Autophagy Modulation Mechanism and Application: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Oncology
Contributor: , , ,

Autophagy is a vital cellular process that functions to degrade and recycle damaged organelles into basic metabolites. This allows a cell to adapt to a diverse range of challenging conditions. Autophagy assists in maintaining homeostasis, and it is tightly regulated by the cell. The disruption of autophagy has been associated with many diseases, such as neurodegenerative disorders and cancer. 

  • autophagy
  • glioblastoma
  • treatment
  • JAK2/STAT3 signaling pathway
  • PI3K/AKT/mTOR signaling pathway

1. Introduction

Gliomas are considered the most common primary brain tumors in adults, and over half of all brain tumors are gliomas [1,2,3,4]. Glioblastoma multiforme (GBM) is regarded as one of the most aggressive and lethal types of gliomas [5,6,7,8]. Unfortunately, the cause of the disease is unknown in most GBM patients [9]. The current standard of care treatment includes surgical resection followed by radiotherapy and chemotherapy. Even after treatment, patients with GBM typically have a median survival of approximately 14 months [10,11,12]. In recent studies, glioblastoma has been classified into three subtypes based on transcriptome analysis of the GBM tumors: proneural, classical, and mesenchymal [13,14]. The mesenchymal subtype of GBM is associated with the worst prognosis due to its expression of neural stem cell markers and the creation of an inflammatory microenvironment with increased angiogenesis [15]. Recognizing and utilizing these molecular subtypes in GBM therapy could facilitate personalized treatment and improve patient outcomes. Research has shown that glioma cells exhibit a greater responsiveness to treatment through autophagy as opposed to apoptosis [16], making the autophagy pathway a compelling target in the GBM treatments. Also, according to a recent whole transcriptome expression data analysis, high expression levels of autophagy-related genes are detected in classical and mesenchymal subtypes, the more malignant tumor subtypes [17,18].
Autophagy is a natural cellular process that plays a crucial role in maintaining cellular homeostasis, adapting to stress conditions, and promoting cell survival [19,20]. There are three different primary types of autophagy, which include microautophagy, chaperone-mediated autophagy, and macroautophagy [21]. These pathways are differentiated by the method by which cytosolic cargoes are delivered to the lysosome [22]. Microautophagy involves the lysosome utilizing direct membrane protrusion to take up cytoplasmic material for degradation [23]. Microautophagy has gained significant attention in recent research endeavors. It not only relates to macroautophagy regulation [21] but also plays a role in cancer and neurodegenerative disorders [24,25,26,27]. Chaperone-mediated autophagy (CMA) is a selective form of autophagy that targets specific individual proteins for degradation [28,29]. CMA involves cytosolic proteins with KFERQ-like motifs that are recognized by chaperone HSC70, and these proteins are delivered to the lysosomal-associated membrane protein 2A (LAMP2A), which allows for the translocation of these proteins into the lysosome for degradation [30]. Quite a few proteins involved in neurodegenerative disorders, including α-synuclein [31,32,33] and β-amyloid precursor protein [32], contain this motif. Thus, inhibiting CMA in neurons impairs neuronal function and leads to the accumulation of harmful proteins, thereby raising susceptibility to Alzheimer’s disease [34]. Macroautophagy (autophagy) plays a key role in cell physiology. Some of these roles include the degradation and recycling of damaged organelles, as well as the maintenance of normal cellular homeostasis [35]. The basic mechanism of autophagy has been well documented and involves the formation of a cytosolic double-membrane vesicle known as the autophagosome, which fuses with lysosomes to degrade its contents [36]. Macroautophagy generally occurs at low levels and can be further induced under specific stress conditions. Macroautophagy deteriorates cytoplasmic material into metabolites that can be used in biosynthetic processes or energy production for cell survival [37]. While autophagy primarily functions as a cytoprotective mechanism for degrading damaged organelles, uncontrolled autophagy can lead to detrimental consequences for the cell.
Glioblastoma, the most common and aggressive brain tumor, is characterized by its highly invasive and therapy-resistant nature. Autophagy has been found to play a significant role in the various stages of glioblastoma development, including tumor initiation and progression, cell proliferation, and response to treatment. Elevated levels of autophagy have been associated with glioblastoma development and are correlated with poor glioblastoma survival [62]. Suppressing autophagy has shown promise in inhibiting glioblastoma development; for instance, the knockdown of key autophagy-related genes, such as Atg7, Atg13, or Ulk1, disrupts autophagy and inhibits glioblastoma development in KRAS-driven glioblastoma mouse models [63], indicating the critical role of autophagy in tumor initiation. In established tumors, conditions like hypoxia and nutrient deficiency can induce high autophagy activity. This autophagy activity enhancement could be induced by yes-associated protein (YAP)-mediated high mobility group box 1(HMGB1) translocation. When HMGB1 translocates from the nucleus to the cytoplasm, it enhances autophagy, thereby promoting GBM progression [63,64].

2. Exploring the Mechanism and Application of Autophagy Modulation through the JAK/STAT Pathway

2.1. JAK/STAT Signaling Pathway

JAK/STAT pathway is activated via Janus kinase/signal and heavily contributes to tumorigenic functions, including proliferation, anti-apoptosis, angiogenesis, stem cell maintenance, and immune suppression [73,74,75,76]. JAK-mediated pathway is activated when cytokines bind to JAK receptors located on the cell membrane, followed by receptor dimerization and cytoplasmic tail phosphorylation. The JAK family consists of non-receptor protein tyrosine kinases (PTKs), which include JAK1, JAK2, JAK3, and Tyk2 [70,77]. These kinases are upstream of STAT3 and are involved in the signal transduction of cytokines [70]. STAT proteins have SH2 domains that contain tyrosine phosphorylation sites, and their phosphorylation allows STAT to form dimers and alters STAT protein conformation to facilitate their DNA binding [78]. After tyrosine phosphorylation, STAT is translocated into the nucleus. In the nucleus, STAT proteins bind DNA elements and regulate the transcription of associated genes, such as Bcl2, c-Myc, and Mcl-1 [78,79,80]. The STAT family is composed of seven members, which include STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6 [81]. STATs 1, 2, 4, and 6 play limited roles, while STAT3 and STAT5 are involved in more functions, such as resistance to treatments [82,83]. STAT3 serves as an intracellular signaling hub and can be activated when JAK2 is stimulated by various cytokines (IL-6) and growth factors. Once JAK2 is activated, it can phosphorylate STAT3 at the tyrosine 705 residue [73,84]. STAT3 is the primary transcriptional regulator of autophagy genes in the nucleus [70,85], and it can inhibit autophagy via the upregulation of BCL2 [85].

2.2. Drugs Induce Cell Apoptosis via Inhibition of JAK/STAT Pathway

Along with understanding the molecular mechanism of the JAK/STAT pathway, it is also important to understand the drugs that can influence this pathway. Therefore, scholars can modulate autophagy during cancer treatment. Pimozide is an antipsychotic drug that functions as a dopamine antagonist, but it has also been shown to promote tumor apoptosis [78,87,88]. Through studies on GBM cells, such as U87, U251, Daoy, and GBM 28, pimozide has been shown to promote autophagy-mediated apoptosis in vivo and in vitro by inhibiting the JAK/STAT3 pathway [78]. When the JAK/STAT3 pathway is inhibited, the development of malignancies such as brain tumors is suppressed. This is because STAT3 regulates the transcription of various oncogenes, such as Mcl-1, c-Myc, and Bcl-2 [89]. Bcl-2 is cited as one of the anti-apoptotic proteins and forms a complex with Beclin 1 [90,91].
Another potential enhancer of glioma treatment that has been researched is curcumin. Curcumin is an herbal supplement extracted from turmeric with lipophilic characteristics, which allows it to penetrate the blood–brain barrier and potentially affect several glioma tumor processes, such as proliferation, cell death, metastasis, and chemoresistance [95,96,97,98,99]. One of the signaling pathways that curcumin affects is the JAK1,2/STAT3 pathway [99,100,101]. Curcumin can downregulate the JAK1,2/STAT3 signaling pathway and its downstream targets.

2.3. Drugs Enhance the Efficacy of TMZ via JAK/STAT Pathway

Temozolomide (TMZ) is considered the first-line drug for the clinical treatment of gliomas [94,104]. TMZ can induce autophagy by initiating DNA damage and triggering the activation of the ATM/AMPK/ULK1 signaling pathway, which, in turn, promotes autophagy [105,106]. However, TMZ resistance is one of the most difficult challenges to overcome in GBM treatment [107,108]. In order to enhance the chemosensitivity of GBM to TMZ, momelotinib (MTB) is used in combination with TMZ [108,109,110]. MTB is an aminopyrimidine derivative and has been shown to inhibit the phosphorylation of JAK2 and STAT3 in GBM U251 cells [111]. During the treatment, MTB enhances the chemosensitivity of GBM to TMZ via the inactivation of the MTB/JAK2/STAT3/Bcl2 axis [109,110]. This increased chemosensitivity of GBM cells was demonstrated via studies on mouse xenograft models. In the study, co-treatment with MTB and TMZ on GBM U251 cells led to enhanced autophagy followed by apoptosis due to the inhibition of JAK2 and STAT3 phosphorylation [111]. This particular experiment showed that MTB and TMZ co-treated xenografts significantly decreased tumor weight compared with TMZ treatment alone.
Pacritinib is a JAK2 inhibitor with blood–brain barrier permeability and shows promising effects as an inhibitor that can be paired with TMZ to improve the efficacy of TMZ [104,112]. The synergistic effect of combination treatment with pacritinib and TMZ was demonstrated in patient-derived GBM brain-tumor-initiating cells (BTICs) [112]. When BTICs were treated with either 10 μg/mL of TMZ, 1 μM of pacritinib, or a combination of 1 μM pacritinib and 10 μg/mL of TMZ, Western blot protein analysis detected a significant increase in STAT3 activation in the TMZ-only treatment group.

3. Unveiling Autophagy Modulation Mechanism and Application via PI3K/AKT/mTOR Pathway

3.1. PI3K/AKT/mTOR Signaling Pathway

In addition to the JAK/STAT pathway, the phosphatidylinositol 3-kinase /AKT/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway is also involved in autophagy modulation [36,114,115,116]. The PI3K/AKT/mTOR pathway is a modulated signaling pathway in many cancer types, known to regulate different cellular processes such as cell survival, proliferation, growth, metabolism, angiogenesis, and metastasis [117,118,119]. mTOR is a serine/threonine protein kinase, and it is a downstream effector of PI3K and AKT signaling. There are two mTOR complexes: mTORC1 and mTORC2. The function of mTORC2 is not well understood, but it is responsive to growth factors for controlling cytoskeleton organization [120,121]. Activated mTORC2 plays a modulatory role in relation to mTORC1 and phosphorylates AKT at Ser473. The phosphorylated AKT can inhibit TSC1/2, resulting in the hyperactivation of mTORC1 through Rheb. mTORC1 is considered a major negative regulator of autophagy, and the PI3K/AKT pathway is a major upstream modulator of mTORC1 [122]. The suppression of the PI3K/AKT/mTOR pathway promotes autophagy, while the activation of this pathway inhibits autophagy [36]. The PI3K/AKT/mTOR pathway regulates synaptic plasticity in the brain and plays an important role in the development of brain structure; thus, the dysregulation of this pathway is commonly reported in neurodegenerative pathologies [36].

3.2. Drugs Induce Autophagy-Mediated Cell Death by Inhibiting AKT/mTOR Pathway

Several drugs are involved in the PI3K/AKT/mTOR signaling pathway, including rapamycin, an mTORC1 inhibitor and autophagy modulator [125]. Rapamycin is produced by Streptomyces hygroscopicus, and was initially used as an antifungal compound [126,127]. Several studies were carried out to determine whether rapamycin could induce autophagy. In one of the studies, U87-MG cells were treated for 3 days with rapamycin at a dosage of 100 nmol/L, and the ultrastructure of the cells was visualized through electron microscopy [128]. EM revealed that the cells treated with rapamycin developed numerous autophagic vacuoles, but not chromatin fragmentation [128]. These results indicate that rapamycin exerts its effects through autophagy induction as opposed to apoptosis. Rapamycin functions by inducing autophagy through the inhibition of mTORC1 [125]. This autophagy induction initially leads to an antitumor effect, which helps sensitize radioresistant cancer cells to radiotherapy [126]. D
Along with the drugs previously mentioned, ivermectin (IVM), an antiparasitic drug, has been considered a promising anticancer drug [133]. Ivermectin therapy has been shown to enhance autophagy in glioma cells by increasing the production of autophagosomes and activating autophagy-related genes and proteins [134,135,136]. Liu’s group reported the effect of ivermectin on cell growth and metabolism, as well as autophagy modulation [136]. Their group utilized U251 and C6 GBM cells to determine the impact that IVM had on autophagy. Western blot showed p-AKT (Ser473)/AKT and p-mTOR (S2448)/mTOR presented a dose-dependent reduction with the administration of IVM at 0, 5, 10, and 15 μM [136]. 

3.3. Drugs Decrease Cell Proliferation by Targeting AKT/mTOR Pathway

Autophagy inhibition in cancer cells can promote tumor development because genomic defects can accumulate within the cell and foster an environment for cancer cell growth [138]. As mentioned before, curcumin is a promising nutraceutical compound that provides effective treatment for GBM by downregulating the JAK/STAT3 pathway [95,102]. Surprisingly, curcumin is reported to target the AKT/mTOR pathway as well. An in vitro study with U87 human glioma cells treated with 5–10 μM curcumin showed that the cells remain arrested in the G2/M stages, subsequently inhibiting cell proliferation [102]. Additional studies on glioblastoma cell line U251 showed that when cells were treated with curcumin, phosphorylation levels of AKT and mTOR were significantly decreased [139]. This suggests that curcumin led to autophagy induction, and can lead to autophagy-mediated cell death in GBM cells by inhibiting the AKT/mTOR pathway [99].
Ganoderic acid DM (GA-DM) is a lanostanetriterpene that is isolated from the medicinal mushroom Ganoderma lucidum [141]. It shows potential antitumor activity in different cancers, including human glioblastoma [142]. Ganoderic acid DM induces G1 phase cell cycle arrest and activates p53 in the intrinsic apoptotic pathway [143]. Studies have shown that autophagy could be induced through GA-DM via the activation of the AMPK pathway followed by mTORC1 inhibition and the subsequent induction of autophagy [144,145]. This indicates that GA-DM could induce autophagy through modulating mTORC1.

4. Summary

Autophagy is a highly conserved cellular process that allows cells to adapt to a variety of conditions. Autophagy is a dynamic process and multiple networks can modulate and regulate this process. The pathways discussed were the JAK/STAT and PI3K/AKT/mTOR pathways. Autophagy has been known to play dual contrasting roles in cancer. In the initial phases of tumor formation, autophagy may have a tumor-suppressive role. Since autophagy removes damaged organelles and misfolded proteins within the cell, this minimizes the risk of genomic instability and potential tumor development. However, within established tumor cells, autophagy induction can be utilized as a survival mechanism. This allows for the established tumor cells to adapt to nutrient-stressed conditions and promotes resistance to therapeutics. However, when autophagy is overactivated and passes the specific threshold, autophagy-mediated cell death begins. Navigating the paradoxical role that autophagy plays in cancer cells has been one of the biggest challenges in developing effective therapies.

This entry is adapted from the peer-reviewed paper 10.3390/cimb45110546

This entry is offline, you can click here to edit this entry!
Video Production Service