Skip to main content

Advertisement

Log in

The Effects of Bariatric Surgery-Induced Weight Loss on Adipose Tissue in Morbidly Obese Women Depends on the Initial Metabolic Status

  • Original Contributions
  • Published:
Obesity Surgery Aims and scope Submit manuscript

Abstract

Background

Adipose tissue (AT) dysfunction in obesity is commonly linked to insulin resistance and promotes the development of metabolic disease. Bariatric surgery (BS) represents an effective strategy to reduce weight and to improve metabolic health in morbidly obese subjects. However, the mechanisms and pathways that are modified in AT in response to BS are not fully understood, and few information is still available as to whether these may vary depending on the metabolic status of obese subjects.

Methods

Abdominal subcutaneous adipose tissue (SAT) samples were obtained from morbidly obese women (n = 18) before and 13.3 ± 0.37 months after BS. Obese women were stratified into two groups: normoglycemic (NG; Glu < 100 mg/dl, HbA1c <5.7 %) or insulin resistant (IR; Glu 100–126 mg/dl, HbA1c 5.7–6.4 %) (n = 9/group). A multi-comparative proteomic analysis was employed to identify differentially regulated SAT proteins by BS and/or the degree of insulin sensitivity. Serum levels of metabolic, inflammatory, and anti-oxidant markers were also analyzed.

Results

Before surgery, NG and IR subjects exhibited differences in AT proteins related to inflammation, metabolic processes, the cytoskeleton, and mitochondria. BS caused comparable weight reductions and improved glucose homeostasis in both groups. However, BS caused dissimilar changes in metabolic enzymes, inflammatory markers, cytoskeletal components, mitochondrial proteins, and angiogenesis regulators in NG and IR women.

Conclusions

BS evokes significant molecular rearrangements indicative of improved AT function in morbidly obese women at either low or high metabolic risk, though selective adaptive changes in key cellular processes occur depending on the initial individual’s metabolic status.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Ahima RS. Digging deeper into obesity. J Clin Invest. 2011;121(6):2076–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Sun K, Tordjman J, Clement K, et al. Fibrosis and adipose tissue dysfunction. Cell Metab. 2013;18(4):470–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gregor MF, Hotamisligil GS. Inflammatory mechanisms in obesity. Annu Rev Immunol. 2011;29:415–45.

    Article  CAS  PubMed  Google Scholar 

  4. Bluher M. Adipose tissue dysfunction contributes to obesity related metabolic diseases. Best Pract Res Clin Endocrinol Metab. 2013;27(2):163–77.

    Article  PubMed  Google Scholar 

  5. Diaz-Ruiz A, Guzman-Ruiz R, Moreno NR, et al. Proteasome dysfunction associated to oxidative stress and proteotoxicity in adipocytes compromises insulin sensitivity in human obesity. Antioxid Redox Signal. 2015;23(7):597–612.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Matsuda M, Shimomura I. Increased oxidative stress in obesity: implications for metabolic syndrome, diabetes, hypertension, dyslipidemia, atherosclerosis, and cancer. Obes Res Clin Pract. 2013;7(5):e330–41.

    Article  PubMed  Google Scholar 

  7. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell. 2010;140(6):900–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Malkani S. An update on the role of bariatric surgery in diabetes management. Curr Opin Endocrinol Diabetes Obes. 2015;22(2):98–105.

    Article  PubMed  Google Scholar 

  9. Yu J, Zhou X, Li L, et al. 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(1):143–58.

    Article  PubMed  Google Scholar 

  10. Clement K. Bariatric surgery, adipose tissue and gut microbiota. Int J Obes (Lond). 2011;35 Suppl 3:S7–15.

    Article  Google Scholar 

  11. Mendez-Gimenez L, Becerril S, Moncada R, et al. Sleeve gastrectomy reduces hepatic steatosis by improving the coordinated regulation of aquaglyceroporins in adipose tissue and liver in obese rats. Obes Surg. 2015;25(9):1723–34.

    Article  PubMed  Google Scholar 

  12. Appachi S, Kelly KR, Schauer PR, et al. Reduced cardiovascular risk following bariatric surgeries is related to a partial recovery from "adiposopathy". Obes Surg. 2011;21(12):1928–36.

    Article  PubMed  PubMed Central  Google Scholar 

  13. Benraouane F, Litwin SE. Reductions in cardiovascular risk after bariatric surgery. Curr Opin Cardiol. 2011;26(6):555–61.

    Article  PubMed  Google Scholar 

  14. Bays HE, Laferrere B, Dixon J, et al. Adiposopathy and bariatric surgery: is 'sick fat' a surgical disease? Int J Clin Pract. 2009;63(9):1285–300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Dankel SN, Fadnes DJ, Stavrum AK, et al. Switch from stress response to homeobox transcription factors in adipose tissue after profound fat loss. PLoS One. 2010;5(6), e11033.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Henegar C, Tordjman J, Achard V, et al. Adipose tissue transcriptomic signature highlights the pathological relevance of extracellular matrix in human obesity. Genome Biol. 2008;9(1):R14.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Cancello R, Henegar C, Viguerie N, 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(8):2277–86.

    Article  CAS  PubMed  Google Scholar 

  18. Moreno-Navarrete JM, Ortega F, Serrano M, et al. CIDEC/FSP27 and PLIN1 gene expression run in parallel to mitochondrial genes in human adipose tissue, both increasing after weight loss. Int J Obes (Lond). 2014;38(6):865–72.

    Article  CAS  Google Scholar 

  19. Eriksson-Hogling D, Andersson DP, Backdahl J, et al. Adipose tissue morphology predicts improved insulin sensitivity following moderate or pronounced weight loss. Int J Obes (Lond). 2015;39(6):893–8.

  20. Cotillard A, Poitou C, Torcivia A, et al. Adipocyte size threshold matters: link with risk of type 2 diabetes and improved insulin resistance after gastric bypass. J Clin Endocrinol Metab. 2014;99(8):E1466–70.

    Article  CAS  PubMed  Google Scholar 

  21. Divoux A, Tordjman J, Lacasa D, et al. Fibrosis in human adipose tissue: composition, distribution, and link with lipid metabolism and fat mass loss. Diabetes. 2010;59(11):2817–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bluher S, Schwarz P. Metabolically healthy obesity from childhood to adulthood—Does weight status alone matter? Metabolism. 2014;63(9):1084–92.

    Article  PubMed  Google Scholar 

  23. Denis GV, Obin MS. 'Metabolically healthy obesity': origins and implications. Mol Aspects Med. 2013;34(1):59–70.

    Article  CAS  PubMed  Google Scholar 

  24. Primeau V, Coderre L, Karelis AD, et al. Characterizing the profile of obese patients who are metabolically healthy. Int J Obes (Lond). 2011;35(7):971–81.

    Article  CAS  Google Scholar 

  25. American DA. Diagnosis and classification of diabetes mellitus. Diabetes Care. 2012;35 Suppl 1:S64–71.

    Article  Google Scholar 

  26. Martin-Rodriguez JF, Cervera-Barajas A, Madrazo-Atutxa A, et al. Effect of bariatric surgery on microvascular dysfunction associated to metabolic syndrome: a 12-month prospective study. Int J Obes (Lond). 2014;38(11):1410–5.

    Article  CAS  Google Scholar 

  27. Scarpulla RC. Metabolic control of mitochondrial biogenesis through the PGC-1 family regulatory network. Biochim Biophys Acta. 2011;1813(7):1269–78.

    Article  CAS  PubMed  Google Scholar 

  28. Peinado JR, Quiros PM, Pulido MR, et al. Proteomic profiling of adipose tissue from Zmpste24-/- mice, a model of lipodystrophy and premature aging, reveals major changes in mitochondrial function and vimentin processing. Mol Cell Proteomics. 2011;10(11):M111 008094.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Brackley KI, Grantham J. Activities of the chaperonin containing TCP-1 (CCT): implications for cell cycle progression and cytoskeletal organisation. Cell Stress Chaperones. 2009;14(1):23–31.

    Article  CAS  PubMed  Google Scholar 

  30. Mirando AC, Francklyn CS, Lounsbury KM. Regulation of angiogenesis by aminoacyl-tRNA synthetases. Int J Mol Sci. 2014;15(12):23725–48.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Klimcakova E, Roussel B, Marquez-Quinones A, et al. Worsening of obesity and metabolic status yields similar molecular adaptations in human subcutaneous and visceral adipose tissue: decreased metabolism and increased immune response. J Clin Endocrinol Metab. 2011;96(1):E73–82.

    Article  CAS  PubMed  Google Scholar 

  32. del Pozo CH, Calvo RM, Vesperinas-Garcia G, et al. Expression profile in omental and subcutaneous adipose tissue from lean and obese subjects. Repression of lipolytic and lipogenic genes. Obes Surg. 2011;21(5):633–43.

    Article  PubMed  Google Scholar 

  33. Dharuri H, 't Hoen PA, van Klinken JB, et al. Downregulation of the acetyl-CoA metabolic network in adipose tissue of obese diabetic individuals and recovery after weight loss. Diabetologia. 2014;57(11):2384–92.

    Article  CAS  PubMed  Google Scholar 

  34. Reshef L, Olswang Y, Cassuto H, et al. Glyceroneogenesis and the triglyceride/fatty acid cycle. J Biol Chem. 2003;278(33):30413–6.

    Article  CAS  PubMed  Google Scholar 

  35. Camastra S, Gastaldelli A, Mari A, et al. Early and longer term effects of gastric bypass surgery on tissue-specific insulin sensitivity and beta cell function in morbidly obese patients with and without type 2 diabetes. Diabetologia. 2011;54(8):2093–102.

    Article  CAS  PubMed  Google Scholar 

  36. da Silva VR, Moreira EA, Wilhelm-Filho D, et al. Proinflammatory and oxidative stress markers in patients submitted to Roux-en-Y gastric bypass after 1 year of follow-up. Eur J Clin Nutr. 2012;66(8):891–9.

    Article  PubMed  Google Scholar 

  37. Joao Cabrera E, Valezi AC, Delfino VD, et al. Reduction in plasma levels of inflammatory and oxidative stress indicators after Roux-en-Y gastric bypass. Obes Surg. 2010;20(1):42–9.

    Article  PubMed  Google Scholar 

  38. Naukkarinen J, Heinonen S, Hakkarainen A, et al. Characterising metabolically healthy obesity in weight-discordant monozygotic twins. Diabetologia. 2014;57(1):167–76.

    Article  CAS  PubMed  Google Scholar 

  39. van Beek L, Lips MA, Visser A, et al. Increased systemic and adipose tissue inflammation differentiates obese women with T2DM from obese women with normal glucose tolerance. Metabolism. 2014;63(4):492–501.

    Article  PubMed  Google Scholar 

  40. von der Malsburg K, Muller JM, Bohnert M, et al. Dual role of mitofilin in mitochondrial membrane organization and protein biogenesis. Dev Cell. 2011;21(4):694–707.

    Article  PubMed  Google Scholar 

  41. Sjostrom L. Review of the key results from the Swedish Obese Subjects (SOS) trial—a prospective controlled intervention study of bariatric surgery. J Intern Med. 2013;273(3):219–34.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded by MINECO/FEDER (BFU2010-17116; BFU2013-44229-R), J. Andalucia/FEDER (PI-0269/2008; CTS-03039, CTS-6606), and CIBEROBN (Instituto de Salud Carlos III), Spain. D.A.C. was supported by the Nicolás Monardes program of the Andalusian Ministry of Health (C-0015-2014). We thank Jana Alonso (Proteomic platform of the Health Research Institute of Santiago (IDIS), University of Santiago de Compostela, Spain) and the Proteomics Facilities of the IMIBIC/University of Córdoba-SCAI (ProteoRed, PRB2-ISCIII, supported by grant PT13/0001) for their help with mass spectrometry studies. We thank Laura Molero (Dept. of Cell Biology, Physiology and Immunology; IMIBIC/Reina Sofia University Hospital/University of Cordoba, CIBEROBN, Spain) for her technical assistance.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Alfonso Leal-Cerro or María M. Malagón.

Ethics declarations

Informed consent was obtained from all individual participants included in the study, which was approved by the Hospital’s Ethical Committee. All reported investigations were carried out in accordance with the principles of the Declaration of Helsinki.

Conflict of Interest

The authors declare that they have no competing interests.

Financial Support

This work was funded by MINECO/FEDER (BFU2010-17116; BFU2013-44229-R), J. Andalucia/FEDER (PI-0269/2008; CTS-03039; CTS-6606), and CIBERobn (Instituto de Salud Carlos III), Spain.

Electronic Supplementary Material

Below is the link to the electronic supplementary material.

Supplementary Figure 1

Uncropped Ponceau images for the different proteins measured by western blotting. (A) DLDH, (B) TCPB and VEGF, (C) AL1A1, (D) SYWC, (E) PGC-1α and JNK, (F) perilipin, (G) IMMT, Adiponectin and p-JNK, and (H) vimentin. (GIF 23 kb)

High resolution image (TIF 421 kb)

Supplementary Figure 2

Decyder software outputs showing the relative abundance of the spots in relation to the internal standard. Graphs show quantitative data of 30 differentially expressed proteins (p < 0.05) identified by the 2D-DIGE analysis in the multicomparative analysis (Suppl. Table 2). TRFE (serotransferrin), PCKGM (Phosphoenolpyruvate carboxykinase), A1BG (Alpha-1B-glycoprotein), FIBB (Fibrinogen beta), CATA (Catalase), TUBB5 (Tubulin beta), PLCD1 (Phosphodiesterase delta-1), AMPL (Cytosol aminopeptidase), ANT3 (Serpin C1), A2MG (Alpha-2-macroglobulin), ANXA6 (Annexin A6), BLVRB (Flavin reductase), ALBU (Serum albumin), IGHM (Ig mu chain C region), IGHG4 (Ig gamma-4 chain C region), KCRB (Creatine kinase B-type), PLIN1 (Perilipin-1), VAT1 (Synaptic vesicle membrane 1), PRDX6 (Peroxiredoxin-6), CH60 (60 kDa heat shock protein), FIBG (Fibrinogen gamma), ANXA1 (Annexin A1), UGPA (Uridylyl transferase), ACOT1 (Acyl-coenzyme A thioesterase), CRYAB (Alpha-crystallin B), HSPB1 (Heat shock protein beta-1), HS71L (Heat shock 70 kDa), SODM (Superoxide dismutase mitochondrial), GRP78 (78 kDa glucose-regulated protein), SEPT11 (Septin 11). (GIF 176 kb)

High resolution image (TIF 726 kb)

Supplementary Figure 3

Metabolic pathway analysis and chart of biological processes of the 37 differentially expressed proteins identified by 2D-DIGE in SAT of NG vs. IR obese subjects at PRE- and POST-BS (see Table 2). (A) Metabolic pathway analysis of the proteomic data by IPA allowed for the identification of one statistically significant interaction map corresponding to Cell Death and Survival, Carbohydrate Metabolism and Inflammatory Disease. The proteins that were identified to belong to this pathway are labeled in orange. (B) Chart of biological processes of the proteins identified by 2D-DIGE. Classification of the identified proteins was performed using PANTHER. Proteins related to metabolic process, cellular process, and immune system process were significantly regulated as determined by the Bonferroni correction for multiple testing used in the calculation of PANTHER p values. Significant differences were considered at p < 0.05. (C) Comparison of human adipose tissue proteome data sets. The colored circles represent lists of proteins differentially expressed (both up and down) between groups with p < 0.05. The resulting protein groups represent proteins differentially expressed between all obese participants Post-BS vs. Pre-BS (pink), IR vs. NG obese women at Pre-BS (purple), NG Post-BS vs. Pre-BS (green), IR Post-BS vs. Pre-BS (yellow), and IR vs. NG at Post-BS (blue). The Venn diagram comparison indicates proteins whose expression changes are shared by the different comparison groups. The overlapping regions indicate the protein expression changes induced by different conditions. (GIF 185 kb)

High resolution image (TIF 819 kb)

Supplementary Table 1

Antibodies used for Western blot studies. (DOCX 16 kb)

Supplementary Table 2

Proteins identified by MALDI-TOF/TOF differentially expressed between normoglycemic (NG) and insulin resistant (IR) women in subcutaneous adipose tissue before (PRE) and after (POST) bariatric surgery (BS). (DOCX 49 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Moreno-Castellanos, N., Guzmán-Ruiz, R., Cano, D.A. et al. The Effects of Bariatric Surgery-Induced Weight Loss on Adipose Tissue in Morbidly Obese Women Depends on the Initial Metabolic Status. OBES SURG 26, 1757–1767 (2016). https://doi.org/10.1007/s11695-015-1995-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11695-015-1995-x

Keywords

Navigation