Skip to main content

RETRACTED ARTICLE: Progranulin causes adipose insulin resistance via increased autophagy resulting from activated oxidative stress and endoplasmic reticulum stress

This article was retracted on 13 December 2022

This article has been updated

Abstract

Background

Progranulin (PGRN) has recently emerged as an important regulator for insulin resistance. However, the direct effect of progranulin in adipose insulin resistance associated with the autophagy mechanism is not fully understood.

Methods

In the present study, progranulin was administered to 3T3-L1 adipocytes and C57BL/6 J mice with/without specific inhibitors of oxidative stress and endoplasmic reticulum stress, and metabolic parameters, oxidative stress, endoplasmic reticulum stress and autophagy markers were assessed.

Results

Progranulin treatment increased iNOS expression, NO synthesis and ROS generation, and elevated protein expressions of CHOP, GRP78 and the phosphorylation of PERK, and caused a significant increase in Atg7 and LC3-II protein expression and a decreased p62 expression, and decreased insulin-stimulated tyrosine phosphorylation of IRS-1 and glucose uptake, demonstrating that progranulin activated oxidative stress and ER stress, elevated autophagy and induced insulin insensitivity in adipocytes and adipose tissue of mice. Interestingly, inhibition of iNOS and ER stress both reversed progranulin-induced stress response and increased autophagy, protecting against insulin resistance in adipocytes. Furthermore, the administration of the ER stress inhibitor 4-phenyl butyric acid reversed the negative effect of progranulin in vivo.

Conclusion

Our findings showed the clinical potential of the novel adipokine progranulin in the regulation of insulin resistance, suggesting that progranulin might mediate adipose insulin resistance, at least in part, by inducing autophagy via activated oxidative stress and ER stress.

Background

Progranulin (PGRN), as an autocrine growth factor, plays a key role on a variety of physiological and pathological processes, including inflammation, glucose and lipid metabolism and so on [1, 2]. A growing body of evidence indicated that progranulin could emerge as an important regulator for insulin resistance. These studies showed that progranulin knockout mice prevented from diet-induced obesity and insulin resistance via the regulation of inflammation, while progranulin treatment in adipocytes resulted in insulin insensitivity [3]. Recently, our results also found that administration of progranulin caused glucose intolerance and insulin insensitivity through triggering autophagy in adipose tissue of mice [4], suggesting that progranulin could be a critical adipokine regulating glucose and lipid metabolism. Although the potential role of progranulin in activating autophagy and inducing insulin resistance has been identified, the intracellular events responsible for progranulin-mediated effects in autophagy and insulin resistance remain not fully understood.

Recently it has been proved that oxidative stress could cause ER stress, which is a known inducer of autophagy [5, 6], and the strong association among autophagy, endoplasmic reticulum (ER) stress and oxidative stress in a variety of physiological and pathological processes in kinds of cell types has been identified. For example, the development of cardiomyoblast death was found to result from activated ER stress, and elevated levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) production via triggering autophagy [7]. Additionally, diet-induced obese mice exhibited activated ER stress and increased autophagy, leading to developing insulin resistance [8]. On the basis of these findings, we postulated that progranulin may cause adipose insulin resistance via increased autophagy, resulting from activated ER stress and oxidative stress.

In the present studies, we provided the evidence that the administration of progranulin activated ER stress and oxidative stress, elevated autophagy and induced insulin insensitivity in adipocytes. We also assessed the potential intracellular signalings required for progranulin-mediated insulin resistance. These results supported the hypothesis that progranulin aggravated insulin resistance through increased autophagy, resulting from activated ER stress and oxidative stress, suggesting the significance of the novel adipokine progranulin in the regulation of glucose and lipid metabolism.

Methods

Materials

Chemicals of analytical grade were purchased from Sigma (St Louis, MO, USA) except where stated otherwise. The following antibodies were used: anti-Atg7, anti-p62, anti-LC3 (light chain 3), anti-iNOS (inducible NO synthase), anti-CHOP (C/EBP homologous protein), anti-GRP78 (guaninenucleotide-releasing protein 78), anti-p-PERK (PKR-like ER kinase; Thr980) and anti-PERK (all from Cell Signaling Technology, Danvers, MA, USA); and anti-IRS-1 (insulin receptor substrate 1), anti-pY20, anti-GAPDH, peroxidase-conjugated goat anti-rabbit IgG and peroxidase-conjugated goat anti-mouse IgG (all from Santa Cruz Biotechnology, Santa Cruz, CA, USA).

Cell culture, differentiation and treatment

Mouse 3T3-L1 cells were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in Dulbecco’s modified Eagle’s medium with 10% fetal bovine serum (HyClone, Thermo Fisher Scientific, Logan, UT, USA). Induction medium containing 3T3-L1 cells was used for the differentiation of mature fat cells, with differentiation usually being complete by the 8th day. The effects of progranulin were determined by treating cells with 100 ng/ml progranulin for 20 h. Insulin signaling in the cells was stimulated by applying 10 nM insulin for 10 min. The medium was replaced with fresh medium before each experiment.

Animal care

This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Committee on the Ethics of Animal Experiments of Medical School of Xi’an Jiaotong University (Permit number: 2013–025). For the in vivo study, C57BL/6J male mice (8 weeks old) were fed with a normal chow diet and housed under standard conditions with a 12 h light:12 h darkness cycle (darkness from 19:30 to 07:30). Mice were distributed in four groups (n = 10/group): i) vehicle (normal saline solution); ii) 4-phenyl butyric acid (4-PBA i.p. 1 mg/g, once a day); iii) progranulin (i.p. 1 mg/g, once a day); iv) progranulin (i.p. 1 mg/g, once a day) + 4-PBA (i.p. 1 mg/g, once a day). The treatment lasted 21 days. At the end of the 21-day study period, half of the mice in each group were randomly selected and received an intraperitoneal injection of insulin at a dosage of 2 IU/kg; 15 min after the injection, the animals were euthanized, and their omental adipose tissues and blood samples were obtained and stored at −80 °C for subsequent analysis.

Western blotting

The tissues and cells that were subjected to various treatments were lysed in lysis buffer containing 25 mM Tris HCl (pH 6.8), 2% sodium dodecyl sulfate, 6% glycerol, 1% 2-mercaptoethanol, 2 mM phenylmethylsulfonyl fluoride, 0.2% bromophenol blue and a protease inhibitor cocktail for 20 min. Western blotting was performed in accordance with a standard protocol [9].

Immunoprecipitation

Cytoplasmic lysate (200 μg) was incubated for 2 h at 4 °C with the corresponding antibodies coupled to 20 μl of packed protein A + G sepharose beads (Beyotime, Jiangsu, China). Immunocomplexes were resolved by means of sodium dodecyl sulfate-polyacrylamide gel electrophoresis and immunoblotted with the indicated antibodies.

Measurement of nitrite concentration

Nitrite concentrations in the culture media were measured by the Total Nitric Oxide Assay kit (Assay Designs, Ann Arbor, MI) according to the manufacturer’s instructions.

Measurement of ROS levels

Intracellular ROS generation was measured by flow cytometry using DCFH2-DA. For measurement of intracellular ROS levels, cells were incubated with 2.5 μmol/ ml DCFH2-DA at 37 °C for 30 min. The increase in DCFH2-DA oxidation was measured by a flow cytometry. Fluorescence was measured at an excitation wavelength of 488 nm and an emission wavelength of 530 nm.

Glucose uptake

After transfer of 3T3L1 cells to medium without glucose, mouse adipocytes were incubated with 10 nmol/l insulin for 15 min, when glucose transport was determined as uptake of 50 mmol/l (10 mCi/ml) 2-deoxy-D-[1-3H] glucose, and then incubated 30 min. Uptake was linear for at least 30 min.

Measurement of blood parameters

Glucose tolerance testing (GTT) and insulin tolerance testing (ITT) were performed by utilizing a standard protocol as described [10]. Glucose tolerance testing (GTT) was performed after the mice were fasted overnight. A total of 2 g/kg glucose was administrated through an i.p. injection, and blood glucose was measured at the indicated time points. Insulin tolerance testing (ITT) was performed after the animals had fasted for 4 h. Then, 0.75 U/kg insulin was administered via i.p. injection, and blood glucose was measured at the indicated time points.

Statistics

Statistical analysis was performed using SPSS 17.0 Software. Statistical analysis between the two groups was performed using unpaired, two-tailed Student t-test or ANOVA. Differences were considered significant when the P value was < 0.05.

Results

Progranulin treatment activated oxidative stress and ER stress, elevated autophagy and induced insulin insensitivity in adipocytes

To identify the potential key role of progranulin in glucose and lipid metabolism, 3T3-L1 adipocytes were cultured and pretreated with progranulin. Progranulin significantly increased iNOS expression in cultured adipocytes (Fig. 1a and b), which was associated with increased NO synthesis as measured by the nitrite concentration in the media (Fig. 1c) and elevated ROS generation (Fig. 1d). In agreement with a previous study [3, 4], progranulin also activated ER stress in adipocytes, as evident by increase in protein expression of CHOP, GRP78 and the phosphorylation of PERK (Fig. 1e and f). Additionally, progranulin caused a significant increase in Atg7 and LC3-II protein expression and a decreased p62 expression, demonstrating up-regulation of autophagy in adipocytes (Fig. 1g and h). Meanwhile, progranulin decreased insulin-stimulated tyrosine phosphorylation of IRS-1 and glucose uptake (Fig. 1i-k), leading to insulin insensitivity in adipocytes.

Fig. 1
figure 1

Progranulin treatment activated oxidative stress and ER stress, elevated autophagy and induced insulin insensitivity in adipocytes. a Protein expression of iNOS. b The relative protein quantity of iNOS. c Nitrate levels in the culture media. d Intracellular ROS levels. e Protein expression of CHOP and GRP78, and the phosphorylation of PERK. f The relative protein quantity of CHOP, GRP78 and p-PERK. g Protein expression of Atg7, p62 and LC3. h The relative protein quantity of Atg7, p62 and LC3. i IRS-1 tyrosine phosphorylation. j The relative protein quantity of IRS-1 tyrosine phosphorylation. k Glucose uptake. The relative quantity of proteins was analyzed using Quantity One software. A representative blot is shown and the data was expressed as mean ± SEM in each bar graph. *P < 0.05 (PGRN vs Control)

Inhibition of iNOS reversed progranulin-induced ER stress response and increased autophagy, preventing from insulin resistance in adipocytes

Recently, the relationship among autophagy, ER stress and oxidative stress in some physiological and pathological processes has been identified, so we reasoned that iNOS inhibition may recover progranulin-induced ER stress response and increased autophagy, preventing from insulin resistance in adipocytes. As expected, 30 μM S-methylisothiourea sulfate (SMT), an iNOS-specific inhibitor, was used to inhibit iNOS expression, NO synthesis and ROS generation (Fig. 2a-d). Moreover, SMT not only significantly decreased protein expression of CHOP, GRP78 and the phosphorylation of PERK in adipocytes treated with progranulin (Fig. 2e and f), but also increased insulin-induced IRS-1 tyrosine phosphorylation and glucose uptake (Fig. 2i-k). Furthermore, the iNOS-specific inhibitor SMT reversed progranulin-induced up-regulation of autophagy in adipocytes, as evidenced by decreased Atg7 and LC3-II protein expression levels and elevated p62 expression (Fig. 2g and h).

Fig. 2
figure 2

Inhibition of iNOS reversed progranulin-induced ER stress response and increased autophagy, preventing from insulin resistance in adipocytes. a Protein expression of iNOS. b The relative protein quantity of iNOS. c Nitrate levels in the culture media. d Intracellular ROS levels. e Protein expression of CHOP and GRP78, and the phosphorylation of PERK. f The relative protein quantity of CHOP, GRP78 and p-PERK. g Protein expression of Atg7, p62 and LC3. h The relative protein quantity of Atg7, p62 and LC3. i IRS-1 tyrosine phosphorylation. j The relative protein quantity of IRS-1 tyrosine phosphorylation. k Glucose uptake. The relative quantity of proteins was analyzed using Quantity One software. A representative blot is shown and the data was expressed as mean ± SEM in each bar graph. *P < 0.05 (PGRN vs Control); # P < 0.05 (PGRN + SMT vs PGRN)

Inhibition of ER stress reversed progranulin-induced oxidative stress response and increased autophagy, preventing from insulin resistance in adipocytes

In addition, the ER stress specific inhibitor 4-PBA was used to suppress progranulin-induced ER stress response in adipocytes. As expected, 4-PBA markedly reduced protein expression of CHOP, GRP78 and the phosphorylation of PERK in adipocytes treated with progranulin (Fig. 3e and f). Interestingly, inhibition of ER stress with 4-PBA also resulted in reduced iNOS expression, NO synthesis and ROS generation (Fig. 3a-d). Furthermore, adipocytes treated with 4-PBA in the presence of progranulin displayed recovered the abnormal levels of autophagy indicators such as Atg7, p62 and LC3-II (Fig. 3g and h), and up-regulation of insulin-induced IRS-1 tyrosine phosphorylation and glucose uptake (Fig. 3i-k).

Fig. 3
figure 3

Inhibition of ER stress reversed progranulin-induced oxidative stress response and increased autophagy, preventing from insulin resistance in adipocytes. a Protein expression of iNOS. b The relative protein quantity of iNOS. c Nitrate levels in the culture media. d Intracellular ROS levels. e Protein expression of CHOP and GRP78, and the phosphorylation of PERK. f The relative protein quantity of CHOP, GRP78 and p-PERK. g Protein expression of Atg7, p62 and LC3. h The relative protein quantity of Atg7, p62 and LC3. i IRS-1 tyrosine phosphorylation. j The relative protein quantity of IRS-1 tyrosine phosphorylation. k Glucose uptake. The relative quantity of proteins was analyzed using Quantity One software. A representative blot is shown and the data was expressed as mean ± SEM in each bar graph. *P < 0.05 (PGRN vs Control); # P < 0.05 (PGRN + 4-PBA vs PGRN)

The administration of the ER stress inhibitor 4-PBA reversed the negative effect of progranulin in vivo

Meanwhile, we investigated the effects of the ER stress inhibitor 4-PBA on oxidative stress, autophagy and insulin sensitivity in vivo. As expected, those mice injected with progranulin exhibited activated ER stress and oxidative stress, up-regulation of autophagy, and developed glucose intolerance and insulin insensitivity as measured by GTT and ITT (Fig. 4a-k). In accordance with our findings in vitro, 4-PBA treatment in the mice injected with progranulin reduced iNOS expression and nitrite concentrations in adipose tissue (Fig. 4a-c), inhibited activated ER stress as demonstrated by reduced protein expression of CHOP, GRP78 and the phosphorylation of PERK (Fig. 4d and e), reversed adipose autophagic imbalance (Fig. 4f and g), increased IRS-1 tyrosine phosphorylation and glucose uptake (Fig. 4h and i), and improved glucose tolerance and insulin sensitivity (Fig. 4j and k), in comparison with the mice just injected with progranulin. These findings demonstrated the potential association among autophagy, ER stress and oxidative stress required for progranulin-mediated adipose insulin resistance.

Fig. 4
figure 4

The administration of the ER stress inhibitor 4-PBA reversed the negative effect of progranulin in vivo. a Protein expression of iNOS in adipose tissue. b The relative protein quantity of iNOS in adipose tissue. c Nitrite concentrations in adipose tissue. d Protein expression of CHOP and GRP78, and the phosphorylation of PERK in adipose tissue. e The relative protein quantity of CHOP, GRP78 and p-PERK in adipose tissue. f Protein expression of Atg7, p62 and LC3 in adipose tissue. g The relative protein quantity of Atg7, p62 and LC3 in adipose tissue. h IRS-1 tyrosine phosphorylation in adipose tissue. i The relative protein quantity of IRS-1 tyrosine phosphorylation in adipose tissue. j GTT. k ITT. The relative quantity of proteins was analyzed using Quantity One software. A representative blot is shown and the data was expressed as mean ± SEM in each bar graph. *P < 0.05 (PGRN vs Control); # P < 0.05 (PGRN + 4-PBA vs PGRN)

Discussion

The previous study indicated that progranulin induced adipose insulin resistance and autophagic imbalance via TNFR1 in vivo, and ablation of progranulin prevented from diet-induced insulin resistance [3, 4]. Consistent with these results, our findings showed that mice injected with progranulin developed insulin insensitivity, and the ER stress inhibitor 4-PBA treatment reversed the negative effect of progranulin in vivo and in vitro. Furthermore, we explored the mechanism of progranulin action, and our results revealed that progranulin treatment activated oxidative stress and ER stress, elevated autophagy and induced insulin insensitivity in adipocytes and adipose tissue of mice. Interestingly, inhibition of iNOS and ER stress both reversed progranulin-induced stress response and increased autophagy, protecting against insulin resistance in adipocytes. Therefore, progranulin may partially participate in the development of insulin resistance, which was associated with oxidative stress, ER stress and autophagy, but the definite effects of progranulin on insulin insensitivity in humans need to be further studied.

Accumulating evidence suggested that oxidative stress played a key role on the development of insulin resistance [11]. The expression of iNOS was elevated in adipose tissue of mice in dietary and genetic obesity [12], while iNOS−/− mice were prevented from diet-induced insulin resistance [13]. In line with these findings, our results showed that oxidative stress was involved in progranulin-induced insulin resistance, and the iNOS-specific inhibitor SMT helped to protect from insulin insensitivity in adipocytes treated with progranulin. It has been proved that iNOS caused high concentration of NO in response to kinds of inflammatory signaling [14], so inflammatory signaling associated with progranulin remains to be determined.

Recent studies showed that antioxidant activity of \( \beta \)-carotene and lycopene opposed inflammatory oxidative stress and increased vascular nitric oxide bioavailability allowing protective effects against cardiovascular disease [15]. Additionally, previous research found that omega-3 polyunsaturated fatty acids became more significant in reducing the inflammatory and insulin resistant condition [16]. With this background, we inferred that the antioxidant properties of fatty acids could improve insulin resistance via inhibited oxidative stress and ER stress, suggesting an innovative strategy for the treatment against insulin resistance.

As is known, ER stress also plays a crucial role as a chronic stimulus on the development of insulin resistance [1719]. In the present study, we found that progranulin caused adipose insulin insensitivity via increased autophagy, resulting from activated oxidative stress and ER stress. Of note, inhibition of iNOS with SMT resulted in a significant decrease in the expression levels of ER stress markers such as CHOP, GRP78 and p-PERK in adipocytes, while inhibition of ER stress with 4-PBA also lead to reduced the expression of iNOS and ROS production. Thus the mechanisms of interactions between oxidative stress and ER stress in adipocytes treated with progranulin remain to be identified in the future.

A growing number of evidences supported the link between autophagy and insulin resistance. Some studies found that the expressions of autophagy indicators were elevated in adipose tissue of humans and mice in obesity [2022], and the induction of autophagy might benefit in ER stress-induced unfolded protein response, thus we speculated that increased autophagy in adipocytes treated with progranulin could be a decompensatory response to activated ER stress. The relationship among autophagy, oxidative stress and ER stress still needs to be further explored.

Conclusions

In summary, we demonstrated that progranulin treatment activated oxidative stress and ER stress, elevated autophagy and induced insulin insensitivity in adipocytes and adipose tissue of mice. Additionally, inhibition of iNOS and ER stress both reversed the negative effect of progranulin on insulin sensitivity. Although further studies are warranted to address inflammatory signaling associated with progranulin in glucose and lipid metabolism, our findings provided new insights into the clinical potential of the novel adipokine progranulin in the regulation of insulin resistance, suggesting that progranulin may mediate insulin resistance, at least in part, by inducing autophagy via activated oxidative stress and ER stress.

Change history

References

  1. Kessenbrock K, Fröhlich L, Sixt M, et al. Proteinase 3 and neutrophil elastase enhance inflammation in mice by inactivating antiinflammatory progranulin. J Clin Invest. 2008;118:2438–47.

    CAS  Google Scholar 

  2. Zhu J, Nathan C, Jin W, et al. Conversion of proepithelin to epithelins: roles of SLPI and elastase in host defense and wound repair. Cell. 2002;111:867–78.

    Article  CAS  Google Scholar 

  3. Matsubara T, Mita A, Minami K, et al. PGRN is a key adipokine mediating high fat diet-induced insulin resistance and obesity through IL-6 in adipose tissue. Cell Metab. 2012;15:38–50.

    Article  CAS  Google Scholar 

  4. Zhou B, Li H, Liu J, et al. Progranulin induces adipose insulin resistance and autophagic imbalance via TNFR1 in mice. J Mol Endocrinol. 2015;55:231–43.

    Article  CAS  Google Scholar 

  5. Jeon MJ, Leem J, Ko MS, et al. Mitochondrial dysfunction and activation of iNOS are responsible for the palmitate-induced decrease in adiponectin synthesis in 3T3L1 adipocytes. Exp Mol Med. 2012;44:562–70.

    Article  CAS  Google Scholar 

  6. Yang L, Li P, Fu S, et al. Defective Hepatic Autophagy in Obesity Promotes ER Stress and Causes Insulin Resistance. Cell Metab. 2010;11:467–78.

    Article  CAS  Google Scholar 

  7. Younce CW, Kolattukudy PE. MCP-1 causes cardiomyoblast death via autophagy resulting from ER stress caused by oxidative stress generated by inducing a novel zinc-finger protein, MCPIP. Biochem J. 2010;426:43–53.

    Article  CAS  Google Scholar 

  8. Zhou B, Li H, Liu J, et al. Autophagic dysfunction is improved by intermittent administration of osteocalcin in obese mice. Int J Obes (Lond). 2013;40:833–43.

    Article  Google Scholar 

  9. Zhou B, Li H, Xu L, et al. Osteocalcin Reverses Endoplasmic Reticulum Stress and Improves Impaired Insulin Sensitivity Secondary to Diet-Induced Obesity Through Nuclear Factor-kB Signaling Pathway. Endocrinology. 2013;154:1055–68.

    Article  CAS  Google Scholar 

  10. Zhou B, Li H, Liu J, et al. Intermittent injections of osteocalcin reverse autophagic dysfunction and endoplasmic reticulum stress resulting from diet-induced obesity in the vascular tissue via the NF-kB-p65-dependent mechanism. Cell Cycle. 2013;12:1901–13.

    Article  CAS  Google Scholar 

  11. Kapur S, Picard F, Perreault M, et al. Nitricoxide: a new player in the modulation of energy metabolism. Int J Obes Relat Metab Disord. 2000;24:S36–40.

    Article  CAS  Google Scholar 

  12. Zhou YT, Grayburn P, Karim A, et al. Lipotoxic heart disease in obese rats: implications for human obesity. Proc Natl Acad Sci U S A. 2000;97:1784–9.

    Article  CAS  Google Scholar 

  13. Perreault M, Marette A. Targeted disruption of inducible nitricoxide synthase protects against obesity-linked insulin resistance in muscle. Nat Med. 2001;7:1138–43.

    Article  CAS  Google Scholar 

  14. Nathan C, Xie QW. Nitric oxide synthases: roles, tolls, and controls. Cell. 1994;78:915–8.

    Article  CAS  Google Scholar 

  15. Ciccone MM, Cortese F, Gesualdo M, et al. Dietary intake of carotenoids and their antioxidant and anti-inflammatory effects in cardiovascular care. Mediators Inflamm. 2013;2013:782137.

    Article  Google Scholar 

  16. Ciccone MM, Scicchitano P, Gesualdo M, et al. The role of omega-3 polyunsaturated fatty acids supplementation in childhood: a review. Recent Pat Cardiovasc Drug Discov. 2013;8:42–55.

    Article  CAS  Google Scholar 

  17. Ozawa K, Miyazaki M, Matsuhisa M, et al. The endoplasmic reticulum chaperone improves insulin resistance in type 2 diabetes. Diabetes. 2005;54:657–63.

    Article  CAS  Google Scholar 

  18. Nakatani Y, Kaneto H, Kawamori D, et al. Involvement of endoplasmic reticulum stress in insulin resistance and diabetes. J Biol Chem. 2005;280:847–51.

    Article  CAS  Google Scholar 

  19. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. J Clin Invest. 2005;115:1111–9.

    Article  CAS  Google Scholar 

  20. Kovsan J, Blüher M, Tarnovscki T, et al. Altered autophagy in human adipose tissues in obesity. J Clin Endocrinol Metab. 2011;96:E268–77.

    Article  CAS  Google Scholar 

  21. Zhou L, Zhang J, Fang Q, et al. Autophagy-mediated insulin receptor down-regulation contributes to endoplasmic reticulum stress-induced insulin resistance. Mol Pharmacol. 2009;76:596–603.

    Article  CAS  Google Scholar 

  22. Xu L, Zhou B, Li H, et al. Serum levels of progranulin are closely associated with microvascular complication in type 2 diabetes. Dis Markers. 2015;2015:357279.

    Article  Google Scholar 

Download references

Acknowledgments

Not applicable.

Funding

This work was supported by the programs from the National Natural Science Foundation of China (no. 81472038 and no.81500016), and the Young Innovators Awards of the First Affiliated Hospital of Xi’an Jiaotong University (2014YK3 and 2015YK6).

Availability of data and materials

Please contact author for data requests.

Authors’ contributions

BZ and QG designed and executed the experiments and drafted the manuscript. LX, JL and HL conducted most of the experiments and contributed to manuscript preparation. BZ, SW and HS contributed to the experimental design. All authors revised, edited and approved the final version of the manuscript.

Competing interests

The authors declare that they have no competing interests.

Consent for publication

Not applicable.

Ethics approval

This study was carried out in strict accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Committee on the Ethics of Animal Experiments of Medical School of Xi’an Jiaotong University (Permit number: 2013–025).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bo Zhou.

Additional information

This article has been retracted. Please see the retraction notice for more detail:https://doi.org/10.1186/s12944-022-01746-3

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Guo, Q., Xu, L., Li, H. et al. RETRACTED ARTICLE: Progranulin causes adipose insulin resistance via increased autophagy resulting from activated oxidative stress and endoplasmic reticulum stress. Lipids Health Dis 16, 25 (2017). https://doi.org/10.1186/s12944-017-0425-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12944-017-0425-6

Keywords