Insulin/IGF-1 Signaling Is Downregulated in Barrett's Esophagus Patients Undergoing a Moderate Calorie and Protein Restriction Program: A Randomized 2-Year Trial

Diletta Arcidiacono, Alice Zaramella, Federico Fabris, Ricardo Sánchez-Rodríguez, Daniele Nucci, Matteo Fassan, Mariateresa Nardi, Clara Benna, Chiara Cristofori, Tiziana Morbin, Salvatore Pucciarelli, Alberto Fantin, Stefano Realdon, Diletta Arcidiacono, Alice Zaramella, Federico Fabris, Ricardo Sánchez-Rodríguez, Daniele Nucci, Matteo Fassan, Mariateresa Nardi, Clara Benna, Chiara Cristofori, Tiziana Morbin, Salvatore Pucciarelli, Alberto Fantin, Stefano Realdon

Abstract

Obesity and associated insulin resistance (Ins-R) have been identified as important risk factors for esophageal adenocarcinoma development. Elevated calories and protein consumption are also associated with Ins-R and glucose intolerance. We investigated the effect of a 24-month moderate calorie and protein restriction program on overweight or obese patients affected by Barrett's esophagus (BE), as no similar dietary approach has been attempted to date in this disease context. Anthropometric parameters, levels of serum analytes related to obesity and Ins-R, and the esophageal insulin/IGF-1 signaling pathway were analyzed. This study is registered with ClinicalTrials.gov, number NCT03813381. Insulin, C-peptide, IGF-1, IGF-binding protein 3 (IGFBP3), adipokines, and esophageal expression of the main proteins involved in insulin/IGF-1 signal transduction were quantified using Luminex-XMAP® technology in 46 patients who followed the restriction program (IA) and in 54 controls (CA). Body mass index and waist circumference significantly decreased in 76.1% of IA and 35.2% of CA. IGF-1 levels were reduced in 71.7% of IA and 51.8% of CA. The simultaneous reduction of glycaemia, IGF-1, the IGF-1/IGFBP3 ratio, and the improvement in weight loss-dependent insulin sensitivity, were associated with the downregulation of the insulin/IGF-1 signal on BE tissue. The proposed intervention program was an effective approach to counteract obesity-associated cancer risk factors. The improvement in metabolic condition resulted in a downregulation of the ERK-mediated mitogenic signal in 43.5% of patients, probably affecting the molecular mechanism driving adenocarcinoma development in BE lesions.

Keywords: Barrett’s esophagus; calorie–protein restriction; esophageal adenocarcinoma prevention; insulin/IGF-1 signal transduction; obesity.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
The trial’s CONSORT flow diagram.
Figure 2
Figure 2
Waist circumference variation (ΔWC) and BMI (ΔBMI) in the control arm (CA) and intervention arm (IA) at the end of the 24 months of the intervention program. (a) ΔWC and (b) ΔBMI were calculated as the difference between the value measured at T24 and the baseline value. The dashed red lines (ΔWC = −4 cm and ΔBMI = −1.11 kg/m2) show the cutoffs between the two arms, which were mathematically corresponding to the 25th percentile of the whole data distribution. Each square represents a single patient from the CA and each triangle represents a single patient from the IA.
Figure 3
Figure 3
Total protein expression involved in insulin/IGF-1 signal transduction in esophageal tissue affected by Barrett’s esophagus. Protein expression was analyzed at baseline (T0, white boxes) and 24 months later (T24, black boxes) in both the control arm (CA) and intervention arm (IA). Data are expressed as the median (Q1; Q3) related to an arbitrary unit (A.U.), defined as described in the Materials and Methods Section. The Wilcoxon matched-pairs signed-rank test was used to compare measurements at T0 and T24 within the same group. The Mann–Whitney U test was performed to evaluate differences between the CA and IA at baseline. ** p < 0.01.
Figure 4
Figure 4
Main protein phosphorylation levels in insulin/IGF-1 signal transduction in esophageal tissue affected by Barrett’s esophagus. Protein expression was analyzed at baseline (T0) and 24 months later (T24) in both the control arm (CA) and the intervention arm (IA). Data are expressed as the median (Q1; Q3) of relative phosphorylation levels related to an arbitrary unit (A.U.) defined as described in the Materials and Methods Section. Each symbol represents a different data set: ● CA at T0, ∎ CA at T24, ▲ IA at T0, ▼IA at T24. The Wilcoxon matched-pairs signed-rank test was used to compare measurements at T0 and T24 within the same group. The Mann–Whitney U test was performed to evaluate differences in baseline measurements between the CA and IA. * p < 0.05.
Figure 5
Figure 5
Identification of different patient subpopulations based on the modulation of IRS1 expression after 24 months from the start of the intervention program. Patients were divided on the basis of the downregulation (lower) or upregulation (higher) of IRS1 expression at the end of the intervention program compared to the basal expression at the time of enrollment. Within the lower IRS1 subpopulation, patients who had improved glycaemic control and patients who had unchanged or worsened glycaemic control were analyzed separately. For each group of patients, data regarding anthropometric parameters, serum analytes, total expression, and the relative phosphorylation level of proteins extracted from Barrett’s lesion were reported. Data indicate the median variation (T24−T0) of the parameter considered, followed by a p-value (the Wilcoxon matched-pairs signed-rank test was used to compare measurements at T0 and T24 within the same group). A reduction in protein levels was indicated as the median of the variation expressed as a percentage (%). An increase in protein level was indicated as n-fold of the amount detected at baseline. Parameters not included in this diagram were not significantly modified. 1 The Fisher’s exact test was performed to assess differences between the proportions of patients belonging to the two arms within each subpopulation identified. The up/down arrow indicates an increase/decrease in the parameter’s measure at 24 months compared to baseline. The green arrow indicates a positive change (protective against cancer evolution) in the parameter considered. The red arrow indicates a negative change (procancer) in the parameter considered.

References

    1. PwC Chronic Diseases and Conditions Are on the Rise. [(accessed on 11 October 2021)]. Available online: .
    1. De Pergola G., Silvestris F. Obesity as a Major Risk Factor for Cancer. J. Obes. 2013;2013:2915. doi: 10.1155/2013/291546.
    1. Teng J.A., Wu S.G., Chen J.X., Li Q., Peng F., Zhu Z., Qin J., He Z.Y. The Activation of ERK1/2 and JNK MAPK Signaling by Insulin/IGF-1 Is Responsible for the Development of Colon Cancer with Type 2 Diabetes Mellitus. PLoS ONE. 2016;11:e0149822. doi: 10.1371/journal.pone.0149822.
    1. Zhu N., Zhang Y., Gong Y.I., He J., Chen X. Metformin and Lung Cancer Risk of Patients with Type 2 Diabetes Mellitus: A Meta-Analysis. Biomed. Rep. 2015;3:235–241. doi: 10.3892/br.2015.417.
    1. Arcidiacono D., Antonello A., Fassan M., Nucci D., Morbin T., Agostini M., Nitti D., Rugge M., Alberti A., Battaglia G., et al. Insulin Promotes HER2 Signaling Activation during Barrett’s Esophagus Carcinogenesis. Dig. Liver Dis. 2017;49:630–638. doi: 10.1016/j.dld.2017.01.154.
    1. Elliott J., Reynolds J. Visceral Obesity, Metabolic Syndrome, and Esophageal Adenocarcinoma. Front. Oncol. 2021;11:627270. doi: 10.3389/fonc.2021.627270.
    1. Long E., Beales I.L.P. The Role of Obesity in Oesophageal Cancer Development. Therap. Adv. Gastroenterol. 2014;7:247–268. doi: 10.1177/1756283X14538689.
    1. Shaheen N.J., Richter J.E. Barrett’s Oesophagus. Lancet. 2009;373:850–861. doi: 10.1016/S0140-6736(09)60487-6.
    1. Siegel R., Naishadham D., Jemal A. Cancer Statistics, 2013. CA Cancer J. Clin. 2013;63:11–30. doi: 10.3322/caac.21166.
    1. Howlader N., Noone A., Krapcho M., Miller D., Brest A., Yu M., Ruhl J., Tatalovich Z., Mariotto A., Lewis D., et al. Cancer Statistics Review 1975–2018—SEER Statistics. [(accessed on 17 August 2021)]; Available online:
    1. Gordon L.G., Mayne G.C., Hirst N.G., Bright T., Whiteman D.C., Australian Cancer Study Clinical Follow-Up. Watson D.I. Cost-Effectiveness of Endoscopic Surveillance of Non-Dysplastic Barrett’s Esophagus. Gastrointest. Endosc. 2014;79:242–256.e6. doi: 10.1016/j.gie.2013.07.046.
    1. Spechler S.J. Barrett Esophagus and Risk of Esophageal Cancer: A Clinical Review. JAMA. 2013;310:627–636. doi: 10.1001/jama.2013.226450.
    1. Shaheen N.J. Risk of Cancer in Patients With Barrett Esophagus. Gastroenterol. Hepatol. 2019;15:688.
    1. World Cancer Research Fund and American Institute for Cancer Research . Food, Nutrition, Physical Activity and the Prevention of Cancer: A Global Perspective. World Cancer Research Fund and American Institute for Cancer Research; Washington, DC, USA: 2007.
    1. Romaguera D., Vergnaud A.C., Peeters P.H., Van Gils C.H., Chan D.S.M., Ferrari P., Romieu I., Jenab M., Slimani N., Clavel-Chapelon F., et al. Is Concordance with World Cancer Research Fund/American Institute for Cancer Research Guidelines for Cancer Prevention Related to Subsequent Risk of Cancer? Results from the EPIC Study. Am. J. Clin. Nutr. 2012;96:150–163. doi: 10.3945/ajcn.111.031674.
    1. Gallagher E., LeRoith D. Minireview: IGF, Insulin, and Cancer. Endocrinology. 2011;152:2546–2551. doi: 10.1210/en.2011-0231.
    1. Baker J., Liu J.P., Robertson E.J., Efstratiadis A. Role of Insulin-like Growth Factors in Embryonic and Postnatal Growth. Cell. 1993;75:73–82. doi: 10.1016/S0092-8674(05)80085-6.
    1. Clemmons D. Metabolic Actions of Insulin-like Growth Factor-I in Normal Physiology and Diabetes. Endocrinol. Metab. Clin. North Am. 2012;41:425–443. doi: 10.1016/j.ecl.2012.04.017.
    1. LeRoith D., Werner H., Beitner-Johnson D., Roberts C. Molecular and Cellular Aspects of the Insulin-like Growth Factor I Receptor. Endocr. Rev. 1995;16:143–163. doi: 10.1210/edrv-16-2-143.
    1. Baserga R., Peruzzi F., Reiss K. The IGF-1 Receptor in Cancer Biology. Int. J. Cancer. 2003;107:873–877. doi: 10.1002/ijc.11487.
    1. Guerard M., Robin T., Perron P., Hatat A., David-Boudet L., Vanwonterghem L., Busser B., Coll J., Lantuejoul S., Eymin B., et al. Nuclear Translocation of IGF1R by Intracellular Amphiregulin Contributes to the Resistance of Lung Tumour Cells to EGFR-TKI. Cancer Lett. 2018;420:146–155. doi: 10.1016/j.canlet.2018.01.080.
    1. Arcidiacono D., Dedja A., Giacometti C., Fassan M., Nucci D., Francia S., Fabris F., Zaramella A., Gallagher E., Cassaro M., et al. Hyperinsulinemia Promotes Esophageal Cancer Development in a Surgically-Induced Duodeno-Esophageal Reflux Murine Model. Int. J. Mol. Sci. 2018;19:1198. doi: 10.3390/ijms19041198.
    1. Fontana L., Adelaiye R.M., Rastelli A.L., Miles K.M., Ciamporcero E., Longo V.D., Nguyen H., Vessella R., Pili R. Dietary Protein Restriction Inhibits Tumor Growth in Human Xenograft Models. Oncotarget. 2013;4:2451–2461. doi: 10.18632/oncotarget.1586.
    1. Speakman J.R., Mitchell S.E. Caloric Restriction. Mol. Asp. Med. 2011;32:159–221. doi: 10.1016/j.mam.2011.07.001.
    1. Weusten B., Bisschops R., Coron E., Dinis-Ribeiro M., Dumonceau J.-M., Esteban J.-M., Hassan C., Pech O., Repici A., Bergman J., et al. Endoscopic Management of Barrett’s Esophagus: European Society of Gastrointestinal Endoscopy (ESGE) Position Statement. Endoscopy. 2017;49:191–198. doi: 10.1055/s-0042-122140.
    1. Roza A.M., Shizgal H.M. The Harris Benedict Equation Reevaluated: Resting Energy Requirements and the Body Cell Mass. Am. J. Clin. Nutr. 1984;40:168–182. doi: 10.1093/ajcn/40.1.168.
    1. Harvard School of Public Health Now Being Served, Better Nutrition Advice. Our Healthy Eating Plate Alternative to the Government’s MyPlate—PubMed. Harv. Health Lett. 2011;37:4–5.
    1. Diabetes Prevention Program Research Group 10-Year Follow-up of Diabetes Incidence and Weight Loss in the Diabetes Prevention Program Outcomes Study. Lancet. 2009;374:1677–1686. doi: 10.1016/S0140-6736(09)61457-4.
    1. Anderson A.S., Craigie A.M., Caswell S., Treweek S., Stead M., Macleod M., Daly F., Belch J., Rodger J., Kirk A., et al. The Impact of a Bodyweight and Physical Activity Intervention (BeWEL) Initiated through a National Colorectal Cancer Screening Programme: Randomised Controlled Trial. BMJ. 2014;348:g1823. doi: 10.1136/bmj.g1823.
    1. Matthews D.R., Hosker J.P., Rudenski A.S., Naylor B.A., Treacher D.F., Turner R.C. Homeostasis Model Assessment: Insulin Resistance and β-Cell Function from Fasting Plasma Glucose and Insulin Concentrations in Man. Diabetologia. 1985;28:412–419. doi: 10.1007/BF00280883.
    1. Capasso I., Esposito E., Pentimalli F., Montella M., Crispo A., Maurea N., D’Aiuto M., Fucito A., Grimaldi M., Cavalcanti E., et al. Homeostasis Model Assessment to Detect Insulin Resistance and Identify Patients at High Risk of Breast Cancer Development: National Cancer Institute of Naples Experience. J. Exp. Clin. Cancer Res. 2013;32:14. doi: 10.1186/1756-9966-32-14.
    1. International Diabetes Federation (IDF) Global Guideline for Type 2 Diabetes. [(accessed on 2 July 2021)]. Available online: .
    1. Kawai T., Autieri M.V., Scalia R. Adipose Tissue Inflammation and Metabolic Dysfunction in Obesity. Am. J. Physiol. Cell Physiol. 2021;320:C375–C391. doi: 10.1152/ajpcell.00379.2020.
    1. Donohoe C.L., Ofarrell N.J., Doyle S.L., Reynolds J.V. The Role of Obesity in Gastrointestinal Cancer: Evidence and Opinion. Therap. Adv. Gastroenterol. 2014;7:38–50. doi: 10.1177/1756283X13501786.
    1. Marco-Benedí V., Pérez-Calahorra S., Bea A.M., Lamiquiz-Moneo I., Baila-Rueda L., Cenarro A., Civeira F., Mateo-Gallego R. High-Protein Energy-Restricted Diets Induce Greater Improvement in Glucose Homeostasis but Not in Adipokines Comparing to Standard-Protein Diets in Early-Onset Diabetic Adults with Overweight or Obesity. Clin. Nutr. 2020;39:1354–1363. doi: 10.1016/j.clnu.2019.06.005.
    1. Gautier-Stein A., Rajas F., Mithieux G. Intestinal Gluconeogenesis and Protein Diet: Future Directions. Proc. Nutr. Soc. 2021;80:118–125. doi: 10.1017/S0029665120007922.
    1. Tian S., Xu Q., Jiang R., Han T., Sun C., Na L. Dietary Protein Consumption and the Risk of Type 2 Diabetes: A Systematic Review and Meta-Analysis of Cohort Studies. Nutrients. 2017;9:982. doi: 10.3390/nu9090982.
    1. Fontana L., Weiss E.P., Villareal D.T., Klein S., Holloszy J.O. Long-Term Effects of Calorie or Protein Restriction on Serum IGF-1 and IGFBP-3 Concentration in Humans. Aging Cell. 2008;7:681. doi: 10.1111/j.1474-9726.2008.00417.x.
    1. Zick Y. Ser/Thr Phosphorylation of IRS Proteins: A Molecular Basis for Insulin Resistance. Sci. STKE. 2005;2005:pe4. doi: 10.1126/stke.2682005pe4.
    1. Bouskila M., Hunter R.W., Ibrahim A.F.M., Delattre L., Peggie M., Van Diepen J.A., Voshol P.J., Jensen J., Sakamoto K. Allosteric Regulation of Glycogen Synthase Controls Glycogen Synthesis in Muscle. Cell Metab. 2010;12:456–466. doi: 10.1016/j.cmet.2010.10.006.
    1. Huang J., Manning B.D. The TSC1–TSC2 Complex: A Molecular Switchboard Controlling Cell Growth. Biochem. J. 2008;412:179. doi: 10.1042/BJ20080281.
    1. Ma L., Chen Z., Erdjument-Bromage H., Tempst P., Pandolfi P.P. Phosphorylation and Functional Inactivation of TSC2 by Erk: Implications for Tuberous Sclerosisand Cancer Pathogenesis. Cell. 2005;121:179–193. doi: 10.1016/j.cell.2005.02.031.
    1. Um S.H., D’Alessio D., Thomas G. Nutrient Overload, Insulin Resistance, and Ribosomal Protein S6 Kinase 1, S6K1. Cell Metab. 2006;3:393–402. doi: 10.1016/j.cmet.2006.05.003.
    1. Cai Q., Dozmorov M., Oh Y. IGFBP-3/IGFBP-3 Receptor System as an Anti-Tumor and Anti-Metastatic Signaling in Cancer. Cells. 2020;9:1261. doi: 10.3390/cells9051261.

Source: PubMed

3
Tilaa