KE, Schweitzer MA, Lidor AO, Magnuson TH. Suboptimal weight loss after gastric bypass surgery: correlation of demographics, comorbidities, and insurance status with outcomes. J Gastrointest Surg. 2008;12:250–5.
    1. de Toro-Martín J, Guénard F, Tchernof A, Pérusse L, Marceau S, Vohl M-C. Polygenic risk score for predicting weight loss after bariatric surgery. JCI Insight. 2018 doi: 10.1172/jci.insight.122011.
    1. van Rijswijk A-S, van Olst N, Schats W, van der Peet DL, van de Laar AW. What is weight loss after bariatric surgery expressed in percentage total weight Loss (%TWL)? a systematic Review. Obes Surg. 2021;31:3833.
    1. Bioletto F, Pellegrini M, D’Eusebio C, Boschetti S, Rahimi F, de Francesco A, et al. Development and validation of a scoring system for pre-surgical and early post-surgical prediction of bariatric surgery unsuccess at 2 years. Sci Rep. 2021;11:1–10.
    1. Baraboi E-D, Li W, Labbé SM, Roy M-C, Samson P, Hould F-S, et al. Metabolic changes induced by the biliopancreatic diversion in diet-induced obesity in male rats: the contributions of sleeve gastrectomy and duodenal switch. Endocrinology. 2015;156:13106.
    1. Vink RG, Roumans NJ, Fazelzadeh P, Tareen SHK, Boekschoten M, v, van Baak MA,, et al. Adipose tissue gene expression is differentially regulated with different rates of weight loss in overweight and obese humans. Int J Obes. 2017;41:309.
    1. Castagneto-Gissey L, Angelini G, Casella-Mariolo JR, Marini P, Mingrone G, Casella G. The jejunum is the key factor in insulin resistance. Surg Obes Relat Dis. 2020;16:509.
    1. Keleher MR, Zaidi R, Hicks L, Shah S, Xing X, Li D, et al. A high-fat diet alters genome-wide DNA methylation and gene expression in SM/J mice. BMC Geno. 2018 doi: 10.1186/s12864-018-5327-0.
    1. Gaye A, Doumatey AP, Davis SK, Rotimi CN, Gibbons GH. Whole-genome transcriptomic insights into protective molecular mechanisms in metabolically healthy obese African Americans. Geno Med. 2018 doi: 10.1038/s41525-018-0043-x.
    1. Yoshino M, Kayser BD, Yoshino J, Stein RI, Reeds D, Eagon JC, et al. Effects of diet versus gastric bypass on metabolic function in diabetes. New Engl J Med. 2020;383:721.
    1. Proud CG. Regulation of protein synthesis by insulin. Biochem Soci Trans. 2006;34:213.
    1. MacLaren R, Cui W, Simard S, Cianflone K. Influence of obesity and insulin sensitivity on insulin signaling genes in human omental and subcutaneous adipose tissue. J Lipid Res. 2008;49:308.
    1. Scheja L, Heeren J. The endocrine function of adipose tissues in health and cardiometabolic disease. Nat Rev Endocrinol. 2019;15:507.
    1. Zatterale F, Longo M, Naderi J, Raciti GA, Desiderio A, Miele C, et al. Chronic adipose tissue inflammation linking obesity to insulin resistance and type 2 diabetes. Frontiers in physiology. 2020;1607.
    1. DeBari MK, Abbott RD. Adipose tissue fibrosis: mechanisms, models, and importance. Int J Mol Sci. 2020;21:6030.
    1. Duffaut C, Zakaroff-Girard A, Bourlier V, Decaunes P, Maumus M, Chiotasso P, et al. Interplay between human adipocytes and T lymphocytes in obesity. Arteriosclerosis Thrombo Vasc Biol. 2009 doi: 10.1161/ATVBAHA.109.192583.
    1. Villarreal-Calderón JR, Cuéllar RX, Ramos-González MR, Rubio-Infante N, Castillo EC, Elizondo-Montemayor L, et al. Interplay between the adaptive immune system and insulin resistance in weight loss induced by bariatric surgery. Oxid Med Cell Longev. 2019 doi: 10.1155/2019/3940739.
    1. Poitou C, Perret C, Mathieu F, Truong V, Blum Y, Durand H, et al. Bariatric surgery induces disruption in inflammatory signaling pathways mediated by immune cells in adipose tissue: A RNA-Seq study. PLOS ONE. 2015;10:e0125718.
    1. Cancello R, Henegar C, Viguerie N, Taleb S, Poitou C, Rouault C, et al. Reduction of macrophage infiltration and chemoattractant gene expression changes in white adipose tissue of morbidly obese subjects after surgery-induced weight loss. Diabetes. 2005;54:2277.
    1. Ding S, Xu S, Ma Y, Liu G, Jang H, Fang J. Modulatory mechanisms of the NLRP3 inflammasomes in diabetes. Biomolecules. 2019 doi: 10.3390/biom9120850.
    1. Kolehmainen M, Salopuro T, Schwab US, Kekäläinen J, Kallio P, Laaksonen DE, et al. Weight reduction modulates expression of genes involved in extracellular matrix and cell death: the GENOBIN study. Int J Obes. 2008;32(1):292–303.
    1. Abdennour M, Reggio S, le Naour G, Liu Y, Poitou C, Aron-Wisnewsky J, et al. Association of adipose tissue and liver fibrosis with tissue stiffness in morbid obesity: links with diabetes and bmi loss after gastric bypass. J Clin Endocrinol Metab. 2014;99(3):898–907.
    1. Valdez Y, Maia M, M. Conway E. CD248: reviewing its role in health and disease. Current Drug Targets. 2012;13:432.
    1. Petrus P, Fernandez TL, Kwon MM, Huang JL, Lei V, Safikhan NS, et al. Specific loss of adipocyte CD248 improves metabolic health via reduced white adipose tissue hypoxia, fibrosis and inflammation. EBioMedicine. 2019;44:489.
    1. Lu Z, Meng L, Sun Z, Shi X, Shao W, Zheng Y, et al. Differentially expressed genes and enriched signaling pathways in the adipose tissue of obese people. Front Genet. 2021 doi: 10.3389/fgene.2021.620740.
    1. He H, Sun D, Zeng Y, Wang R, Zhu W, Cao S, et al. A Systems genetics approach identified GPD1L and its molecular mechanism for obesity in human adipose tissue. Sci Rep. 2017 doi: 10.1038/s41598-017-01517-6.
    1. Joshi H, Vastrad B, Joshi N, Vastrad C, Tengli A, Kotturshetti I. Identification of key pathways and genes in obesity using bioinformatics analysis and molecular docking studies. Front Endocrinol. 2021 doi: 10.3389/fendo.2021.628907.
    1. Courcoulas AP, King WC, Belle SH, Berk P, Flum DR, Garcia L, et al. 7-Year weight trajectories and health outcomes in the longitudinal assessment of bariatric surgery (LABS) study. JAMA Surg. 2018;153:427.
    1. Topart P, Becouarn G, Delarue J. Weight loss and nutritional outcomes 10 years after biliopancreatic diversion with duodenal switch. Obes Surg. 2017;27(11):1645–50.
    1. Gastrointestinal surgery for severe obesity: National institutes of health consensus development conference statement. Am J Clini Nutr. 1992;55.
    1. Marceau P, Biron S, Hould F-S, Lebel S, Marceau S, Lescelleur O, et al. Duodenal switch longterm results. Obes Surg. 2007;17(11):1421–30.
    1. Grenier-Larouche T, Galinier A, Casteilla L, Carpentier AC, Tchernof A. Omental adipocyte hypertrophy relates to coenzyme Q10 redox state and lipid peroxidation in obese women. J Lipid Res. 2015;56:1985.
    1. Krueger F. Trim Galore. 2019. .
    1. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J EMBnet Stichting. 2011;17:10–12.
    1. Andrews S. FastQC: a quality control tool for high throughput sequence data. 2019. .
    1. Bray NL, Pimentel H, Melsted P, Pachter L. Near-optimal probabilistic RNA-seq quantification. Nat Biotechnol Nat Publ Gr. 2016;34:525–527.
    1. Soneson C, Love MI, Robinson MD. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res. Faculty of 1000 Ltd; 2015;4.
    1. Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics Oxf Univ Press. 2009;26:139–140.
    1. Touleimat N, Tost J. Complete pipeline for Infinium® Human Methylation 450K BeadChip data processing using subset quantile normalization for accurate DNA methylation estimation. Epigenomics. 2012 doi: 10.2217/epi1221.
    1. Fortin J-P, Triche TJ, Hansen KD. Preprocessing, normalization and integration of the illumina humanmethylationEPIC array with minfi. Bioinformatics. 2016;33:btw691.
    1. Yu G, Wang LG, Han Y, ClusterProfiler He QY. An R package for comparing biological themes among gene clusters. OMICS J Integr Biol. 2012;16:284–7.
    1. R Core Team. R: A language and environment for statistical computing. R Foundation for Statistical Computing, Vienna, Austria. 2021. .

Source: PubMed

3
Subskrybuj