Document Type : Review

Authors

1 Department of Basic Science, Faculty of Veterinary Medicine, Islamic Azad Branch, University of Shushtar, Khuzestan, Iran

2 Department of Basic Science, Veterinary Medicine Faculty, Tabriz University, Tabriz, Iran.

Abstract

In recent decades, special attention has been paid to cell death mechanisms, with the exception of apoptosis. This could be due to the resistance of cells, particularly cancer cells, to apoptosis. Among novel pathways, autophagy and endoplasmic reticulum (ER) stress have attracted the attention of researchers. A large number of antitumor drugs have been developed based on their modulatory effects on autophagy and ER stress. On the other hand, ER stress could stimulate autophagy and apoptosis, which is indicative of the dual role of this pathway. Therefore, the monitoring of these pathways could contribute to the treatment of pathological conditions. Among the multiple synthetic and natural modulators of autophagy and ER stress, natural agents are used more extensively owing to their few side-effects, valuable biological activities, and cost-efficiency. Honokiol as a lignin extracted from the bark of magnolia tree. This compound has been reported to have antioxidant, anti-inflammatory, anti-diabetic, and antitumor effects. The present study aimed to first introduce honokiol, autophagy, and ER stress and assess the modulatory effects of honokiol on the autophagy and ER stress mechanisms so as to demonstrate the therapeutic efficacy of this natural compound.

Keywords

Introduction
There is an absolute need for organisms to maintain their homeostasis in physiological and pathological conditions. Regulated cell death (RCD) is involved in this homeostasis through the degradation of impaired or potentially toxic organelles and macromolecules. Furthermore, RCD is observed in physiological and pathological conditions in a large number of organisms, such as eukaryotes and some prokaryotes (1-13). In general, cell death is classified into three distinct categories, including apoptosis, autophagy, and necrosis (14).
Apoptosis has not exhibited a promising profile in cancer therapy. In an experiment performed by Roumane et al. (15), the results indicated that the effective treatment of cancer requires the proper understanding cell death mechanisms rather than apoptosis, which is mainly due to the proliferative effects of apoptosis on the surrounding surviving cells; this is referred to as apoptosis-induced proliferation. From an organelle standpoint, endoplasmic reticulum (ER) also plays a key role in homeostasis. As such, the proper targeting of these pathways and organelles could be beneficial in the treatment of the pathological conditions associated with the impairment of these pathways. It is of paramount importance to find compounds with modulatory effects on these mechanisms as a promising strategy in the management of pathological conditions. It is also notable that synthetic drugs have been reported to have numerous side-effects, whereas plant-derived products have shown favorable biological activities (16-27).
The present study aimed to describe the beneficial effects of honokiol as a natural compound on the targeting of autophagy and ER stress.

Literature Review
Molecular Mechanisms of Autophagy
Autophagy is a catabolic process, which is responsible for the monitoring and conservation of cellular energetic balance during the degradation of proteins and organelles in lysosomes (28-37). Autophagy is stimulated by starvation and other stress conditions so as to provide adequate energy and maintain survival (38). If the energy level is insufficient (nutrient deprivation), autophagy is activated to partly decompose its deposits for the provision of energy. The periodic incidence of autophagy occurs at least once during the day; for instance, autophagy is induced between meals in organs such as the liver in order to preserve its primary functions, while providing amino acids and energy as well (39-44). As an evolutionary process, autophagy is also observed in yeasts, and its main function is to respond to starvation.
Based on mechanisms and functions, autophagy is classified into three distinct types, including micro-autophagy, macro-autophagy (autophagy), and chaperone-mediated autophagy (CMA). In micro-autophagy, cytoplasmic structures are surrounded by the pre-existing lysosomes or vacuole membranes. In CMA, the guidance of cargos toward the lysosome is mediated by chaperone proteins, such as HSPA8 (HSC70). In macro-autophagy (autophagy), proteins and lipids are recruited from some intracellular membranes so as to form autophagosomes (double-membrane vesicles). Following that, they are infused with lysosomes to degrade their content (45-49).
Based on the procedures of cargo delivery to the lysosome, autophagy is divided into the selective and non-selective types. Seemingly, CMA is more selective and specific compared to micro- and macro-autophagy (50,51). Furthermore, a basal level of autophagy is required to maintain homeostasis and prevent the accumulation of impaired macromolecules and organelles in normal conditions. Therefore, the strict regulation of autophagy mechanisms is considered essential (39,40,52).
According to the literature, the PI3K/Akt/mTOR pathway is the most important regulator of autophagy. In normal conditions (i.e., balanced energy level), the mammalian target of rapamycin complex 1 (mTORC1) suppresses the autophagy process through the phosphorylation of Unc-51-like kinase 1/2 (ULK1/2) and autophagy-related gene 13 (ATG13). In addition, class-I PI3K (phosphatidylinositol-3-phosphate kinase) and class-III PI3K are considered to be the inhibitors and stimulators of autophagy, respectively (53,54).
Under stress conditions (e.g., starvation), ATG1 activates autophagy (55,56). After activation, ATG1 produces the phagophore (cup-shaped structure) that surrounds the cargo. Afterwards, this complex phosphorylates the Beclin-1 at Ser14 residues in order to accelerate the formation of autophagosome through the activation of other ATGs on the membranes of the phagophore (57). Following that, the signal of the triggering autophagy is induced via the ubiquitination and degradation of ULK-1 by Cul3-KLHL20 ligase complex (58). At this phase, autophagosome has been formed and is in the nucleation stage, where the VPS34-Beclin-1 complex plays a remarkable role (59,60). Finally, the elongation of the autophagosome occurs, in which two ubiquitin-like conjugation systems (ATG5-ATG12 and light chain 3 complexes [LC3]) are involved (61).

Molecular Mechanisms of Endoplasmic Reticulum Stress
The regulation of protein homeostasis is referred to as proteostasis, which is a mechanism investigating the synthesis, folding, assembly, translocation, and decomposition of proteins. Endoplasmic reticulum (ER) is an organized, double-membrane compartment that is involved in the proper folding of a large number of eukaryotic cell proteins (proteostasis), Ca2+ storage and release, and lipid and carbohydrate metabolism (62-64). In this regard, it is of utmost importance to consider the key role of these proteins as some must lodge in ER, Golgi apparatus, lysosomes, and plasma membrane, while the others are involved in intracellular signaling pathways, which confirms their pivotal role in cellular processes as well. Therefore, it is crucial to maintain ER homeostasis, which results in normal organismal physiology. The quality control mechanisms in ER guarantee the proper folding and assembly of proteins, and disturbances in ER homeostasis are mainly caused by redox imbalance, protein folding defects, infections, hypoxia, and impairments in Ca2+ homeostasis, which in turn lead to ER stress. In order to restore normal ER function, the quality control mechanisms of ER are activated, including the unfolding of the protein response (UPR) and ER-associated degradation (ERAD) (65-71).
Three important pathways are available to preserve ER homeostasis. Inositol-requiring protein-1 (IRE1) is a membrane-localized kinase/endoribonuclease pathway, which affects ER homeostasis through alterations in mRNA, so that IRE1 converts the transcription factor X-box binding protein 1 (XBP1) into its active form through eliminating a specific intron from the mRNA of XBP1. Following that, XBP1 activates the transcription of the genes that are associated with protein degradation and folding (72-74). In addition, IRE1 uses regulated IRE1-dependent decay, which is a process for the decomposition of ER-localized mRNAs, in order to decrease the synthesis of proteins. The subsequent reduction in protein synthesis is associated with diminution in the required folding in ER, while the quality control mechanisms of ER achieve this time to degrade and re-fold the misfolded and unfolded proteins (75). Following the mentioned strategy of IRE1 for the stimulation of the alterations in the transcription factor, c-Jun N-terminal kinase (JNA) signaling pathway are affected (76).
Protein kinase RNA-like ER (PERK) is another major pathway, which is involved in the reduction of the protein load into ER through phosphorylation, which in turn leads to the suppression of the eukaryotic translation initiation factor 2α (elF2α) (77). During ER stress, stimulating transcription factor 6 (ATF6) is transferred to the Golgi apparatus, where it is converted into its active form by cleavage. Afterwards, ATF6 is translocated to the nucleus, positively affecting the expression of the genes associated with protein and lipid synthesis. ATF6 and PERK have been reported to stimulate the expression of a pro-apoptotic factor, known as CCAAT/enhancerbinding protein homologous protein (CHOP) (78,79).

Association of ER Stress with Autophagy
Previous findings have confirmed the induction of autophagy by ER stress. However, it is essential to determine whether the autophagy induction under ER stress is associated with cell death or cell survival. In a study, Song et al. investigated the role of intermittent-hypoxia (IH)-induced autophagy on pancreatic cells (80), reporting that IH could enhance the autophagy level and increase the concentrations of the proteins associated with ER stress (e.g., CHOP, PERK, p-elF2, and ATF4). The inhibition of these signaling pathways resulted in the inhibition of autophagy, indicating that ER stress could stimulate autophagy. In the mentioned study, rapamycin and chloroquine were used as the stimulator and inhibitor of autophagy, respectively in order to investigate the effects of autophagy on cell viability. Interestingly, the increased level of autophagy by rapamycin decreased the cell death caused by IH, and the reduced autophagy by chloroquine was reported to increase cell death in pancreatic cells. Therefore, it was concluded that autophagy induction by IH through ER stress plays a key role in the protection of pancreatic cells against cell death.
In this regard, the findings of Li et al. were inconsistent with the mentioned research (81). According to the latter, sodium fluoride could increase the level of ER stress in MC3T3-E1 osteoblastic cells, demonstrating that the increased level of ER stress is associated with higher autophagy and apoptosis in osteoblastic cells, which confirms the adverse effects of ER stress-induced autophagy. In fact, Li et al. claimed that the ameliorative or adverse effects of autophagy depend on the excitation level of autophagy, so that a specific level of autophagy induction could alleviate ER stress, while high levels of autophagy induction are associated with the self-digestion of the cell and apoptosis.

Honokiol
Honokiol is a lignan, which is extracted from the bark of magnolia tree. It is a small molecule with the molecular weight of 266 and molecular formula of C18H18O2. In recent decades, multiple derivatives of honokiol with antitumor activities have been developed, including 3/-formyl-honokiol, 5-formyl-honokiol, and 3,5/-diformyl-honokiol (82). Honokiol has remarkable biological properties, including anti-inflammatory (83,84), antimicrobial (85), antioxidant (86), hepatoprotective (87, 88), neuroprotective (81,89), and protective effects against thrombosis (90) and angiopathy (91-95).
According to the literature, honokiol was extensively used in Chinese traditional medicine for the treatment of some diseases, such as thrombotic stroke, gastrointestinal disorders, anxiety, and nervous system impairment (96,97). Moreover, honokiol is a popular compound in Japan with extensive usage (98,99).
Recently, Cen et al. examined the effects of honokiol on lung squamous cell carcinoma (100), reporting that honokiol exerted inhibitory effects on the FGF2-FGFR1 signaling pathway, thereby stimulating apoptosis in lung squamous cell carcinoma and decreasing the viability and proliferation of cancer cells.
Honokiol is also beneficial in the prevention of type II diabetes (101). Previous findings have indicated that the consumption of honokiol is associated with the reduction of the abnormal alterations of hepatic cytochrome P450 and transporter mRNA expression (e.g., hepatic Oat2 and Oatp2b1) in rats with type II diabetes induced by a high-fat diet and streptozotocin, which are involved in the incidence and development of diabetes. Furthermore, honokiol has been reported to ameliorate insulin resistance, nonalcoholic steatosis, and liver dysfunction (102-104). Considering its anti-inflammatory properties, honokiol could significantly inhibit the expression of cyclooxygenase-2 and activation of nuclear factor-kappa B (105).

Honokiol and Autophagy
Several studies have confirmed the protective and antitumor effects of honokiol through autophagy (Figure 1). For instance, XQ et al. examined the beneficial effects of honokiol on anti-b1-adrenergic receptor autoantibody-induced myocardial dysfunction (106), reporting that honokiol treatment could inhibit b1-AAB-induced effects, conserving the myocardial tissues against dysfunction. Moreover, honokiol could enhance the contractile ability of the heart and remarkably decrease the activity of lactate dehydrogenase.
In another study, the potential antitumor activity of honokiol in human thyroid cancer cells was evaluated (107), and the obtained results indicated that honokiol could stimulate autophagy in human thyroid cancer cells via the Akt/mTOR signaling pathway, thereby reducing the viability and proliferation of these cancer cells.
Honokiol and ER Stress
In general, honokiol uses the ER stress pathway to exert its protective or antitumor effects (Figure 2). In a study in this regard, Chiu et al. applied honokiol to suppress tumor growth and metastasis (117). According to the obtained results, honokiol stimulated ER stress through CHOP activation, thereby significantly decreasing the viability of melanoma cells and suppressing the epithelial mesenchymal transition of these cancer cells. Similarly, Jangra et al. investigated the neuroprotective effects of honokiol mediated by ER stress (118), reporting that honokiol significantly reduced the CHOP level in the hippocampus of stressed mice in order to inhibit ER stress, which in turn results in the inhibition of cognitive impairment and depressive behaviors in the stressed mice. Other findings in this regard have demonstrated that honokiol exerts stimulatory effects on ER stress through enhancing the level of glucose-regulated protein 78, leading to the induction of autophagy and reduced migration of neuroblastoma cells (93).

Conclusion
ER stress is involved in the pathogenesis of some disorders, such as neurological disorders, pulmonary fibrosis, and cancer. In these pathological conditions, UPR is activated to partly restore the homeostasis of ER by the proper folding of proteins and through the degradation of unfolded and misfolded proteins in severe conditions. In addition, autophagy is associated with pathological conditions; for instance, in Alzheimer’s disease, the low level or lack of autophagy contributes to the aggregation of amyloid-α. Therefore, the targeting of these mechanisms could be beneficial in the treatment of various pathological conditions. According to the information in Table 2, honokiol has stimulatory and inhibitory effects on ER stress in two modes depending on the condition. As such, it plays a key role in maintaining homeostasis and proteostasis. According to the information in Table 1, in some conditions, honokiol could stimulate autophagy, while in other conditions, it inhibits autophagy. In both its modes, honokiol exerts protective and antitumor effects. Further investigations are required in order to clarify the association of honokiol with ER stress and autophagy, with the findings of this review laying the groundwork in this regard.

Acknowledgements
None.

Conflict of Interest
The authors declare no conflict of interest.

  1. Fuchs Y, Steller H. Programmed cell death in animal development and disease. Cell. 2011; 147:742-758.
  2. Galluzzi L, Bravo-San Pedro JM, Kepp O, et al. Regulated cell death and adaptive stress responses. Cell Mol Life Sci. 2016; 73:2405-2410.
  3. Galluzzi L, López-Soto A, Kumar S, et al. Caspases connect cell-death signaling to organismal homeostasis. Immunity 2016;44:221-231.
  4. Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 2018;25:486-541.
  5. Jorgensen I, Rayamajhi M, Miao EA. Programmed cell death as a defence against infection. Nat Rev Immunol. 2017;17:151-164.
  6. Nagata S, Tanaka M. Programmed cell death and the immune system. Nat Rev Immunol. 2017;17:333-340.
  7. Büttner S, Eisenberg T, Herker E, et al. Why yeast cells can undergo apoptosis: death in times of peace, love, and war. J Cell Biol. 2006; 175:521-525.
  8. Cornillon S, Foa C, Davoust J, et al. Programmed cell death in Dictyostelium. J Cell Sci. 1994; 107:2691-2704.
  9. Cornillon S, Olie RA, Golstein P. An insertional mutagenesis approach to Dictyostelium cell death. Cell Death Differ. 1998;5:416-425.
  10. Eisenberg T, Büttner S, Kroemer G, et al. The mitochondrial pathway in yeast apoptosis. Apoptosis. 2007;12:1011-1023.
  11. Green DR, Fitzgerald P. Just so stories about the evolution of apoptosis. Curr Biol. 2016;26:R620-R627.
  12. Madeo F, Fröhlich E, Fröhlich K-U. A yeast mutant showing diagnostic markers of early and late apoptosis. J Cell Biol. 1997;139:729-734.
  13. Olie R, Durrieu F, Cornillon S, et al. Apparent caspase independence of programmed cell death in Dictyostelium. Curr Biol. 1998; 8:955-S951.
  14. Martin-Sanchez D, Poveda J, Fontecha-Barriuso M, et al. Targeting of regulated necrosis in kidney disease. Nefrología. 2018; 38:125-135.
  15. Roumane A, Berthenet K, El Fassi C, et al. Caspase-independent cell death does not elicit a proliferative response in melanoma cancer cells. BMC cell biol. 2018;19:11.
  16. Abdollahzadeh Soreshjani S, Ashrafizadeh M. The Effects of the Exercise on the Testosterone Level, Heat Shock Proteins and Fertility Potential. Rev Clin Med. 2018; 5:12-15.
  17. Ahmadi Z, Ashrafizadeh M. Downregulation of Osteocalcin Gene in Chickens Treated with Lead Acetate II. IBBJ. 2018; 4:177-182.
  18. Ahmadi Z, Ashrafizadeh M. Down Regulation of Osteocalcin Gene in Chickens Treated with Cadmium. Iran J Toxicol. 2019;13:1-4.
  19. Ahmadi Z, Mohammadinejad R, Ashrafizadeh M. Drug delivery systems for resveratrol, a non-flavonoid polyphenol: Emerging evidence in last decades. J Drug Delivery Sci Technol. 2019;51:591-604.
  20. Ashrafizadeh M, Ahmadi Z. The effects of astaxanthin treatment on the sperm quality of mice treated with nicotine. Rev Clin Med. 2019; 6:156-158.
  21. Ashrafizadeh M, Rafiei H, Ahmadi Z. Histological changes in the liver and biochemical parameters of chickens treated with lead acetate II. Iran J Toxicol. 2018; 12:1-5.
  22. Rafiei H, Ahmadi Z, Ashrafizadeh M. Effects of orally administered lead acetate II on rat femur histology, mineralization properties and expression of osteocalcin gene. Int Biol Biomed J. 2018; 4:149-155.
  23. Rafiei H, Ashrafizadeh M. Expression of collagen type II and osteocalcin genes in mesenchymal stem cells from rats treated with lead acetate II. Iran J Toxicol. 2018;12:35-40.
  24. Sobhani B, Roomiani S, Ahmadi Z, et al. Histopathological Analysis of Testis: Effects of Astaxanthin Treatment against Nicotine Toxicity. Iran J Toxicol. 2019; 13:41-44.
  25. Abdollahzadeh Soreshjani S, Ashrafizadeh M. Effects of exercise on testosterone level, heat shock protein, and fertility potential. Rev Clin Med. 2018; 5:141-145.
  26. Mohammadinejad R, Ahmadi Z, Tavakol S, et al. Berberine as a potential autophagy modulator. J Cell Physiol. 2019 Feb 15. doi: 10.1002/jcp.28325.
  27. Mohammadinejad R, Dadashzadeh A, Moghassemi S, et al. Shedding light on gene therapy: carbon dots for the minimally invasive image-guided delivery of plasmids and noncoding RNAs. J Adv Res. 2019;18:81-93.
  28. Mizushima N, Levine B, Cuervo AM, et al. Autophagy fights disease through cellular self-digestion. Nature. 2008;451:1069-1075.
  29. Limpert AS, Lambert LJ, Bakas NA, et al. Autophagy in cancer: regulation by small molecules. Trends Pharmacol Sci. 2018;39:1021-1032.
  30. Ren J, Zhang Y. Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci. 2018;39:1064-1076.
  31. Vidal RL, Matus S, Bargsted L, et al. Targeting autophagy in neurodegenerative diseases. Trends Pharmacol Sci. 2014; 35:583-591.
  32. Moosavi MA, Haghi A, Rahmati M, et al. Phytochemicals as potent modulators of autophagy for cancer therapy. Cancer lett. 2018; 424:46-69.
  33. Marzban H. Development of the Cerebellum from Molecular Aspects to Diseases. In: Moosavi MA, Rahmati M, Ashtari N, et al, editors. Apoptosis, Autophagy, and Unfolded Protein Response and Cerebellar Development. Development of the Cerebellum from Molecular Aspects to Diseases. Switzerland: Springer; 2017 p. 153-178.
  34. Mohammadinejad R, Moosavi MA, Tavakol S, et al. Necrotic, apoptotic and autophagic cell fates triggered by nanoparticles. Autophagy. 2019; 15:4-33.
  35. Hosseinpour‐Moghaddam K, Caraglia M, Sahebkar A. Autophagy induction by trehalose: Molecular mechanisms and therapeutic impacts. J Cell Physiol. 2018; 233:6524-6543.
  36. Shakeri A, Cicero AF, Panahi Y, et al. Curcumin: a naturally occurring autophagy modulator. J Cell Physiol. 2019;234:5643-5654.
  37. Zhou B, Liu J, Kang R, et al. Ferroptosis is a type of autophagy-dependent cell death. Semin Cancer Biol. 2019 Mar 14. pii: S1044-579X(19)30006-9.
  38. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet. 2009; 43:67-93.
  39. Botti J, Djavaheri-Mergny M, Pilatte Y, et al. Autophagy signaling and the cogwheels of cancer. Autophagy. 2006;2:67-73.
  40. Gozuacik D, Kimchi A. Autophagy and cell death. Curr Top Dev Biol. 2007;78:217-245.
  41. Kuma A, Hatano M, Matsui M, et al. The role of autophagy during the early neonatal starvation period. Nature. 2004; 432:1032-1036.
  42. Massey AC, Zhang C, Cuervo AM. Chaperone‐mediated autophagy in aging and disease. Curr Top Dev Biol. 2006; 73:205-235.
  43. Mizushima N, Klionsky DJ. Protein turnover via autophagy: implications for metabolism. Annu Rev Nutr. 2007;27:19-40.
  44. Yorimitsu T, Klionsky DJ. Autophagy: molecular machinery for self-eating. Cell Death Differ. 2005;12:1542-1552.
  45. Bandyopadhyay U, Kaushik S, Varticovski L, et al. The chaperone-mediated autophagy receptor organizes in dynamic protein complexes at the lysosomal membrane. Mol Cell Biol. 2008; 28:5747-5763.
  46. Feng Y, He D, Yao Z, et al. The machinery of macroautophagy. Cell Res. 2014;24:24-41.
  47. Galluzzi L, Baehrecke EH, Ballabio A, et al. Molecular definitions of autophagy and related processes. EMBO J. 2017; 36:1811-1836.
  48. Galluzzi L, Bravo-San Pedro JM, Levine B, et al. Pharmacological modulation of autophagy: therapeutic potential and persisting obstacles. Nat Rev Drug Discov. 2017;16:487-511.
  49. Ravanan P, Srikumar IF, Talwar P. Autophagy: the spotlight for cellular stress responses. Life sci. 2017; 188:53-67.
  50. Sica V, Galluzzi L, Bravo-San Pedro JM, et al. Organelle-specific initiation of autophagy. Mol cell. 2015; 59:522-539.
  51. Zaffagnini G, Martens S. Mechanisms of selective autophagy. J Mol Biol. 2016; 428:1714-1724.
  52. Meijer AJ, Codogno P. Signalling and autophagy regulation in health, aging and disease. Mol Aspects Med. 2006; 27:411-425.
  53. Klionsky DJ. Autophagy: from phenomenology to molecular understanding in less than a decade. Nat Rev Mol Cell Biol. 2007;8:931-937.
  54. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008; 132:27-42.
  55. Klionsky DJ. The molecular machinery of autophagy: unanswered questions. J Cell Sci. 2005; 118:7-18.
  56. Yang Z, Klionsky DJ. Mammalian autophagy: core molecular machinery and signaling regulation. Curr Opin Cell Biol. 2010;22:124-131.
  57. Russell RC, Tian Y, Yuan H, et al. ULK1 induces autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat Cell Biol. 2013;15:741-750.
  58. Liu CC, Lin YC, Chen YH, et al. Cul3-KLHL20 ubiquitin ligase governs the turnover of ULK1 and VPS34 complexes to control autophagy termination. Mol cell. 2016; 61:84-97.
  59. Di Bartolomeo S, Corazzari M, Nazio F, et al. The dynamic interaction of AMBRA1 with the dynein motor complex regulates mammalian autophagy. J Cell Biol. 2010; 191:155-168.
  60. Glick D, Barth S, Macleod KF. Autophagy: cellular and molecular mechanisms. J Pathol. 2010; 221:3-12.
  61. Otomo C, Metlagel Z, Takaesu G, et al. Structure of the human ATG12~ ATG5 conjugate required for LC3 lipidation in autophagy.Nat Struct Mol Biol. 2013;20:59-66. 
  62. Rahmati M, Amanpour S, Kharman-Biz A, et al. Endoplasmic reticulum stress as a therapeutic target in cancer: a mini review. Basic Clin Cancer Res. 2017; 9:38-48.
  63. Rahmati M, Moosavi MA, McDermott MF. ER Stress: A Therapeutic Target in Rheumatoid Arthritis? Trends Pharmacol Sci. 2018;39:610-623.
  64. Mollazadeh H, Atkin SL, Butler AE, et al. The effect of statin therapy on endoplasmic reticulum stress. Pharmacol Res. 2018;137:150-158.
  65. Chakrabarti A, Chen AW, Varner JD. A review of the mammalian unfolded protein response. Biotechnol Bioeng. 2011; 108:2777-2793.
  66. Kelsen SG. The unfolded protein response in chronic obstructive pulmonary disease. Ann Am Thorac Soc. 2016; 13:S138-S145.
  67. Fujita E, Kouroku Y, Isoai A, et al. Two endoplasmic reticulum-associated degradation (ERAD) systems for the novel variant of the mutant dysferlin: ubiquitin/proteasome ERAD (I) and autophagy/lysosome ERAD (II). Hum Mol Genet.2007;16:618-629.
  68. Schröder M, Kaufman RJ. ER stress and the unfolded protein response. Mutat Res. 2005;569:29-63.
  69. Zhang B, Wang XQ, Chen HY, et al. Involvement of the Nrf2 pathway in the regulation of pterostilbene-induced apoptosis in HeLa cells via ER stress. J Pharmacol Sci. 2014; 126:216-229.
  70. Zhang S, Zheng H, Chen Q, et al. The lectin chaperone calnexin is involved in endoplasmic reticulum stress response by regulating Ca2+ homeostasis in Aspergillus nidulans. Appl Environ Microbiol. 2017;83: e00673-17.
  71. Zhong Q, Zhou B, Ann DK, et al. Role of endoplasmic reticulum stress in epithelial–mesenchymal transition of alveolar epithelial cells: effects of misfolded surfactant protein. Am J Respir Cell Mol Biol. 2011; 45:498-509.
  72. Calfon M, Zeng H, Urano F, et al. IRE1 couples endoplasmic reticulum load to secretory capacity by processing the XBP-1 mRNA. Nature. 2002; 415:92-96.
  73. Cox JS, Walter P. A novel mechanism for regulating activity of a transcription factor that controls the unfolded protein response. Cell. 1996;87:391-404.
  74. Sidrauski C, Walter P. The transmembrane kinase Ire1p is a site-specific endonuclease that initiates mRNA splicing in the unfolded protein response. Cell. 1997;90:1031-1039.
  75. Hollien J, Lin JH, Li H, et al. Regulated Ire1-dependent decay of messenger RNAs in mammalian cells. J Cell Biol. 2009; 186:323-331.
  76. Urano F, Wang X, Bertolotti A, et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science. 2000; 287:664-666.
  77. Harding HP, Zhang Y, Bertolotti A, et al. Perk is essential for translational regulation and cell survival during the unfolded protein response. Mol Cell. 2000; 5:897-904.
  78. Xu C, Bailly-Maitre B, Reed JC. Endoplasmic reticulum stress: cell life and death decisions. J Clin Invest. 2005; 115:2656-2664.
  79. Yoshida H. ER stress and diseases. FEBS J. 2007; 274:630-658.
  80. Song S, Tan J, Miao Y, et al. Intermittent-Hypoxia-Induced Autophagy Activation Through the ER-Stress-Related PERK/eIF2α/ATF4 Pathway is a Protective Response to Pancreatic β-Cell Apoptosis. Cell Physiol Biochem. 2018; 51:2955-2971.
  81. Li X, Meng L, Wang F, et al. Sodium fluoride induces apoptosis and autophagy via the endoplasmic reticulum stress pathway in MC3T3-E1 osteoblastic cells. Mol Cell Biochem. 2019;454:77-85.
  82. Ma L, Chen J, Wang X, et al. Structural modification of honokiol, a biphenyl occurring in Magnolia officinalis: the evaluation of honokiol analogues as inhibitors of angiogenesis and for their cytotoxicity and structure–activity relationship. J Med Chem. 2011; 54:6469-6481.
  83. Fang JY, Huang TH, Hung CF, et al. Derivatization of honokiol by integrated acetylation and methylation for improved cutaneous delivery and anti-inflammatory potency. Eur J Pharm Sci. 2018; 114:189-198.
  84. Wijesuriya YK, Lappas M. Potent anti-inflammatory effects of honokiol in human fetal membranes and myometrium. Phytomedicine. 2018;49:11-22.
  85. Liao K, Sun L. Roles of the Hsp90-Calcineurin Pathway in the Antifungal Activity of Honokiol. J Microbiol Biotechnol. 2018;28:1086-1093.
  86. Ramachandran C, Wilk B, Melnick SJ, et al. Synergistic antioxidant and anti-inflammatory effects between modified citrus pectin and honokiol. Evid Based Complement Alternat Med. 2017;2017:8379843.
  87. Jeong YH, Hur HJ, Jeon EJ, et al. Honokiol Improves Liver Steatosis in Ovariectomized Mice. Molecules. 2018; 23:194.
  88. Liu JX, Shen SN, Tong Q, et al. Honokiol protects hepatocytes from oxidative injury through mitochondrial deacetylase SIRT3. Eur J Pharmacol. 2018; 834:176-187.
  89. Hou Y, Peng S, Li X, et al. Honokiol alleviates oxidative stress-induced neurotoxicity via activation of Nrf2. ACS Chem Neurosci. 2018;9:3108-3116.
  90. Hu H, Zhang Xx, Wang Yy, et al. Honokiol inhibits arterial thrombosis through endothelial cell protection and stimulation of prostacyclin. Acta Pharmacol Sin. 2005;26:1063-1068.
  91. Fried LE, Arbiser JL. Honokiol, a multifunctional antiangiogenic and antitumor agent. Antioxid Redox Signal. 2009; 11:1139-1148.
  92. Lee TY, Chang CC, Lu WJ, et al. Honokiol as a specific collagen receptor glycoprotein VI antagonist on human platelets: Functional ex vivo and in vivo studies. Sci Rep. 2017; 7:40002.
  93. Yeh PS, Wang W, Chang YA, et al. Honokiol induces autophagy of neuroblastoma cells through activating the PI3K/Akt/mTOR and endoplasmic reticular stress/ERK1/2 signaling pathways and suppressing cell migration. Cancer lett. 2016; 370:66-77.
  94. Liou KT, Lin SM, Huang SS, et al. Honokiol ameliorates cerebral infarction from ischemia-reperfusion injury in rats. Planta med. 2003;69:130-134.
  95. Sulakhiya K, Kumar P, Gurjar SS, et al. Beneficial effect of honokiol on lipopolysaccharide induced anxiety-like behavior and liver damage in mice. Pharmacol Biochem Behav. 2015; 132:79-87.
  96. Cheng Z. Comparative studies on the interactions of honokiol and magnolol with human serum albumin. J Pharm Biomed Anal. 2012;66:240-251.
  97. Yang B, Ni X, Chen L, et al. Honokiol-loaded polymeric nanoparticles: an active targeting drug delivery system for the treatment of nasopharyngeal carcinoma. Drug deliv. 2017;24:660-669.
  98. Lee YJ, Lee YM, Lee CK, et al. Therapeutic applications of compounds in the Magnolia family. Pharmacol Ther. 2011; 130:157-176.
  99. Xu C, Tang Y, Hu W, et al. Investigation of inclusion complex of honokiol with sulfobutyl ether-β-cyclodextrin. Carbohydr Polym. 2014; 113:9-15.
  100. Cen M, Yao Y, Cui L, et al. Honokiol induces apoptosis of lung squamous cell carcinoma by targeting FGF2‐FGFR1 autocrine loop. Cancer Med. 2018;7:6205-6218.
  101. Wang J, Zhai T, Chen Y. Effects of Honokiol on CYP450 Activity and Transporter mRNA Expression in Type 2 Diabetic Rats. Int J Mol Sci. 2018; 19:815.
  102. Kim YJ, Choi MS, Cha BY, et al. Long‐term supplementation of honokiol and magnolol ameliorates body fat accumulation, insulin resistance, and adipose inflammation in high‐fat fed mice. Mol Nutr Food Res. 2013; 57:1988-1998.
  103. Lee JH, Jung JY, Jang EJ, et al. Combination of honokiol and magnolol inhibits hepatic steatosis through AMPK-SREBP-1 c pathway. Exp Biol Med (Maywood). 2015;240:508-518.
  104. Seo MS, Kim JH, Kim HJ, et al. Honokiol activates the LKB1–AMPK signaling pathway and attenuates the lipid accumulation in hepatocytes. Toxicol Appl Pharmacol. 2015; 284:113-124.
  105. Murakami Y, Kawata A, Seki Y, et al. Comparative inhibitory effects of magnolol, honokiol, eugenol and bis-eugenol on cyclooxygenase-2 expression and nuclear factor-kappa B activation in RAW264. 7 macrophage-like cells stimulated with fimbriae of Porphyromonas gingivalis. In Vivo. 2012;26:941-950.
  106. Wei Xq, Zhang Hs, Wei Gh, et al. Honokiol Protects against Anti-β1-Adrenergic Receptor Autoantibody-Induced Myocardial Dysfunction via Activation of Autophagy. Oxid Med Cell Longev. 2018;2018:1640804.
  107. Lu CH, Chen SH, Chang YS, et al. Honokiol, a potential therapeutic agent, induces cell cycle arrest and program cell death in vitro and in vivo in human thyroid cancer cells. Pharmacol Res. 2017;115:288-298.
  108. Luo LX, Li Y, Liu ZQ, et al. Honokiol induces apoptosis, G1 arrest, and autophagy in KRAS mutant lung cancer cells. Front Pharmacol. 2017; 8:199.
  109. Lin CJ, Chen TL, Tseng YY, et al. Honokiol induces autophagic cell death in malignant glioma through reactive oxygen species-mediated regulation of the p53/PI3K/Akt/mTOR signaling pathway. Toxicol Appl Pharmacol. 2016; 304:59-69.
  110. Chio CC, Chen KY, Chang CK, et al. Improved effects of honokiol on temozolomide-induced autophagy and apoptosis of drug-sensitive and-tolerant glioma cells. BMC cancer 2018; 18:379.
  111. Huang K, Chen Y, Zhang R, et al. Honokiol induces apoptosis and autophagy via the ROS/ERK1/2 signaling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis. 2018; 9:157.
  112. Huang KJ, Kuo CH, Chen SH, et al. Honokiol inhibits in vitro and in vivo growth of oral squamous cell carcinoma through induction of apoptosis, cell cycle arrest and autophagy. J Cell Mol Med. 2018; 22:1894-1908.
  113. Cheng YC, Hueng DY, Huang HY, et al. Magnolol and honokiol exert a synergistic anti-tumor effect through autophagy and apoptosis in human glioblastomas.Oncotarget. 2016;7:29116-29130.
  114. Lv X, Liu F, Shang Y, et al. Honokiol exhibits enhanced antitumor effects with chloroquine by inducing cell death and inhibiting autophagy in human non-small cell lung cancer cells. Oncol Rep. 2015; 34:1289-1300.
  115. Hahm ER, Sakao K, Singh SV. Honokiol activates reactive oxygen species‐mediated cytoprotective autophagy in human prostate cancer cells. Prostate 2014; 74:1209-1221.
  116. Chang KH, Yan MD, Yao CJ, et al. Honokiol‑induced apoptosis and autophagy in glioblastoma multiforme cells. Oncol lett. 2013; 6:1435-1438.
  117. Chiu CS, Tsai CH, Hsieh MS, et al. Exploiting Honokiol-induced ER stress CHOP activation inhibits the growth and metastasis of melanoma by suppressing the MITF and β-catenin pathways. Cancer lett. 2019;442:113-125.
  118. Jangra A, Dwivedi S, Sriram CS, et al. Honokiol abrogates chronic restraint stress-induced cognitive impairment and depressive-like behaviour by blocking endoplasmic reticulum stress in the hippocampus of mice. Eur J Pharmacol. 2016;770:25-32.
  119. Liu SH, Lee WJ, Lai DW, et al. Honokiol confers immunogenicity by dictating calreticulin exposure, activating ER stress and inhibiting epithelial‐to‐mesenchymal transition. Mol Oncol. 2015; 9:834-849.
  120. Huang KH, Weng TI, Huang HY, et al. Honokiol attenuates torsion/detorsion-induced testicular injury in rat testis by way of suppressing endoplasmic reticulum stress-related apoptosis. Urology. 2012;79:967.e5-11.
  121. Weng TI, Wu HY, Chen BL, et al. Honokiol attenuates the severity of acute pancreatitis and associated lung injury via acceleration of acinar cell apoptosis. Shock .2012; 37:478-484.
  122. Chen YJ, Wu CL, Liu JF, et al. Honokiol induces cell apoptosis in human chondrosarcoma cells through mitochondrial dysfunction and endoplasmic reticulum stress. Cancer lett. 2010; 291:20-30.