Skip to main content

Advertisement

Log in

Modulation of PPAR-γ, SREBP-1c and inflammatory mediators by luteolin ameliorates β-cell dysfunction and renal damage in a rat model of type-2 diabetes mellitus

  • Original Article
  • Published:
Molecular Biology Reports Aims and scope Submit manuscript

Abstract

Background

Natural products have been recommended as a complementary therapy for type 2 diabetes mellitus (T2DM) due to constraints of safety and tolerability of existing anti-diabetic agents. Luteolin exhibits anti-diabetic and anti-inflammatory effects. Hence, the impact of luteolin on glucose homoeostasis and organ damage was investigated in high-fat diet (HFD) and streptozotocin (STZ) induced T2DM in rats.

Methods and results

Male Wistar rats were maintained on HFD (provided 55% energy as fat) for 10 days. Subsequently, a single dose of 40 mg/kg STZ was injected intraperitoneally on the 11th day. Seventy-two hours after STZ administration, diabetic rats with established hyperglycemia (fasting serum glucose > 200 mg/dL) were randomized into different groups having six rats each and orally administered either 0.5% hydroxy propyl cellulose or pioglitazone (10 mg/kg) or luteolin (50 mg/kg or 100 mg/kg) once daily for 28 days, while continuing HFD for respective groups. Luteolin significantly reduced hyperglycaemia, homoeostasis model assessment (HOMA) of insulin resistance (HOMA-IR) levels, and improved hypoinsulinemia and HOMA of b-cell function (HOMA-B) in a dose-dependent manner. Increased TNF-α, IL-6 and NFκB levels in diabetic rats were significantly regulated. Additionally, luteolin significantly augmented PPAR-γ expression while attenuating sterol regulatory element binding protein-1c (SREBP-1c) expression. Histopathological scrutiny validated that luteolin effectively attenuated HFD-STZ-induced injury in pancreatic β-cells and kidneys to near normalcy.

Conclusion

Our study showed that luteolin ameliorated hyperglycemia and improved hypoinsulinemia, β-cell dysfunction, and renal impairment in HFD-STZ-induced diabetic rats by attenuating inflammation and dysregulated cytokine secretion through modulation of PPAR-γ, TNF-α, IL-6 and NF-kB expression and down-regulation of SREBP-1c.

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

Similar content being viewed by others

Data availability

The data generated during the current study are available from the corresponding author upon reasonable request.

Abbreviations

T2DM:

Type 2 diabetes mellitus

HFD:

High-fat diet

STZ:

Streptozotocin

HOMA:

Homoeostasis model assessment

HOMA-IR:

Homoeostasis model assessment of insulin resistance

HOMA-B:

Homoeostasis model assessment of β-cell function

IR:

Insulin resistance

TNF-α:

Tumor necrosis factor-α

IL-6:

Interleukin-6

NFκB:

Nuclear factor-κB

PPAR-γ:

Peroxisome proliferator-activated receptor-γ

SREBP-1c:

Sterol regulatory element binding protein-1c

HPC:

Hydroxy propyl cellulose

QUICKI:

Quantitative Insulin Sensitivity Check Index

References

  1. Saeedi P, Petersohn I, Salpea P et al (2019) Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract 157:107843. https://doi.org/10.1016/j.diabres.2019.107843

    Article  PubMed  Google Scholar 

  2. Rochlani Y, Pothineni NV, Kovelamudi S, Mehta JL (2017) Metabolic syndrome: pathophysiology, management, and modulation by natural compounds. Ther Adv Cardiovasc Dis 11:215–225. https://doi.org/10.1177/1753944717711379

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Wellen KE (2005) Inflammation, stress, and diabetes. J Clin Investig 115:1111–1119. https://doi.org/10.1172/JCI200525102

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ricote M, Glass CK (2007) PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta 1771:926–935. https://doi.org/10.1016/j.bbalip.2007.02.013

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Ferré P, Foufelle F (2007) SREBP-1c transcription factor and lipid homeostasis: clinical perspective. Horm Res 68:72–82. https://doi.org/10.1159/000100426

    Article  CAS  PubMed  Google Scholar 

  6. Luo Y, Shang P, Li D (2017) Luteolin: a flavonoid that has multiple cardio-protective effects and its molecular mechanisms. Front Pharmacol 8:1–10. https://doi.org/10.3389/fphar.2017.00692

    Article  CAS  Google Scholar 

  7. Aziz N, Kim MY, Cho JY (2018) Anti-inflammatory effects of luteolin: a review of in vitro, in vivo, and in silico studies. J Ethnopharmacol 225:342–358. https://doi.org/10.1016/j.jep.2018.05.019

    Article  CAS  PubMed  Google Scholar 

  8. El-Bassossy HM, Abo-Warda SM, Fahmy A (2014) Chrysin and luteolin alleviate vascular complications associated with insulin resistance mainly through PPAR-γ activation. Am J Chin Med 42:1153–1167. https://doi.org/10.1142/S0192415X14500724

    Article  CAS  PubMed  Google Scholar 

  9. Li L, Luo W, Qian Y et al (2019) Luteolin protects against diabetic cardiomyopathy by inhibiting NF-κB-mediated inflammation and activating the Nrf2-mediated antioxidant responses. Phytomedicine. https://doi.org/10.1016/j.phymed.2018.11.034

    Article  PubMed  PubMed Central  Google Scholar 

  10. Wang GG, Lu XH, Li W et al (2011) Protective effects of luteolin on diabetic nephropathy in STZ-induced diabetic rats. Evid Based Complement Altern Med. https://doi.org/10.1155/2011/323171

    Article  Google Scholar 

  11. Lu HE, Chen Y, Sun XB et al (2015) Effects of luteolin on retinal oxidative stress and inflammation in diabetes. RSC Adv 5:4898–4904. https://doi.org/10.1039/c4ra10756j

    Article  CAS  Google Scholar 

  12. Gheibi S, Kashfi K, Ghasemi A (2017) A practical guide for induction of type-2 diabetes in rat: incorporating a high-fat diet and streptozotocin. Biomed Pharmacother 95:605–613. https://doi.org/10.1016/J.BIOPHA.2017.08.098

    Article  CAS  PubMed  Google Scholar 

  13. Srinivasan K, Viswanad B, Asrat L et al (2005) Combination of high-fat diet-fed and low-dose streptozotocin-treated rat: a model for type 2 diabetes and pharmacological screening. Pharmacol Res 52:313–320. https://doi.org/10.1016/j.phrs.2005.05.004

    Article  CAS  PubMed  Google Scholar 

  14. Vital P, Larrieta E, Hiriart M (2006) Sexual dimorphism in insulin sensitivity and susceptibility to develop diabetes in rats. J Endocrinol 190:425–432. https://doi.org/10.1677/joe.1.06596

    Article  CAS  PubMed  Google Scholar 

  15. Shehnaz SI, Roy A, Vijayaraghavan R, Sivanesan S (2023) Luteolin mitigates diabetic dyslipidemia in rats by modulating ACAT-2, PPARα, SREBP-2 proteins, and oxidative stress. Appl Biochem Biotechnol 195:4893–4914. https://doi.org/10.1007/s12010-023-04544-4

    Article  CAS  PubMed  Google Scholar 

  16. Albasher G (2020) Modulation of reproductive dysfunctions associated with streptozocin-induced diabetes by Artemisia judaica extract in rats fed a high-fat diet. Mol Biol Rep 47:7517–7527. https://doi.org/10.1007/s11033-020-05814-8

    Article  CAS  PubMed  Google Scholar 

  17. Sharma AK, Bharti S, Ojha S et al (2011) Up-regulation of PPARγ, heat shock protein-27 and -72 by naringin attenuates insulin resistance, β-cell dysfunction, hepatic steatosis and kidney damage in a rat model of type 2 diabetes. Br J Nutr 106:1713–1723. https://doi.org/10.1017/S000711451100225X

    Article  CAS  PubMed  Google Scholar 

  18. Abu-Elsaad N, El-Karef A (2018) The falconoid luteolin mitigates the myocardial inflammatory response induced by high-carbohydrate/high-fat diet in wistar rats. Inflammation 41:221–231. https://doi.org/10.1007/s10753-017-0680-8

    Article  CAS  PubMed  Google Scholar 

  19. Matthews DR, Hosker JP, Rudenski AS et al (1985) Homeostasis model assessment: insulin resistance and β-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28:412–419. https://doi.org/10.1007/BF00280883

    Article  CAS  PubMed  Google Scholar 

  20. Katz A, Nambi SS, Mather K et al (2000) Quantitative insulin sensitivity check index: a simple, accurate method for assessing insulin sensitivity in humans. J Clin Endocrinol Metab 85:2402–2410. https://doi.org/10.1210/jcem.85.7.6661

    Article  CAS  PubMed  Google Scholar 

  21. Bancroft J, Gamble M (2008) Theory and practice of histopathological techniques, 6th edn. Churchill Livingstone, Philadelphia

    Google Scholar 

  22. Tayeb W, Nakbi A, Trabelsi M et al (2012) Biochemical and histological evaluation of kidney damage after sub-acute exposure to 2,4-dichlorophenoxyacetic herbicide in rats: involvement of oxidative stress. Toxicol Mech Methods 22:696–704. https://doi.org/10.3109/15376516.2012.717650

    Article  CAS  PubMed  Google Scholar 

  23. Elaidy SM, Hussain MA, El-Kherbetawy MK (2018) Time-dependent therapeutic roles of nitazoxanide on high-fat diet/streptozotocin-induced diabetes in rats: effects on hepatic peroxisome proliferator-activated receptor-gamma receptors. Can J Physiol Pharmacol 96:485–497. https://doi.org/10.1139/cjpp-2017-0533

    Article  CAS  PubMed  Google Scholar 

  24. Soetikno V, Andini P, Iskandar M et al (2023) Alpha-mangosteen lessens high-fat/high-glucose diet and low-dose streptozotocin induced-hepatic manifestations in the insulin resistance rat model. Pharm Biol 61:241–248. https://doi.org/10.1080/13880209.2023.2166086

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zang Y, Igarashi K, Li YL (2016) Anti-diabetic effects of luteolin and luteolin-7-O-glucoside on KK-Ay mice. Biosci Biotechnol Biochem 80:1580–1586. https://doi.org/10.1080/09168451.2015.1116928

    Article  CAS  PubMed  Google Scholar 

  26. Liu Y, Fu X, Lan N et al (2014) Luteolin protects against high fat diet-induced cognitive deficits in obesity mice. Behav Brain Res 267:178–188. https://doi.org/10.1016/j.bbr.2014.02.040

    Article  CAS  PubMed  Google Scholar 

  27. Xu N, Zhang L, Dong J et al (2014) Low-dose diet supplement of a natural flavonoid, luteolin, ameliorates diet-induced obesity and insulin resistance in mice. Mol Nutr Food Res 58:1258–1268. https://doi.org/10.1002/mnfr.201300830

    Article  CAS  PubMed  Google Scholar 

  28. Kwon EY, Jung UJ, Park T et al (2015) Luteolin attenuates hepatic steatosis and insulin resistance through the interplay between the liver and adipose tissue in mice with diet-induced obesity. Diabetes 64:1658–1669. https://doi.org/10.2337/db14-0631

    Article  CAS  PubMed  Google Scholar 

  29. Ding L, Jin D, Chen X (2010) Luteolin enhances insulin sensitivity via activation of PPARγ transcriptional activity in adipocytes. J Nutr Biochem 21:941–947. https://doi.org/10.1016/j.jnutbio.2009.07.009

    Article  CAS  PubMed  Google Scholar 

  30. Deqiu Z, Kang L, Jiali Y et al (2011) Luteolin inhibits inflammatory response and improves insulin sensitivity in the endothelium. Biochimie 93:506–512. https://doi.org/10.1016/j.biochi.2010.11.002

    Article  CAS  PubMed  Google Scholar 

  31. Guo F, Xu S, Zhu Y et al (2020) PPARγ transcription deficiency exacerbates high-fat diet-induced adipocyte hypertrophy and insulin resistance in mice. Front Pharmacol 11:1285. https://doi.org/10.3389/fphar.2020.01285

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li B, Du P, Du Y et al (2021) Luteolin alleviates inflammation and modulates gut microbiota in ulcerative colitis rats. Life Sci 269:119008. https://doi.org/10.1016/j.lfs.2020.119008

    Article  CAS  PubMed  Google Scholar 

  33. Puhl AC, Bernardes A, Silveira RL et al (2012) Mode of peroxisome proliferator-activated receptor γ activation by luteolin. Mol Pharmacol 81:788–799. https://doi.org/10.1124/mol.111.076216

    Article  CAS  PubMed  Google Scholar 

  34. Chen L, Tian G, Tang W et al (2016) Protective effect of luteolin on streptozotocin-induced diabetic renal damage in mice via the regulation of RIP140/NF-ΚB pathway and insulin signalling pathway. J Funct Foods 22:93–100. https://doi.org/10.1016/j.jff.2016.01.023

    Article  CAS  Google Scholar 

  35. Sun D, Huang J, Zhang Z et al (2012) Luteolin limits infarct size and improves cardiac function after myocardium ischemia/reperfusion injury in diabetic rats. PLoS ONE 7:1–10. https://doi.org/10.1371/journal.pone.0033491

    Article  CAS  Google Scholar 

  36. Horton JD, Bashmakov Y, Shimomura I, Shimano H (1998) Regulation of sterol regulatory element binding proteins in livers of fasted and refed mice. Proc Natl Acad Sci USA 95:5987–5992. https://doi.org/10.1073/pnas.95.11.5987

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Liu JF, Ma Y, Wang Y et al (2011) Reduction of lipid accumulation in HepG2 cells by luteolin is associated with activation of AMPK and mitigation of oxidative stress. Phyther Res 25:588–596. https://doi.org/10.1002/ptr.3305

    Article  CAS  Google Scholar 

  38. Yin Y, Gao L, Lin H et al (2017) Luteolin improves non-alcoholic fatty liver disease in db/db mice by inhibition of liver X receptor activation to down-regulate expression of sterol regulatory element binding protein 1c. Biochem Biophys Res Commun 482:720–726. https://doi.org/10.1016/j.bbrc.2016.11.101

    Article  CAS  PubMed  Google Scholar 

  39. Lee FTH, Cao Z, Long DM et al (2004) Interactions between angiotensin II and NF-κB-dependent pathways in modulating macrophage infiltration in experimental diabetic nephropathy. J Am Soc Nephrol 15:2139–2151. https://doi.org/10.1097/01.ASN.0000135055.61833.A8

    Article  CAS  PubMed  Google Scholar 

  40. Zhang M, He L, Liu J, Zhou L (2021) Luteolin attenuates diabetic nephropathy through suppressing inflammatory response and oxidative stress by inhibiting STAT3 pathway. Exp Clin Endocrinol Diabetes 129:729–739. https://doi.org/10.1055/a-0998-7985

    Article  CAS  PubMed  Google Scholar 

  41. Albarakati AJA, Baty RS, Aljoudi AM et al (2020) Luteolin protects against lead acetate-induced nephrotoxicity through antioxidant, anti-inflammatory, anti-apoptotic, and Nrf2/HO-1 signaling pathways. Mol Biol Rep 47:2591–2603. https://doi.org/10.1007/s11033-020-05346-1

    Article  CAS  PubMed  Google Scholar 

  42. Kalbolandi SM, Gorji AV, Babaahmadi-Rezaei H, Mansouri E (2019) Luteolin confers renoprotection against ischemia–reperfusion injury via involving Nrf2 pathway and regulating miR320. Mol Biol Rep 46:4039–4047. https://doi.org/10.1007/s11033-019-04853-0

    Article  CAS  PubMed  Google Scholar 

  43. Fu Z, Gilbert ER, Liu D (2013) Regulation of insulin synthesis and secretion and pancreatic beta-cell dysfunction in diabetes. Curr Diabetes Rev 9:25–53

    Article  PubMed  PubMed Central  Google Scholar 

  44. Kim EK, Kwon KB, Song MY et al (2007) Flavonoids protect against cytokine-induced pancreatic β-cell damage through suppression of nuclear factor κb activation. Pancreas 35:1–9. https://doi.org/10.1097/mpa.0b013e31811ed0d2

    Article  Google Scholar 

  45. Ding Y, Shi X, Shuai X et al (2014) Luteolin prevents uric acid-induced pancreatic β-cell dysfunction. J Biomed Res 28:292–298. https://doi.org/10.7555/JBR.28.20130170

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Orji CE, Okpoko CK, Agbata CA et al (2020) Evaluation of the effect of luteolin on the hepatic and hematopoietic systems in albino rats. J Clin Toxicol 10:2–6. https://doi.org/10.35248/2161-0495.20.10.434

    Article  Google Scholar 

  47. Xiong J, Wang K, Yuan C et al (2017) Luteolin protects mice from severe acute pancreatitis by exerting HO-1-mediated anti-inflammatory and antioxidant effects. Int J Mol Med 39:113–125. https://doi.org/10.3892/ijmm.2016.2809

    Article  CAS  PubMed  Google Scholar 

  48. Lin LC, Pai YF, Tsai TH (2015) Isolation of luteolin and luteolin-7-O-glucoside from Dendranthema morifolium Ramat Tzvel and their pharmacokinetics in rats. J Agric Food Chem 63:7700–7706. https://doi.org/10.1021/jf505848z

    Article  CAS  PubMed  Google Scholar 

  49. Dang H, Meng MHW, Zhao H et al (2014) Luteolin-loaded solid lipid nanoparticles synthesis, characterization, & improvement of bioavailability, pharmacokinetics in vitro and vivo studies. J Nanopart Res. https://doi.org/10.1007/s11051-014-2347-9

    Article  Google Scholar 

  50. Sinha A, Suresh PK (2019) Enhanced induction of apoptosis in HaCaT cells by luteolin encapsulated in PEGylated liposomes—role of caspase-3/caspase-14. Appl Biochem Biotechnol 188:147–164. https://doi.org/10.1007/s12010-018-2907-z

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the technical support rendered by Mr. P. Praveen Kumar, Laboratory assistant of the Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai, India during the study.

Funding

The authors declare that no funds, grants, or other support were received during the preparation of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

SIS conceived and designed the experiments, guided by AR. SIS performed the experiments, guided by RV and SS. NP and SIS contributed to the analysis and interpretation of histopathological data. SIS and RV performed the statistical analysis. The first draft of the manuscript was written by SIS with contributions to the methodology section by RV, SS and NP. All authors commented on previous versions of the manuscript. All authors read and approved the final manuscript. The authors declare that all data was generated in-house and that no paper mill was used.

Corresponding author

Correspondence to Syed Ilyas Shehnaz.

Ethics declarations

Conflict of interest

The authors have no relevant financial or non-financial interests to disclose.

Ethical approval

The experiments were designed in accordance with the Committee for Control and Supervision of Experiments on Animals; Ministry of Forests, Environment and Climate Change, Government of India, and were approved by the Institutional Animal Ethics Committee (Approval No. SU/CLAR/RD/007/ 2019, dated: 21.12.2019).

Consent to participate

Not applicable.

Consent to publish

Not applicable.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shehnaz, S.I., Roy, A., Vijayaraghavan, R. et al. Modulation of PPAR-γ, SREBP-1c and inflammatory mediators by luteolin ameliorates β-cell dysfunction and renal damage in a rat model of type-2 diabetes mellitus. Mol Biol Rep 50, 9129–9142 (2023). https://doi.org/10.1007/s11033-023-08804-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11033-023-08804-8

Keywords

Navigation