You are currently viewing a new version of our website. To view the old version click .
Medicina
  • Review
  • Open Access

10 July 2019

Role of Autophagy on Heavy Metal-Induced Renal Damage and the Protective Effects of Curcumin in Autophagy and Kidney Preservation

,
,
,
and
Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico (UNAM), Mexico City 04510, Mexico
*
Author to whom correspondence should be addressed.

Abstract

Curcumin is a hydrophobic polyphenol compound extracted from the rhizome of turmeric. The protective effect of curcumin on kidney damage in multiple experimental models has been widely described. Its protective effect is mainly associated with its antioxidant and anti-inflammatory properties, as well as with mitochondrial function maintenance. On the other hand, occupational or environmental exposure to heavy metals is a serious public health problem. For a long time, heavy metals-induced nephrotoxicity was mainly associated with reactive oxygen species overproduction and loss of endogenous antioxidant activity. However, recent studies have shown that in addition to oxidative stress, heavy metals also suppress the autophagy flux, enhancing cell damage. Thus, natural compounds with the ability to modulate and restore autophagy flux represent a promising new therapeutic strategy. Furthermore, it has been reported in other renal damage models that curcumin’s nephroprotective effects are related to its ability to regulate autophagic flow. The data indicate that curcumin modulates autophagy by classic signaling pathways (suppression of protein kinase B (Akt)/mammalian target of rapamycin (mTOR) and/or by stimulation of adenosine monophosphate-activated protein kinase (AMPK) and extracellular signal-dependent kinase (ERK) pathways). Moreover, it allows lysosomal function preservation, which is crucial for the later stage of autophagy. However, future studies of autophagy modulation by curcumin in heavy metals-induced autophagy flux impairment are still needed.

1. Introduction

Curcumin or diferuloylmethane (1,7-bis(4-hydroxy-3-methoxyphenyl)-16-heptadiene-3,5-dione) is a hydrophobic polyphenol extracted from the rhizome of Curcuma longa L., also known as turmeric [1], which is widely grown in the southern and south western tropical regions of Asia [2]. Curcumin is formed by two aromatic rings (with o-methoxy phenolic groups) linked to an α,β-unsaturated β-diketone moiety [3]. Curcumin can exist in tautomeric forms, keto and enol [4]. It has been widely used in traditional medicine due to its multiple antioxidant [5], antitumor [6], antiviral [7] and anti-inflammatory properties [8], among others [1,3]. Furthermore, studies have shown its protective effect in multiple diseases such as cancer [6], and neurological [9], metabolic [10], lung [11], liver [5], renal [12], and cardiovascular diseases [2]. In addition to its many therapeutic activities, curcumin has the advantage of its biosafety in animals as well in healthy humans, even at high doses (12 g per day), without undesirable effects [13]. In addition, it has been shown that curcumin provides a nephroprotective effect against various xenobiotics, including heavy metals [12,14,15,16,17].
Heavy metals comprise elements like cadmium (Cd), lead (Pb), arsenic (As), mercury (Hg), and chromium (Cr), which are located along the Earth’s crust in various forms [18]. These elements are widely used in different anthropogenic activities, ranging from agriculture to industry [19]. Heavy metals can enter to the body by three routes: Intake (contaminated water or food), inhalation, and dermal contact [20]. Prolonged exposure (occupational or environmental) can cause serious problems in human health, especially in the kidneys, since they represent the major route of heavy metal excretion from the body [18]. Adverse effects of heavy metals have been usually associated with oxidative stress, which involves an overproduction of reactive oxygen species (ROS) and the loss of the endogenous antioxidant defense, as well as with mitochondrial dysfunction [18,21]. However, it was recently found that heavy metals can also impair autophagy flux [22,23,24], an evolutionarily-conserved self-digestive process, which is generally considered to be a cell survival mechanism [25]. In vitro and in vivo studies related to the participation of autophagy in kidney damage have described that autophagy helps to remove damaged mitochondria, avoiding ROS overproduction and cellular damage [26,27].
In this review we summarized the mechanisms involved in autophagy regulation by curcumin, as well heavy metals-induced autophagy flux impairment in the kidney. On the other hand, curcumin administration has been able to restore renal function in several models of kidney damage [8,14,28,29]. Furthermore, in hyperoxaluria, maleate and contrast-induced nephropathy, curcumin’s nephroprotection has been related to its ability to modulate the autophagy flux [17,30,31]. However, curcumin’s effect on heavy metals-induced autophagy flux impairment has not been explored yet, generating the opportunity for exploration in future studies.

2. Curcumin’s Antioxidant Effects and Mitochondrial Protection in Kidney Damage Models

Curcumin’s direct antioxidant effect derives from the presence of conjugated double bonds in its structure, allowing curcumin to act as an electron donor [32]. Therefore, it scavenges superoxide anion (O2●−), hydroxyl radical (OH), singlet oxygen (1O2), hydrogen peroxide (H2O2), nitric oxide (NO), peroxynitrite (ONOO), and peroxyl radical [33]. Moreover, curcumin also activates the antioxidant response element (ARE) by the kelch-like ECH-associating protein (Keap1)/nuclear factor erythroid 2-related factor 2 (Nrf2) system [34]. The protective effect of curcumin has been described in kidney damage models induced by ischemia/reperfusion [8], cisplatin [35], 5/6 nephrectomy [16], and heavy metals (Pb, Cd, Cr) [12,14,15], where its antioxidant activity is highlighted. In addition, it also preserves mitochondrial function [16,29,35], which also contributes to renal function preservation in kidney function. The anti-inflammatory effect of curcumin also plays a prominent role in acute and chronic kidney damage models [36,37,38]. In this regard, curcumin has an anti-fibrotic effect in glomerulonephritis model, which involves the reduction of transforming growth factor-β1 and fibronectin production [36]. This effect is dependent on induction of heme oxygenase-1 (HO-1), a target of Nrf2. Following the same route, Ghosh et al. [37] in the 5/6 nephrectomy model, highlighted the effect of curcumin on the development of chronic renal failure, where inflammation plays an important role through tumor necrosis factor alpha (TNFα) and the transcription factor nuclear kappa B (NF-κB). In this model, curcumin partially suppressed the TNFα-mediated NF-κB activity and avoided macrophage infiltration, as well as the functional (such as proteinuria, blood urea nitrogen, and plasma creatinine) and structural (tubular atrophy, hyperplasia, and glomerulosclerosis) alterations. Furthermore, in the renal ischemia/reperfusion model, curcumin attenuated interferon gamma (IFNγ) expression, while increasing the IL-10 levels [38]. On the other hand, in an in vitro and in vivo model of rhabdomyolysis, curcumin reduced renal damage associated with rhabdomyolysis. In addition to its antioxidant and anti-inflammatory effect, curcumin decreased ferroptosis-mediated cell death [39]. Interestingly, the inhibition of this type of autophagy associated to iron metabolism, is able to attenuate the function and structural alterations in the kidney. Finally, the protective effect of curcumin was also mediated by HO-1 [39].

3. Autophagy and Its Evaluation

Autophagy is a biological process that allows the preservation of cellular homeostasis by the removal of damaged macromolecules and/or organelles in response to a variety of stimuli [40]. Autophagy consists of five steps: (1) Formation of the phagophore, a complex of Beclin-1, phosphatidylinositol 3-kinase/vacuolar protein sorting 34 (VPS34) and VPS15, (2) phagophore elongation and cargo recruitment, (3) autophagosome maturation, (4) fusion between the autophagosome and lysosome, and (5) autolysosome degradation [41,42]. In addition, formation of autophagosomes and autolysosomal degradation are essential stages to evaluate if the autophagy flux is functional. The usual hallmarks evaluated in these steps are the levels of: Microtubule-associated protein 1 light chain 3 (LC3-I) and its phosphatidyethanolamine form (LC3-II), as well as the ubiquitin binding protein p62, also called sequestosome 1. The LC3-II is an essential protein for elongation and closure of the phagophore, localized in the autophagosome membranes. Meanwhile, p62 is a receptor protein, which binds by its ubiquitin domain to the specific cargo, to anchor them to the LC3-II, present inside the autophagosome, to the subsequent cargo’s degradation in autophagolysosomes [43,44].
In relation to the signaling pathway, key regulators of autophagy include the class I phosphatidylinositol 3-kinase (PI3K) and adenosine monophosphate-activated protein kinase (AMPK) and the autophagy inhibitor the mammalian target of rapamycin (mTOR) [34]. mTOR activation is associated to the PI3K/protein kinase B (AKT)/p70 ribosomal protein S6 kinase (p70S6K) pathway and linked to growth factors. Thus, mTOR negatively regulates autophagy. Meanwhile, the AMPK kinase senses intracellular adenosine triphosphate levels and inhibits formation of the multiprotein kinase complex 1 of mTOR (mTORC1), leading to its dissociation from the Atg2/unc-51-like kinases complex (ULK) and to the dephosphorylation/activation of ULK1 and/ or ULK2, which triggers autophagy’s initiation [45]. Likewise, mTORC1 is also regulated by tuberous sclerosis complex 1 (TSC1)/TSC2 and acts as a brake for this pathway, therefore modulating autophagy [42]. In this regard, Akt phosphorylates TSC2 and inactivates the TSC1/TSC2 complex, meanwhile its phosphorylation by AMPK has the opposite effect [42].

4. The Autophagy in Heavy Metals Kidney Damage

The role of autophagy in renal injury is still under debate [46]. It has been described in tubular epithelial cells that autophagy acts as a survival mechanism in multiple renal damage models, including cisplatin, cyclosporine, and ischemia [47]. However, its overactivation can be counterproductive, leading to autophagic cell death [48]. Therefore, due to the dual role of autophagy, factors such as the temporality and intensity of the stimulus have to be considered a priori, to determine the role of this process.
In relation to heavy metal-nephrotoxicity (as summarized in Table 1), the participation of autophagy in cadmium chloride (CdCl2)-induced damage in mice has been demonstrated [49]. It was shown that high doses of CdCl2 increase the LC3-II/LC3-I ratio and promote the formation of autophagosomes, which lead to autophagic cell death, instead of mitigating the renal damage [49]. This is consistent with the study of Shi et al. (2019), which revealed that prolonged exposure to CdCl2 increased apoptosis in chicken kidneys by c-Jun N-terminal kinase (JNK)-dependent autophagy [50]. In the same sense, Liu et al. (2017) found in rat proximal tubule (rPT) cells that exposure to CdCl2 increases LC3-II and Beclin-1 expression, as well as the number of autophagic vacuoles, in a dose-dependent manner [51].
Table 1. In vitro and in vivo effects of heavy metals on autophagy in kidney.
Nevertheless, the increase in LC3-II expression and in autophagosome numbers are not irrefutable proof of autophagy flux. The lack of lysosomal degradation evidence represents a more reliable parameter to confirm the autophagy flux [52], since autophagosome accumulation can derive from the increase in autophagosome formation or the suppression of lysosomal degradation [53]. In this regard it was shown that exposure to a subtoxic dose (0.3 mg Cd/kg) for 5 days did not affect tubular or glomerular function in rat kidneys, although CdCl2 accumulation in the renal cortex was observed [54]. However, proximal convoluted tubule cells (PCT)-exposure to a low dose of CdCl2, showed significant morphological changes associated to autophagy, but not to apoptosis. Furthermore, autophagy upregulation derived from the binding of CdCl2 to the sulfhydryl groups of proteins, oxidative stress, and the endoplasmic reticulum stress-dependent autophagy induction [54]. Similarly, Liu et al. (2017) showed in rPT cells exposed to CdCl2, the anti-apoptotic effect of autophagy with rapamycin treatment, which decreased the apoptosis rate by Fas/Fas ligand (FasL) pathway inhibition, in contrast to 3-methyladenine (3-MA) treatment, an autophagy inhibitor, which increased the apoptosis rate [51].
In support of this, Liu et al. [23] found in rPT cells treated with cadmium acetate (CdAc2) an increase in LC3-II and p62 protein expression, whose expression is inversely correlated with autophagic activity. This suggests autophagy flux inhibition, which was associated with reduction of the autophagosome-lysosome fusion, as a consequence of the cellular fusion machinery depletion and cytosolic calcium increase.
It is important to highlight that cadmium is not the only heavy metal able to impair autophagy flux. In vitro studies in rPT cells treated with lead nitrate (PbNO3) have shown the accumulation of LC3-II and p62 proteins [22,56]. Furthermore, autophagy inhibition increased the levels of cleaved caspase-3 and poly (ADP-ribose) polymerase (PARP), which evidenced its participation in lead-induced apoptosis in rPT cells [55]. The autophagy flux alterations in this condition were associated to autophagolysosome alkalinization, as a consequence of the suppression of the two V-ATPase subunits (which hydrolyze ATP to pump protons into the lysosome lumen). Furthermore, lead induced lysosomal membrane permeabilization (LMP), allowing cathepsins-release to the cytoplasm, which induced caspases-mediated apoptosis [22]. The authors also demonstrated the deregulation of the AMPK/mTOR pathway and suggested its participation in the lead-induced impairment of autophagy flux [56].
Autophagy flux disruption has also been observed in arsenic models. Kimura et al. (2016) found in female mice administered with sodium arsenite (NaAsO2), an accumulation of LC3-II and p62 proteins. The highlight of this study is that the participation of estrogens in the autophagic flux impairment was demonstrated, which affects more the females than the males. This phenomenon is associated with ERK overactivation, by SOCS3-dependent IL-6/STAT3 signaling pathway suppression [24].
About Cr (VI), its effect on autophagy in the kidneys is still unknown, but indirect evidence suggest that it may also inhibit the autophagy flux [58]. In rat kidney cortex and HK2 cells exposed to potassium dichromate (K2Cr2O7), there was an increase in p-mTOR and in phosphorylated-p70 ribosomal protein S6 kinase (p-p70S6K) (mTOR target) levels, which suggests the inhibition of autophagy flux [58].

6. Conclusions

Curcumin administration has demonstrated the ability to restore autophagy flux balance. Therefore, its administration in heavy metals-induced renal damage could be a possible treatment strategy to reverse the autophagy impairment, thus contributing to the preservation of renal function.

Author Contributions

Conceptualization, S.H.A.-R.; writing—original draft preparation, S.H.A.-R., A.L.-L.; writing—review and editing, S.H.A.-R., O.E.A.-T., A.L.-L., L.M.R.-F., J.P.-C.; visualization, S.H.A.-R., O.E.A.-T., J.P.-C.; funding acquisition, J.P.-C.

Funding

This work was supported by Consejo Nacional de Ciencia y Tecnología (CONACyT, Mexico Grant No. 220646); Programa de Apoyo a Proyecto de Investigación e Innovación Tecnológica (PAPIIT, UNAM, Mexico, Grants IN201316 and IN202219); Programa de Apoyo a la Investigación y el Posgrado (PAIP, Mexico, Grant No. 5000-9105). S.H.A.-R. is student from Programa de Maestría y Doctorado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México (UNAM) and received a fellowship from CONACyT.

Acknowledgments

We thank Elena Martínez-Klimova for her assisting the review of the present text.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Moghaddam, N.S.A.; Oskouie, M.N.; Butler, A.E.; Petit, P.X.; Barreto, G.E.; Sahebkar, A. Hormetic effects of curcumin: What is the evidence? J. Cell. Physiol. 2019, 234, 10060–10071. [Google Scholar] [CrossRef] [PubMed]
  2. Kocaadam, B.; Şanlier, N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health, Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef] [PubMed]
  3. de Oliveira, M.R.; Jardim, F.R.; Setzer, W.N.; Nabavi, S.M.; Nabavi, S.F. Curcumin, mitochondrial biogenesis, and mitophagy: Exploring recent data and indicating future needs. Biotechnol. Adv. 2016, 34, 813–826. [Google Scholar] [CrossRef] [PubMed]
  4. Rainey, N.; Motte, L.; Aggarwal, B.B.; Petit, P.X. Curcumin hormesis mediates a cross-talk between autophagy and cell death. Cell Death Dis. 2015, 6, e2003. [Google Scholar] [CrossRef] [PubMed]
  5. García-Niño, W.R.; Pedraza-Chaverrí, J. Protective effect of curcumin against heavy metals-induced liver damage. Food Chem. Toxicol. 2014, 69, 182–201. [Google Scholar] [CrossRef]
  6. Liu, F.; Gao, S.; Yang, Y.; Zhao, X.; Fan, Y.; Ma, W.; Yang, D.; Yang, A.; Yu, Y. Antitumor activity of curcumin by modulation of apoptosis and autophagy in human lung cancer A549 cells through inhibiting PI3K/Akt/mTOR pathway. Oncol. Rep. 2018, 39, 1523–1531. [Google Scholar] [CrossRef]
  7. Mounce, B.C.; Cesaro, T.; Carrau, L.; Vallet, T.; Vignuzzi, M. Curcumin inhibits Zika and chikungunya virus infection by inhibiting cell binding. Antivir. Res. 2017, 142, 148–157. [Google Scholar] [CrossRef]
  8. Zhang, J.; Tang, L.; Li, G.S.; Wang, J. The anti-inflammatory effects of curcumin on renal ischemia-reperfusion injury in rats. Ren. Fail. 2018, 40, 680–686. [Google Scholar] [CrossRef]
  9. Huang, L.; Chen, C.; Zhang, X.; Li, X.; Chen, Z.; Yang, C.; Liang, X.; Zhu, G.; Xu, Z. Neuroprotective Effect of Curcumin Against Cerebral Ischemia-Reperfusion Via Mediating Autophagy and Inflammation. J. Mol. Neurosci. 2018, 64, 129–139. [Google Scholar] [CrossRef]
  10. Jin, T.; Song, Z.; Weng, J.; Fantus, I.G. Curcumin and other dietary polyphenols: Potential mechanisms of metabolic actions and therapy for diabetes and obesity. Am. J. Physiol. Metab. 2018, 314, E201–E205. [Google Scholar] [CrossRef]
  11. Patel, S.S.; Acharya, A.; Ray, R.S.; Agrawal, R.; Raghuwanshi, R.; Jain, P. Cellular and molecular mechanisms of curcumin in prevention and treatment of disease. Crit. Rev. Food Sci. Nutr. 2019, 1–53. [Google Scholar] [CrossRef] [PubMed]
  12. Avila-Rojas, S.H.; Tapia, E.; Briones-Herrera, A.; Aparicio-Trejo, O.E.; León-Contreras, J.C.; Hernández-Pando, R.; Pedraza-Chaverri, J. Curcumin prevents potassium dichromate (K2Cr2O7)-induced renal hypoxia. Food Chem. Toxicol. 2018, 121, 472–482. [Google Scholar] [CrossRef] [PubMed]
  13. Lao, C.D.; Ruffin, M.T.; Normolle, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E. Dose escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 2006, 6, 10. [Google Scholar] [CrossRef] [PubMed]
  14. Kim, K.S.; Lim, H.J.; Lim, J.S.; Son, J.Y.; Lee, J.; Lee, B.M.; Chang, S.C.; Kim, H.S. Curcumin ameliorates cadmium-induced nephrotoxicity in Sprague-Dawley rats. Food Chem. Toxicol. 2018, 114, 34–40. [Google Scholar] [CrossRef] [PubMed]
  15. Soliman, M.M.; Baiomy, A.A.; Yassin, M.H. Molecular and Histopathological Study on the Ameliorative Effects of Curcumin Against Lead Acetate-Induced Hepatotoxicity and Nephrototoxicity in Wistar Rats. Biol. Trace Elem. Res. 2015, 167, 91–102. [Google Scholar] [CrossRef]
  16. Aparicio-Trejo, O.E.; Tapia, E.; Molina-Jijón, E.; Medina-Campos, O.N.; Macías-Ruvalcaba, N.A.; León-Contreras, J.C.; Hernández-Pando, R.; García-Arroyo, F.E.; Cristóbal, M.; Sánchez-Lozada, L.G.; et al. Curcumin prevents mitochondrial dynamics disturbances in early 5/6 nephrectomy: Relation to oxidative stress and mitochondrial bioenergetics. Biofactors 2017, 43, 293–310. [Google Scholar] [CrossRef] [PubMed]
  17. Buyuklu, M.; Kandemir, F.M.; Ozkaraca, M.; Set, T.; Bakirci, E.M.; Topal, E. Protective effect of curcumin against contrast induced nephropathy in rat kidney: What is happening to oxidative stress, inflammation, autophagy and apoptosis? Eur. Rev. Med. Pharmacol. Sci. 2014, 18, 461–470. Available online: http://www.ncbi.nlm.nih.gov/pubmed/24610611 (accessed on 28 April 2019).
  18. Rana, M.N.; Tangpong, J.; Rahman, M.M. Toxicodynamics of Lead, Cadmium, Mercury and Arsenic- induced kidney toxicity and treatment strategy: A mini review. Toxicol. Rep. 2018, 5, 704–713. [Google Scholar] [CrossRef]
  19. Lentini, P.; Zanoli, L.; Granata, A.; Signorelli, S.S.; Castellino, P.; Dellaquila, R. Kidney and heavy metals - The role of environmental exposure. Mol. Med. Rep. 2017, 15, 3413–3419. [Google Scholar] [CrossRef]
  20. Wu, W.; Zhang, K.; Jiang, S.; Liu, D.; Zhou, H.; Zhong, R.; Zeng, Q.; Cheng, L.; Miao, X.; Tong, Y.; et al. Association of co-exposure to heavy metals with renal function in a hypertensive population. Environ. Int. 2018, 112, 198–206. [Google Scholar] [CrossRef]
  21. Abdel-Moneim, A.M.; El-Toweissy, M.Y.; Ali, A.M.; Allah, A.A.M.A.; Darwish, H.S.; Sadek, I.A. Curcumin Ameliorates Lead (Pb2+)-Induced Hemato-Biochemical Alterations and Renal Oxidative Damage in a Rat Model. Biol. Trace Elem. Res. 2015, 168, 206–220. [Google Scholar] [CrossRef] [PubMed]
  22. Song, X.B.; Liu, G.; Liu, F.; Yan, Z.G.; Wang, Z.Y.; Liu, Z.P.; Wang, L. Autophagy blockade and lysosomal membrane permeabilization contribute to lead-induced nephrotoxicity in primary rat proximal tubular cells. Cell Death Dis. 2017, 8, e2863. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, F.; Wang, X.Y.; Zhou, X.P.; Liu, Z.P.; Song, X.B.; Wang, Z.Y.; Wang, L. Cadmium disrupts autophagic flux by inhibiting cytosolic Ca 2+ -dependent autophagosome-lysosome fusion in primary rat proximal tubular cells. Toxicology 2017, 383, 13–23. [Google Scholar] [CrossRef] [PubMed]
  24. Kimura, A.; Ishida, Y.; Nosaka, M.; Kuninaka, Y.; Hama, M.; Kawaguchi, T.; Sakamoto, S.; Shinozaki, K.; Iwahashi, Y.; Takayasu, T.; et al. Exaggerated arsenic nephrotoxicity in female mice through estrogen-dependent impairments in the autophagic flux. Toxicology 2016, 339, 9–18. [Google Scholar] [CrossRef] [PubMed]
  25. Yan, P.; Sun, X.; Chen, X.; Chen, Y.; Wang, X.; Su, D.; Zhou, H.; Gao, L.; Lu, L.; Wang, J.; et al. The Polyphenolic Compound Curcumin Conjugation with an Alkyne Moiety in the Process of Autophagy. Am. J. Chin. Med. 2018, 46, 673–687. [Google Scholar] [CrossRef]
  26. Tan, J.; Wang, M.; Song, S.; Miao, Y.; Zhang, Q. Autophagy activation promotes removal of damaged mitochondria and protects against renal tubular injury induced by albumin overload. Histol. Histopathol. 2018, 33, 681–690. [Google Scholar] [CrossRef]
  27. Yang, X.; Yan, X.; Yang, D.; Zhou, J.; Song, J.; Yang, D. Rapamycin attenuates mitochondrial injury and renal tubular cell apoptosis in experimental contrast-induced acute kidney injury in rats. Biosci. Rep. 2018, 38, BSR20180876. [Google Scholar] [CrossRef]
  28. Zhu, F.; Chen, M.; Zhu, M.; Zhao, R.; Qiu, W.; Xu, X.; Liu, H.; Zhao, H.; Yu, R.; Wu, X.; et al. Curcumin Suppresses Epithelial—Mesenchymal Transition of Renal Tubular Epithelial Cells through the Inhibition of Akt/mTOR Pathway. Biol. Pharm. Bull. 2017, 40, 17–24. [Google Scholar] [CrossRef]
  29. Molina-Jijón, E.; Tapia, E.; Zazueta, C.; el Hafidi, M.; Zatarain-Barrón, Z.L.; Hernández-Pando, R.; Medina-Campos, O.N.; Zarco-Márquez, G.; Torres, I.; Pedraza-Chaverri, J. Curcumin prevents Cr(VI)-induced renal oxidant damage by a mitochondrial pathway, Free Radic. Biol. Med. 2011, 51, 1543–1557. [Google Scholar] [CrossRef]
  30. Li, Y.; Zhang, J.; Liu, H.; Yuan, J.; Yin, Y.; Wang, T.; Cheng, B.; Sun, S.; Guo, Z. Curcumin ameliorates glyoxylate-induced calcium oxalate deposition and renal injuries in mice. Phytomedicine 2019, 61, 152861. [Google Scholar] [CrossRef]
  31. Molina-Jijón, E.; Aparicio-Trejo, O.E.; Rodríguez-Muñoz, R.; León-Contreras, J.C.; Cárdenas-Aguayo, M.d.; Medina-Campos, O.N.; Tapia, E.; Sánchez-Lozada, L.G.; Hernández-Pando, R.; Reyes, J.L.; et al. The nephroprotection exerted by curcumin in maleate-induced renal damage is associated with decreased mitochondrial fission and autophagy. BioFactors 2016, 42, 686–702. [Google Scholar] [CrossRef] [PubMed]
  32. Shakeri, A.; Cicero, A.F.G.; Panahi, Y.; Mohajeri, M.; Sahebkar, A. Curcumin: A naturally occurring autophagy modulator. J. Cell. Physiol. 2019, 234, 5643–5654. [Google Scholar] [CrossRef] [PubMed]
  33. Trujillo, J.; Granados-Castro, L.F.; Zazueta, C.; Andérica-Romero, A.C.; Chirino, Y.I.; Pedraza-Chaverrí, J. Mitochondria as a Target in the Therapeutic Properties of Curcumin. Arch. Pharm. 2014, 347, 873–884. [Google Scholar] [CrossRef] [PubMed]
  34. Zenkov, N.K.; Chechushkov, A.V.; Kozhin, P.M.; Kandalintseva, N.V.; Martinovich, G.G.; Menshchikova, E.B. Plant phenols and autophagy. Biochemistry 2016, 81, 297–314. [Google Scholar] [CrossRef] [PubMed]
  35. Ortega-Domínguez, B.; Aparicio-Trejo, O.E.; García-Arroyo, F.E.; León-Contreras, J.C.; Tapia, E.; Molina-Jijón, E.; Hernández-Pando, R.; Sánchez-Lozada, L.G.; Barrera-Oviedo, D.; Pedraza-Chaverri, J. Curcumin prevents cisplatin-induced renal alterations in mitochondrial bioenergetics and dynamic. Food Chem. Toxicol. 2017, 107, 373–385. [Google Scholar] [CrossRef] [PubMed]
  36. Gaedeke, J.; Noble, N.A.; Border, W.A. Curcumin blocks fibrosis in anti-Thy 1 glomerulonephritis through up-regulation of heme oxygenase 1. Kidney Int. 2005, 68, 2042–2049. [Google Scholar] [CrossRef]
  37. Ghosh, S.S.; Massey, H.D.; Krieg, R.; Fazelbhoy, Z.A.; Ghosh, S.; Sica, D.A.; Fakhry, I.; Gehr, T.W.B. Curcumin ameliorates renal failure in 5/6 nephrectomized rats: Role of inflammation. Am. J. Physiol. 2009, 296, F1146–F1157. [Google Scholar] [CrossRef]
  38. Liu, F.H.; Ni, W.J.; Wang, G.K.; Zhang, J.J. Protective role of curcumin on renal ischemia reperfusion injury via attenuating the inflammatory mediators and Caspase-3. Cell. Mol. Biol. 2016, 62, 95–99. Available online: http://www.ncbi.nlm.nih.gov/pubmed/27755959 (accessed on 20 June 2019).
  39. Guerrero-Hue, M.; García-Caballero, C.; Palomino-Antolín, A.; Rubio-Navarro, A.; Vázquez-Carballo, C.; Herencia, C.; Martín-Sanchez, D.; Farré-Alins, V.; Egea, J.; Cannata, P.; et al. Curcumin reduces renal damage associated with rhabdomyolysis by decreasing ferroptosis-mediated cell death. FASEB J. 2019. [Google Scholar] [CrossRef]
  40. Pickles, S.; Vigié, P.; Youle, R.J. Mitophagy and Quality Control Mechanisms in Mitochondrial Maintenance. Curr. Biol. 2018, 28, R170–R185. [Google Scholar] [CrossRef]
  41. Sureshbabu, A.; Ryter, S.W.; Choi, M.E. Oxidative stress and autophagy: Crucial modulators of kidney injury. Redox Biol. 2015, 4, 208–214. [Google Scholar] [CrossRef] [PubMed]
  42. Hashemzaei, M.; Heravi, R.E.; Rezaee, R.; Roohbakhsh, A.; Karimi, G. Regulation of autophagy by some natural products as a potential therapeutic strategy for cardiovascular disorders. Eur. J. Pharmacol. 2017, 802, 44–51. [Google Scholar] [CrossRef] [PubMed]
  43. Kaushal, G.P.; Shah, S.V. Autophagy in acute kidney injury. Kidney Int. 2016, 89, 779–791. [Google Scholar] [CrossRef] [PubMed]
  44. Dikic, I.; Elazar, Z. Mechanism and medical implications of mammalian autophagy. Nat. Rev. Mol. Cell Biol. 2018, 19, 349–364. [Google Scholar] [CrossRef] [PubMed]
  45. Navarro-Yepes, J.; Burns, M.; Anandhan, A.; Khalimonchuk, O.; del Razo, L.M.; Quintanilla-Vega, B.; Pappa, A.; Panayiotidis, M.I.; Franco, R. Oxidative stress, redox signaling, and autophagy: Cell death versus survival. Antioxid. Redox Signal. 2014, 21, 66–85. [Google Scholar] [CrossRef]
  46. Wang, Z.; Choi, M.E. Autophagy in Kidney Health and Disease. Antioxid. Redox Signal. 2014, 20, 519–537. [Google Scholar] [CrossRef]
  47. Lin, F. Autophagy in renal tubular injury and repair. Acta Physiol. 2017, 220, 229–237. [Google Scholar] [CrossRef]
  48. Luo, B.; Lin, Y.; Jiang, S.; Huang, L.; Yao, H.; Zhuang, Q.; Zhao, R.; Liu, H.; He, C.; Lin, Z. Endoplasmic reticulum stress eIF2α–ATF4 pathway-mediated cyclooxygenase-2 induction regulates cadmium-induced autophagy in kidney. Cell Death Dis. 2016, 7, e2251. [Google Scholar] [CrossRef]
  49. Yuan, Y.; Ma, S.; Qi, Y.; Wei, X.; Cai, H.; Dong, L.; Lu, Y.; Zhang, Y.; Guo, Q. Quercetin inhibited cadmium-induced autophagy in the mouse kidney via inhibition of oxidative stress. J. Toxicol. Pathol. 2016, 29, 247–252. [Google Scholar] [CrossRef]
  50. Shi, Q.; Jin, X.; Fan, R.; Xing, M.; Guo, J.; Zhang, Z.; Zhang, J.; Xu, S. Cadmium-mediated miR-30a-GRP78 leads to JNK-dependent autophagy in chicken kidney. Chemosphere 2019, 215, 710–715. [Google Scholar] [CrossRef]
  51. Liu, G.; Yuan, Y.; Long, M.; Luo, T.; Bian, J.; Liu, X.; Gu, J.; Zou, H.; Song, R.; Wang, Y.; et al. Beclin-1-mediated Autophagy Protects Against Cadmium-activated Apoptosis via the Fas/FasL Pathway in Primary Rat Proximal Tubular Cell Culture. Sci. Rep. 2017, 7, 977. [Google Scholar] [CrossRef] [PubMed]
  52. Jiang, P.; Mizushima, N. LC3- and p62-based biochemical methods for the analysis of autophagy progression in mammalian cells. Methods 2015, 75, 13–18. [Google Scholar] [CrossRef] [PubMed]
  53. Ni, H.M.; Bockus, A.; Wozniak, A.L.; Jones, K.; Weinman, S.; Yin, X.M.; Ding, W.X. Dissecting the dynamic turnover of GFP-LC3 in the autolysosome. Autophagy 2011, 7, 188–204. Available online: http://www.ncbi.nlm.nih.gov/pubmed/21107021 (accessed on 21 April 2019). [CrossRef] [PubMed]
  54. Chargui, A.; Zekri, S.; Jacquillet, G.; Rubera, I.; Ilie, M.; Belaid, A.; Duranton, C.; Tauc, M.; Hofman, P.; Poujeol, P.; et al. Cadmium-induced autophagy in rat kidney: An early biomarker of subtoxic exposure. Toxicol. Sci. 2011, 121, 31–42. [Google Scholar] [CrossRef] [PubMed]
  55. Chu, B.X.; Fan, R.F.; Lin, S.Q.; Yang, D.B.; Wang, Z.Y.; Wang, L. Interplay between autophagy and apoptosis in lead(II)-induced cytotoxicity of primary rat proximal tubular cells. J. Inorg. Biochem. 2018, 182, 184–193. [Google Scholar] [CrossRef] [PubMed]
  56. Song, X.; Li, Z.; Liu, F.; Wang, Z.; Wang, L. Restoration of autophagy by puerarin in lead-exposed primary rat proximal tubular cells via regulating AMPK-mTOR signaling. J. Biochem. Mol. Toxicol. 2017, 31, e21869. [Google Scholar] [CrossRef] [PubMed]
  57. Luo, T.; Zhang, H.; Yu, Q.; Liu, G.; Long, M.; Zhang, K.; Liu, W.; Song, R.; Bian, J.; Gu, J.; et al. ERK1/2 MAPK promotes autophagy to suppress ER stress-mediated apoptosis induced by cadmium in rat proximal tubular cells. Toxicol. Vitr. 2018, 52, 60–69. [Google Scholar] [CrossRef] [PubMed]
  58. Song, M.F.; Yang, Y.; Yi, Z.W.; Zhang, Z.Q.; Shen, X.D.; Hu, G.H.; Zhu, Y.F. Sema 3A as a biomarker of the activated mTOR pathway during hexavalent chromium-induced acute kidney injury. Toxicol. Lett. 2018, 299, 226–235. [Google Scholar] [CrossRef]
  59. Chen, P.; Huang, H.P.; Wang, Y.; Jin, J.; Long, W.G.; Chen, K.; Zhao, X.H.; Chen, C.G.; Li, J. Curcumin overcome primary gefitinib resistance in non-small-cell lung cancer cells through inducing autophagy-related cell death. J. Exp. Clin. Cancer Res. 2019, 38, 254. [Google Scholar] [CrossRef]
  60. Hsiao, Y.T.; Kuo, C.L.; Chueh, F.S.; Liu, K.C.; Bau, D.T.; Chung, J.G. Curcuminoids Induce Reactive Oxygen Species and Autophagy to Enhance Apoptosis in Human Oral Cancer Cells. Am. J. Chin. Med. 2018, 46, 1145–1168. [Google Scholar] [CrossRef] [PubMed]
  61. Aoki, H.; Takada, Y.; Kondo, S.; Sawaya, R.; Aggarwal, B.B.; Kondo, Y. Evidence That Curcumin Suppresses the Growth of Malignant Gliomas in Vitro and in Vivo through Induction of Autophagy: Role of Akt and Extracellular Signal-Regulated Kinase Signaling Pathways. Mol. Pharmacol. 2007, 72, 29–39. [Google Scholar] [CrossRef] [PubMed]
  62. Gao, Y.; Li, J.; Wu, L.; Zhou, C.; Wang, Q.; Li, X.; Zhou, M.; Wang, H. Tetrahydrocurcumin provides neuroprotection in rats after traumatic brain injury: Autophagy and the PI3K/AKT pathways as a potential mechanism. J. Surg. Res. 2016, 206, 67–76. [Google Scholar] [CrossRef] [PubMed]
  63. Maiti, P.; Scott, J.; Sengupta, D.; Al-Gharaibeh, A.; Dunbar, G. Curcumin and Solid Lipid Curcumin Particles Induce Autophagy, but Inhibit Mitophagy and the PI3K-Akt/mTOR Pathway in Cultured Glioblastoma Cells. Int. J. Mol. Sci. 2019, 20, 399. [Google Scholar] [CrossRef]
  64. Zhu, J.; Zhao, B.; Xiong, P.; Wang, C.; Zhang, J.; Tian, X.; Huang, Y. Curcumin Induces Autophagy via Inhibition of Yes-Associated Protein (YAP) in Human Colon Cancer Cells. Med. Sci. Monit. 2018, 24, 7035–7042. [Google Scholar] [CrossRef] [PubMed]
  65. Liu, R.; Zhang, H.B.; Yang, J.; Wang, J.R.; Liu, J.X.; Li, C.L. Curcumin alleviates isoproterenol-induced cardiac hypertrophy and fibrosis through inhibition of autophagy and activation of mTOR. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 7500–7508. [Google Scholar] [CrossRef] [PubMed]
  66. Hu, J.B.; Li, S.J.; Kang, X.Q.; Qi, J.; Wu, J.H.; Wang, X.J.; Xu, X.L.; Ying, X.Y.; Jiang, S.P.; You, J.; et al. CD44-targeted hyaluronic acid-curcumin prodrug protects renal tubular epithelial cell survival from oxidative stress damage. Carbohydr. Polym. 2018, 193, 268–280. [Google Scholar] [CrossRef] [PubMed]
  67. Wei, Y.; Gao, J.; Qin, L.; Xu, Y.; Shi, H.; Qu, L.; Liu, Y.; Xu, T.; Liu, T. Curcumin suppresses AGEs induced apoptosis in tubular epithelial cells via protective autophagy. Exp. Ther. Med. 2017, 14, 6052–6058. [Google Scholar] [CrossRef]
  68. Yao, Q.; Ke, Z.Q.; Guo, S.; Yang, X.S.; Zhang, F.X.; Liu, X.F.; Chen, X.; Chen, H.G.; Ke, H.Y.; Liu, C. Curcumin protects against diabetic cardiomyopathy by promoting autophagy and alleviating apoptosis. J. Mol. Cell. Cardiol. 2018, 124, 26–34. [Google Scholar] [CrossRef]
  69. Mukhopadhyay, S.; Panda, P.K.; Sinha, N.; Das, D.N.; Bhutia, S.K. Autophagy and apoptosis: Where do they meet? Apoptosis 2014, 19, 555–566. [Google Scholar] [CrossRef]
  70. Li, G.; Chen, L.; Chen, K. Curcumin Promotes Femoral Fracture Healing in a Rat Model by Activation of Autophagy. Med. Sci. Monit. 2018, 24, 4064–4072. [Google Scholar] [CrossRef]
  71. Zhang, J.; Wang, J.; Xu, J.; Lu, Y.; Jiang, J.; Wang, L.; Shen, H.M.; Xia, D. Curcumin targets the TFEB-lysosome pathway for induction of autophagy. Oncotarget 2016, 7, 75659–75671. [Google Scholar] [CrossRef] [PubMed]
  72. Song, J.X.; Sun, Y.R.; Peluso, I.; Zeng, Y.; Yu, X.; Lu, J.H.; Xu, Z.; Wang, M.Z.; Liu, L.F.; Huang, Y.Y.; et al. A novel curcumin analog binds to and activates TFEB in vitro and in vivo independent of MTOR inhibition. Autophagy 2016, 12, 1372–1389. [Google Scholar] [CrossRef] [PubMed]
  73. He, Y.; Lang, X.; Cheng, D.; Yang, Z. Curcumin ameliorates chronic renal failure in 5/6 nephrectomized rats by regulation of the mTOR/HIF-1α/VEGF signaling pathway. Biol. Pharm. Bull. 2019. [Google Scholar] [CrossRef] [PubMed]
  74. Vergadi, E.; Ieronymaki, E.; Lyroni, K.; Vaporidi, K.; Tsatsanis, C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J. Immunol. 2017, 198, 1006–1014. [Google Scholar] [CrossRef] [PubMed]
  75. Heras-Sandoval, D.; Pérez-Rojas, J.M.; Hernández-Damián, J.; Pedraza-Chaverri, J. The role of PI3K/AKT/mTOR pathway in the modulation of autophagy and the clearance of protein aggregates in neurodegeneration. Cell Signal. 2014, 26, 2694–2701. [Google Scholar] [CrossRef] [PubMed]

Article Metrics

Citations

Article Access Statistics

Multiple requests from the same IP address are counted as one view.