Next Article in Journal
The Road Less Traveled: How COVID-19 Patients Use Metaphors to Frame Their Lived Experiences
Previous Article in Journal
Diet Quality, Health, and Wellbeing within the Irish Homeless Sector: A Qualitative Exploration
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

GRP78 Activity Moderation as a Therapeutic Treatment against Obesity

1
Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
2
Department of Molecular Biosciences and Bioengineering, University of Hawaii at Manoa, 1955 East-West Road, Honolulu, HI 96822, USA
*
Authors to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2022, 19(23), 15965; https://doi.org/10.3390/ijerph192315965
Submission received: 5 October 2022 / Revised: 18 November 2022 / Accepted: 26 November 2022 / Published: 30 November 2022

Abstract

:
Glucose-regulated protein 78 (GRP78), a molecular chaperone, is overexpressed in patients suffering from obesity, fatty liver, hyperlipidemia and diabetes. GRP78, therefore, can be not only a biomarker to predict the progression and prognosis of obesity and metabolic diseases but also a potential therapeutic target for anti-obesity treatment. In this paper, GRP78 inhibitors targeting its ATPase domain have been reviewed. Small molecules and proteins that directly bind GRP78 have been described. Putative mechanisms of GRP78 in regulating lipid metabolism were also summarized so as to investigate the role of GRP78 in obesity and other related diseases and provide a theoretical basis for the development and design of anti-obesity drugs targeting GRP78.

1. Introduction

Obesity is listed as a “first-class disease” with greater harm than infectious diseases in the World Health Organization and is mainly caused by energy intake and consumption imbalance, which can induce chronic metabolic diseases such as cancer, cardiovascular disease, diabetes, and hyperlipidemia [1]. The global annual treatment cost of obesity-related diseases is more than 10 billion U.S. dollars [2,3]. The research and development of anti-obesity drugs amount to several billion U.S. dollars [4]. At present, only two drugs (orlistat and sibutramine) have been approved by the U.S. Food and Drug Administration (FDA) for long-term treatment and to improve clinical obesity. Orlistat is the only over-the-counter (OTC) weight loss drug approved by China’s National Medical Products Administration. Orlistat is a hydrated derivative of lipstatin, which lowers body weight via inhibiting gastrointestinal lipase and reducing the absorption of food source fat. At the same time, sibutramine increases physiological hypersatiety and decreases appetite by inhibiting the reuptake of neurotransmitters such as serotonin, norepinephrine and dopamine. In addition, traditional Chinese medicines are commonly used, which include cassia seed, fleece-flower root, cattail pollen, hawthorn, rhubarb, gingko leaf, etc. The main active anti-obesity ingredients of traditional Chinese medicines were flavonoids and anthraquinone compounds [5], such as puerarin, curcumin, gallic acid, polyphenol, berberine, capsaicin, and emodin [6]. The Traditional Chinese Medicine Systems Pharmacology (TCMSP) database listed 25 anti-obesity targets, 467 herbs and 1592 relative ingredients (TCMSP database available online: http://tcmspw.com/tcmsp.php; accessed on 11 November 2022). However, due to the complicated causes and pathogenesis of obesity, there is no drug that can completely cure obesity at present.
What the cause of obesity is and how to balance energy intake and consumption at the cellular level have been unsolved scientific problems. The current hypotheses of obesity development mainly include the reactive oxygen species (ROS) theory, endoplasmic reticulum stress (ERS) hypothesis, energy balance hypothesis, microbial hypothesis, and thrifty gene hypothesis. For example, ERS can lead to mitophagy [7] and further lead to obesity [8]. Furthermore, the overload of energy intake can further aggravate the ERS activated by ROS after mitophagy.
The 78 kDa glucose-regulated protein (GRP78) is an important molecular chaperone, and its up-expression indicates ERS occurrence. Both clinical and animal studies have found that GRP78 overexpression is strongly correlated with obesity, and GRP78 is an essential component of ERS to balance mitochondrial biosynthesis and mitophagy, which is a potential therapeutic target for obesity [9]. However, the downstream mechanisms and pathogenesis of obesity mediated by GRP78, or traditional Chinese medicine, plays weight-loss and lipid-lowering roles via targeting GRP78 are still unclear.
A literature search with the keywords “GRP78” or “BiP/HSPA5” gave more than 20,000 publications in the Web of Science, PubMed, ACS, Elsevier and Wiley databases, but few using the keywords of “obesity” and “GRP78” or “BiP/HSPA5” (accessed on 15 September 2022). Most references cited are those found in Web of Science and PubMed, while some are in the Elsevier and Wiley databases between 2018 and March 2022. Early references are cited for providing the continuity and coverage of the information on the subject.
This paper focuses on the biological functions and roles of GRP78 in weight control and energy restriction via regulating mitochondrial autophagy and adenosine monophosphate-activated protein kinase-PPARγ coactivator 1α-sirtuin1 (AMPK-PGC1α-SIRT1) signal pathway (Figure 1, Pathway 1), revealing the subcellular mechanisms of GRP78 in the pathogenesis of obesity and providing new evidence for effective treatment of obesity and related diseases.

2. Structure, Function, and Subcellular Localization of GRP78

GRP78, also known as immunoglobulin heavy chain binding protein BiP or human heat shock protein 5 (HSPA5), is a member of the heat shock protein 70 family (HSP70) and consists of 654 amino acids. GRP78 structurally contains a C-terminal peptide-binding domain and an N-terminal ATP-binding domain. After ATP binds GRP78, the hydrophobic peptide-binding domain is exposed and binds the misfolded protein to form a complex. Subsequently, the energy released by ATP hydrolysis is utilized to promote protein rearrangement and correct folding [10], which mediates ERS, induces apoptosis and autophagy and maintains calcium homeostasis and internal environment stability [10,11,12]. A protein having a conserved terminal amino acid sequence of EEVD, KDEL/HDEL, PEAEYEEAKK, and PECDVLDAFTDSK can bind GRP78 and reside in the cytoplasm, endoplasmic reticulum, mitochondria, and plastid, respectively. A small amount of GRP78 is expressed in the cytoplasm under normal physiological conditions, while GRP78 is transferred into the nucleus under certain pathological conditions and returned to the cytoplasm after the endoplasmic reticulum homeostasis is restored. Under certain stress, GRP78 can be actively translocated to the surface of lipid droplets [13], mitochondria, cell membrane surface, cytoplasm, and nucleus, and can also be actively secreted to the extracellular microenvironment. GRP78 even can remain in the cytoplasm by disguising its KDEL signal.
GRP78 functions in multifaceted cellular activities in different subcellular compartments. Membrane-associated GRP78 is part of receptors for virus entry and initial infection of host cells, while ER GRP78 maintains ER homeostasis and helps newly and misfolded proteins to assemble, refold and transport. Nuclear GRP78 can inhibit apoptosis induced by DNA damage. Cytoplasmic GRP78 can not only inhibit ERS and apoptotic cascade by directly inactivating caspase-7 and blocking the release of caspase-12 to the cytoplasm but also regulates viral protein assembly and suppresses tumor growth through unfolded protein responses. Recent studies found that changes in the tumor microenvironment, such as glucose deficiency, protein misfolding, hypoxia, and protein glycosylated, could promote cell-surface expression of GRP78, serving as receptors, which can be used as biomarkers and therapeutic targets for cancer and other related diseases. A better understanding of GRP78 subcellular translocation involved in obesity pathogenesis will help to reveal the subcellular mechanism of ER stress-induced obesity, which may contribute to exploiting new effective controlling measures.

3. GRP78 Is Increased in Patients with Obesity and Is a Prognosis Marker

GRP78 regulates insulin resistance in diet-induced obesity, which is closely associated with obesity, type 2 diabetes and cardiovascular diseases. Serum level of GRP78 and mRNA expression in human subcutaneous and omental adipose tissues of obese patients is significantly higher than those of healthy groups, and GRP78 expression is positively correlated with body mass index (BMI) [14]. Therefore, it can be used as a biomarker to predict obesity, type 2 diabetes and cardiovascular diseases [14,15,16,17]. Another clinical study involved 405 patients (obese: 52.5%; Type 2 diabetes: 68.9%; metabolic syndrome: 78.6%) attending the diabetic clinic of the Komfo Anokye Teaching Hospital. The test data showed significantly higher content of serum GRP78 in obese, type 2 diabetes and metabolic syndrome patients as compared to healthy people. Moreover, serum GRP78 is positively correlated with the value of triglyceride fatty acid (TG), non-high-density lipoprotein (HDL)-cholesterol, low-density lipoprotein (LDL)-cholesterol, carotid intima-media thickness (CIMT), and atherosclerosis index in patients [14,16]. In addition, adipose Grp78-knockout mice showed that GRP78 plays an essential role in adipogenesis, lipogenesis and postnatal growth in mice, and inhibition of GRP78 expression can significantly reduce lipid content in mice [18].

4. Roles of GRP78 in Regulating Lipid Metabolism

4.1. GRP78 Promotes Adipogenesis and Lipogenesis

In a high-energy diet state, adipogenesis contributes to the increased adipose tissue mass of obesity. On the contrary, adipogenesis failure makes it unable to differentiate into enough mature adipocytes which can absorb, utilize, and store excess energy, thereby inducing diabetes, etc. [19]. Inhibition of adipogenesis can effectively prevent obesity. Peroxisome proliferator-activated receptor γ (PPARγ), a nuclear transcription factor, plays an important role in adipogenesis, and its molecular targets include CCAAT/enhancer-binding protein α (C/EBPα), perilipin (PLIN), hormone-sensitive lipase (HSL) and glucose transporter type 4 (GLUT4), which participate in adipogenesis, lipid metabolism and glucose metabolism, respectively [20,21] (Figure 1 Pathway 2 & 3). As compared with wild-type, PPARγ expression in Grp78 knockout mouse embryo fibroblasts is reduced [18]. GRP78 overexpression can promote the expression of PPARγ. Heat shock protein family (hsp70) member 12A (HSPA12A), a homolog of GRP78, is also one member of HSP70. Studies have found that HSPA12A regulates the transcription of PPARγ through a positive feedback loop, sustaining overexpression of PPARγ and maintaining the phenotype of adipocytes after differentiation (Figure 1 Pathway 4) [22].
Shown by drug affinity responsive target stability (DARTS) and surface plasmon resonance (SPR) results, epigallocatechin gallate (EGCG), dihydromyricetin (DHM) and berberine, the major active ingredient of flavonoids, induce white fat tissue browning but prevent adipogenesis or obesity via directly binding to GRP78 [23]. The dissociation constants (KD) of DHM and EGCG binding GRP78 were 22 µM and 6 µM, respectively, with significant anti-obesity activity with a maximum half effective concentration (EC50) of 400 µM and 75 µM, respectively [24].
GRP78 plays an important role in adipogenesis, lipogenesis, metabolic homeostasis, fetal and postnatal growth and development of mice [18]. Knockout Grp78 in mouse embryonic fibroblasts, 3T3-L1 cells and adipocyte tissue showed adipogenesis and lipogenesis problems. The aP2-cre-mediated GRP78 deletion leads to a reduction of lipoatrophy in gonadal and subcutaneous white adipose tissue and brown adipose tissue by up to ∼90%, severe growth retardation, bony defects and grossly expanded endoplasmic reticulum in white adipose tissue. However, plasma triglyceride levels and plasma glucose and insulin levels are reduced by 40–60% as compared to wild-type mice, suggesting an improvement of the insulin sensitivity in Grp78 knockout mice. The results indicated that GRP78 is essential for adipogenesis in vivo. Unexpectedly, the mutant mice showed early postnatal death and unique distinction from previously characterized lipodystrophic mouse models.
Sterol regulatory element binding protein-1c (SREBP-1c) is a transcription factor that critically regulates lipid metabolism. Insulin-induced cleavage and activation of SREBP-1c, which is the cause of ectopic fat deposition in the liver. SREBP-1c directly binds GRP78, which remains in the endoplasmic reticulum without transcription activity. Nevertheless, dissociation of SREBP-1c and GRP78 promotes the transport of the SREBPs-SCAP complex to Golgi, where SREBP-1c is cleaved, and then active SREBP-1c is transferred into the nucleus to initiate expression of genes involved in triglyceride and cholesterol synthesis (Figure 1 Pathway 5). Therefore, triglyceride and cholesterol levels are significantly increased [25,26,27]. Through hepatic overexpression of GRP78 in ob/ob mice using an adenovirus vector, it was found that GRP78 overexpression inhibits SREBP-1c cleavage and the expression of SREBP-1c target genes lowers liver triglycerides and cholesterol levels and improves insulin sensitivity [25].
Intermittent fasting can induce pregnancy zone protein (PZP) production and release in the liver, followed by the translocation of GRP78 to the cell surface of brown adipose tissue (BAT). The binding between PZP and GRP78 upregulates UCP1 expression through the p38 mitogen-activated protein kinases-activating the transcription factor-2 (p38 MAPK-ATF2) signaling pathway and promoting the thermogenic metabolism of BAT. These results indicated that GRP78 is an indispensable regulator of PZP-induced thermogenesis [28]. (Figure 1 Pathway 6).

4.2. GRP78 Promotes De Novo Formation of Lipid Droplets

Lipid droplets are the main cellular sites for triglycerides and other lipids storage [29]. Lipid droplet fusion and growth are closely regulated by lipid droplet-coated proteins adapted for cellular energy needs. Proteomic studies have found that HSP70 proteins, including GRP78, are major structural proteins of lipid droplets [30]. Expressed GRP78 is translocated to lipid droplets in rat adipocytes upon heat stimulation. This process occurs neither in a temperature-dependent nor time-dependent manner but occurs suddenly in 30–40 min and rapidly reaches a stable state within 1 h at 40 °C heat shock.
Although GRP78 is co-localized with phospholipids on the surface of lipid droplets, co-immunoprecipitation experiments did not show direct interactions between GRP78 and phospholipids. Alkaline treatment indicated an association of GRP78 with the surface of the droplets through non-hydrophobic interactions. Therefore, it is speculated that GRP78 may non-covalently associate with monolayer microdomains of lipid droplets in a manner like its interaction with lipid bilayer moieties composed of specific fatty acids. As an acute cell-specific response to heat stimulation, the accumulation of GRP78 on adipocytes lipid droplets may be involved in the stabilizing of droplet monolayer phospholipid, transferring or chaperoning denatured proteins to the lipid droplets for subsequent refolding.
Reticular protein 3 (RTN3) plays a key role in regulating triglyceride synthesis, storage, and lipid droplet fusion. Studies have found that RTN3 enhances SREBP-1C and AMPK activity through its interactions with GRP78 and leading to obesity and hyperlipidemia [31] (Figure 1 Pathway 7). SREBP-1c and AMPK are downstream of GRP78. Berbamine inhibits GRP78 and also induces AMPK activation, which regulates the mammalian target of the rapamycin/ SREBP-1c (mTOR/SREBP-1c) axis and the nuclear factor E2-related factor 2 (Nrf2)/antioxidant response element (Nrf2/ARE) pathway to allay lipid accumulation and oxidative stress in steatotic HepG2 cells. [32]

4.3. GRP78 Negatively Regulates Mitochondrial Biosynthesis and Energy Balance

A decrease in mitochondrial numbers and dysfunction can lead to obesity. GRP78 regulates mitophagy, mitochondrial biogenesis and energy balance [33,34]. Mitophagy is inhibited by GRP78 down expression, which is a vital way to activate browning and prevent obesity [35,36,37]. Further research found potential mitophagy mediation by GRP78 through the AMPK/mTOR signaling pathway, which leads to that energy intake exceeding expenditure and obesity. It was also found that GRP78 overexpression in the mitochondria triggers PINK1/IP3R-mediated neuroprotective mitophagy [38].

4.4. GRP78 Causes Insulin Resistance

Insulin resistance is considered an underlying etiology of metabolic syndrome and cardiovascular disease associated with obesity, such as type 2 diabetes. Both human and animal experiments have shown a positive correlation between GRP78 levels and insulin resistance [14,39] and effective improvement in insulin sensitivity and glycemic control via GRP78 down-regulation [15].
Further studies showed that GRP78 could activate the insulin signaling pathway and improve insulin sensitivity through phosphorylation modification of protein kinase B (Akt) [40]. GRP78 down-regulates Akt expression and phosphorylation but does not directly affect upstream pyruvate dehydrogenase kinase 1 (PDK1) activity. PDK1 is a protein kinase B (PKB), also known as Akt, which is a serine/threonine-specific protein kinase. Co-immunoprecipitation of GRP78 and p-Akt (Ser473) immune-complex consists of non-phosphorylated Akt (Ser473 and Thr308) (Figure 1 Pathway 8). In-situ proximity ligation analysis showed co-location of GRP78 with Akt in cell membrane after ER stress induction and an increase in phosphorylation of Akt Ser473 but a decrease (i.e., inhibition) of Thr308. siRNA-mediated GRP78 knockdown enhances phosphorylation at Ser473 by 3.6-fold, but no impact at Thr308 [40].
Human jejunal mucosa secretes GRP78 in vitro, and bariatric surgery improves insulin resistance and type 2 diabetes by reducing intestinal GRP78 secretion. Plasma GRP78 levels in insulin resistance patients are higher than in healthy people and those who returned to normal physiology after duodenal jejunal bypass surgery, and plasma GRP78 level was negatively correlated with insulin sensitivity but positively correlated with body mass index (BMI) [41].
A high-calorie diet can increase plasma GRP78 levels and induce insulin resistance. GRP78 stimulates the accumulation of lipid droplets and inhibits Akt Ser473 phosphorylation and glucose uptake in both immortal liver cells and peripheral blood plasma cells. However, a converse phenomenon occurs when GRP78 serum levels decrease or insulin-resistant patients undergo duodenal jejunal bypass surgery. Intestinal secretion of GRP78 may be the cause of insulin resistance, and duodenal jejunal bypass surgery may reduce GRP78 secretion and improve insulin sensitivity by shortening food transportation or reducing lipid stimulus released from endocrine cells [41]. GRP78 is essential for proinsulin synthesis, and up-regulation of GRP78 can increase insulin secretion in response to hyperglycemia, while down-regulation of GRP78 can decrease insulin secretion and lead to significantly low levels of insulin [42].

4.5. GRP78 Can Eliminate Liver Lipotoxicity and then Improve Liver Steatosis

ER stress plays an important role in hepatic steatosis and insulin resistance in obese mice models [43]. GPR78 is involved in the pathogenesis of nonalcoholic fatty liver disease and is associated with hepatic steatosis, insulin resistance, inflammation, and apoptosis [44,45]. GRP78 plays a key role in maintaining lipid balance in the liver, and GRP78 overexpression can reduce the hydrolysis of SREBP-1c induced by ER stress and liver steatosis [43]. It was also reported that GRP78 overexpression in HepG2 cells prevents ER stress and cytotoxicity induced by palmitic acid, and further studies found that GRP78 may reduce lipid peroxidation and damage induced by oxidative stress [46]. Some studies suggest that GRP78 overexpression in the liver can reduce the expression of SREBP-1c, reduce ectopic triglyceride deposition in the liver, and enhance insulin sensitivity of ob/ob mice [25].

5. Molecular Mechanism of Action of GRP78 during Obesity Development

5.1. Proteins and Small Molecules Directly Bind GRP78

New data on the therapeutic target GRP78 for obesity and its effectiveness are still rare. Ingenuity Pathway Analysis (IPA) analysis showed the relevance of 1216 proteins to obesity and the direct interactions of 139 of them with GRP78 (Figure 2). Studies such as co-immunoprecipitation (co-IP), fluorescence resonance energy transfer assay (FRET), GST pull-down technique, microscale thermophoresis (MST), and bioinformatics analysis have indicated direct interactions between GRP78 and downstream target proteins such as PPARγ, SREBP-1 and p-Akt [47] (Table 1). Research shows that binding of α2-macroglobulin-GRP78 triggers the expression of IP3, RAS and MAPK, PAk2, PI3K, PLD, COX2, and cPLA2 and also increases the intracellular second messengers (Ca2+ and cAMP) level and pH value, which can activate the downstream signal of PPARγ/C/EBPα, PLIN/HSL, etc. (Figure 1 and Figure 2). A secreted form and the cell surface localization of GRP78 bound to phosphoinositide 3-kinases (PI3K) then induced Akt activity to activate the PI3K/Akt/mTOR signaling pathway. GRP78 and Akt form a positive feedback loop [48], and continuous overexpression of mTOR can further enhance triglyceride synthesis and obesity (Figure 1) [49].
Nucleotide-binding domain (NBD) regulates the translocalization and secretion of GRP78. GRP78 translocation mechanism is related to its conserved terminal amino acid sequence, which is discussed in Section 2. Numerous studies using surface plasmon resonance (SPR) and GRP78 ATPase analysis have verified that EGCG and other compounds bind the nucleotide-binding domain of GRP78 (Figure 3). Five active ingredients (salvianolic acid A, naringin, platycodon, platycodon D, diosmin and isovitexin) were identified and extracted from 51 traditional Chinese medicines, which may directly interact with GRP78 [47,50]. EGCG, DHM, berberine and structural analogs have been discovered through drug screening for their inhibition against tumor-secreted GRP78 (Table 2 and Table 3). It is known that direct interactions between GRP78 and the 12 molecules are anti-Glioblastoma (GBM) (Table 2).
Figure 2. Ingenuity Pathway Analysis (IPA) result of GRP78 (HSPA5) with its interacting proteins relevant to obesity. Using “obesity” as the keyword, 1216 proteins, including HSPA5 (GRP78), were obtained from the IPA database. According to the analysis of the “Build-Path Explorer” module, 139 molecules have interactions with HSPA5.
Figure 2. Ingenuity Pathway Analysis (IPA) result of GRP78 (HSPA5) with its interacting proteins relevant to obesity. Using “obesity” as the keyword, 1216 proteins, including HSPA5 (GRP78), were obtained from the IPA database. According to the analysis of the “Build-Path Explorer” module, 139 molecules have interactions with HSPA5.
Ijerph 19 15965 g002
Table 1. List of proteins that directly bind GRP78 [47,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68].
Table 1. List of proteins that directly bind GRP78 [47,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68].
No.ProteinBinding Site and Interaction ModeDownstream Signaling/EffectMethods
1PERK/IRE1/ATF6Calnexin and vimentin interact indirectly with annexin A2 via GRP78Initially activate PERK-eIF2A, IRE1-XBP1 and ATF6 signal pathways and trigger cell apoptosisIP-MS;
Bioinformatics analysis
2PERKDisinhibited by release from GRP78, and then phosphorylates eIF2αActivated phosphorylation of eIF2α by PERK inhibits translation initiation, decreases protein synthesis and protein influx in the ER, then phosphorylation of ATF4 activates CHOP and apoptosis by PERK-eIF2α-ATF4-CHOP pathwayIHC
3IRE1Disinhibited by release from GRP78Splicing mRNA encoding XBP1 triggers endoribonuclease activity of IRE1, and then targets genes in protein folding and ERAD by IRE1-TRAF2-JNK pathwayIHC
4ATF6GRP78 directly activates ATF6 by binding to its luminal domain and inhibiting its Golgi localization signals; ATF6 is disinhibited by release from GRP78, then was cleaved and activated at Golgi apparatusActive ATF6 moves to nucleus and upregulates proteins that promotes ER protein folding;
ATF6 downstream target gene GRP78
IFA; IP.
5SREBP-1GRP78 retains SCAP/SREBP1 in the ER via direct interactionInhibit PI3K/Akt pathway; Positively regulates the transcription of ACC and FASN; SREBP-1 downstream targets involved in fatty acid synthesis, including FASNCo-IP
6CNPY2CNPY2 combines with GRP78 under normal conditions; After UPR inducer chlamycin treatment, CNPY2 disassociates from GRP78 and binds to PERK Activate PERK-CHOP signaling; CNPY2 blocked the PERK-CHOP pathway of the unfolded protein responseCnpy2 knockout mice fed a high-fat diet
7p-AktExocrine GRP78Activate PI3K/AKT signaling, then promote tumor cell proliferation and decrease the sensitivity of HCC cells to sorafenibCo-IP
8SR-ASR-A binds to cytoplasmic GRP78GRP78 involved in SR-A mediates lipid endocytosis; Inhibits inflammatory cytokine expression (TNF-α, IL-1) through MAPK, PI3K-Akt and NF-κB signalingIP;
Indirect IFA;
FRET
9SCNN1BSCNN1B interacts with GRP78 and induces GRP78 degradation via polyubiquitination.Increase ubiquitin-mediated degradation of GRP78, subsequently trigger the unfolded protein response (UPR)Tissue microarray analysis;
SCNN1B ectopic expression and knockdown;
IFA;
IP–MS.
10Anti-EGFR antibodyAnti-EGFR antibody combines and co-locates with GRP78Block the promotion of GRP78 to the invasion of cancer stem cellsTranswell;
Confocal microscopy;
WB
IP: Immunoprecipitation; MS: mass spectrometry; SR-A: Class A scavenger receptor; IHC: Immunohistochemistry; IFA: Immunofluorescence; FRET: Fluorescence resonance energy transfer; WB: Western blot.
Figure 3. GRP78 inhibitors targeting its ATPase/nucleotide-binding domain.
Figure 3. GRP78 inhibitors targeting its ATPase/nucleotide-binding domain.
Ijerph 19 15965 g003
Table 2. GRP78 inhibitors and inducers and their action mechanisms and effects [51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68].
Table 2. GRP78 inhibitors and inducers and their action mechanisms and effects [51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68].
No.Compound/Combination Therapy in GBMAction MechanismEffectsModel
1EGCG;
EGCG+TMZ/5-fluorouracil/taxol/vinblastine/gemcitabine/TRAIL/doxorubicin/paclitaxel/IFN-α2b
GRP78 (NBD)Impair GRP78 function;
Enhance cytotoxicity when used with TMZ or others
Human cell lines, in vivo [51,52]
2Honokiol; Honokiol+TMZ/fenretinide/bortezomibGRP78 (NBD)Interfere with GRP78 folding; Induce ER stress-mediated apoptosis with TMZHuman cell lines [53,54,55]
5NEO100 (Clinical trials);
NEO100+TMZ/DMC/relfinavir
ER stressDisrupt survival pathways; Induce more apoptosis with TMZ and others, reduce GBM invasion capacity, prolong survivalHuman cell lines, in vivo [58,59,60]
8EGF-SubA;
EGF-SubA+radiation+TMZ
Cleave GRP78Correct the ATPase and protein binding domains; Delay tumor growth, enhance effects of TMZ and ionizing radiationHuman cell lines in vivo mouse models [64]
3OSU-03012;
radiotherapy+OSU-03012;
GRP78 (NBD)PDK1 inhibition, GRP78 inhibition, PERK signaling inhibition;
Enhance radiosensitivity; Prolong survival
Human cell lines, in vivo mouse models [52,56]
4Celecoxib and bortezomib Celecoxib+bortezomib+GRP78 inhibitionER stressAugment ER stress; Induce ER stress-mediated apoptosisHuman cell lines [57]
6HA15Bind and inhibit GRP78 Disrupt GRP78 complexes with PERK/IRE1/ATF6; Induce apoptosisHuman cell lines, in vivo mouse models [61,62]
7IT-139GRP78Involve transcriptional and post-transcriptional mechanisms; Decrease therapeutic resistanceHuman cell lines, in vivo human xenograft studies [63]
8EGF-SubA TMZ+radiation therapy+EGF-SubA [64]GRP78; Cleave GRP78TMZ and ionizing radiation; Delay tumor growth, enhanc effects of TMZ and ionizing radiationHuman cell lines in vivo mouse models
9Anti-GRP78 antibody;
Ionizing radiation+anti-GRP78 antibody
Bind to surface GRP78Enhance effects of ionizing radiation via suppression of PI3K/AKT/mTOR signaling, and result in tumor delayHuman cell lines, in vivo mouse xenograft models [65]
10RGD ligand-directed phage with GRP78 promoterBind GRP78RGD tumor homing ligand binds and improves expression of therapeutic transgenes with GRP78 promoterHuman cell lines in vivo [66]
11TMZ-induced AAV phage with GRP78 promoter;
TMZ+phage
RGD4C/AAV/phage/GRP78 bindingActivate therapeutic transgenes expression with GRP78 promoter; Permit dose escalation of TMZHuman cell lines, mouse xenograft models [67]
12GIRLPG;
Radiation+phage
Bind GRP78Allow for adenovirus-mediated gene delivery to target tumor cells; Enhance radiation therapy and therapeutic transgenes expressionHuman cell lines, mouse xenograft models [68]
GBM: Glioblastoma; NBD: Nucleotide-binding domain; TMZ: Temozolomide, which is the leading compound in GBM treatment; TRAIL: Tumor necrosis factor (TNF)-related apoptosis-inducing ligand; IFN-α2b: interferon-α2b; NEO100: Perillyl alcohol (monoterpene); EGF-SubA: Epidermal growth factor; OSU-03012: Celecoxib derivative; GIRLPG: GRP78 binding peptide.
Table 3. GRP78ATPase nucleotide affinities (KD) determined by surface plasmon resonance (SPR) [6,23,47,50,52,53,54,55,56,69,70,71,72,73,74,75,76,77,78].
Table 3. GRP78ATPase nucleotide affinities (KD) determined by surface plasmon resonance (SPR) [6,23,47,50,52,53,54,55,56,69,70,71,72,73,74,75,76,77,78].
No.Chemicals2 mM MgCl2KD (M) a5 mM EDTABiologic Activity/Reference
Untreated b
1ATP(4.5 ± 2.9) × 10−7(7.8 ± 7.1) × 10−7(9.8 ± 4.4) × 10−6[69]
2ADP(1.2 ± 0.9) × 10−8(2.7 ± 4.4) × 10−7(4.3 ± 7.7) × 10−5[69]
37-deazaATP(3.0 ± 2.0) × 10−8(1.5 ± 0.9) × 10−7(9.0 ± 5.4) × 10−7[69]
4AMPPCP(5.9 ± 1.2) × 10−5(5.2 ± 4.1) × 10−5>1 × 10−3[69]
52′-deoxyATP (7.5 ± 5.0) × 10−4>1 × 10−3[69]
6Honokiol Bind GRP78 (NBD) using DSC and ITC [53,54,55,70]
7Mangiferin [71]
8Isoliquiritigenin [72]
9OSU-03012 [56,72,73]
12Luteolin [74]
13DHM 22 × 10−6 Anti-adipogenesis, EC50 284 μM [24]
14HA15 Induce UPR and kill BRAF mutant melanomas [75]
15EGCG 6 × 10−6 bind GRP78 (NBD); Anti-adipogenesis, EC50 103 μM [52,72,73,76]
16Salvianolic acid A Lysine 633 acetylation of GRP78 to block GRP78 secretion [72,77]
17Salvianolic acid B [74]
18Salidroside [74]
19Salubrinal [74]
20Echinacoside [74]
21Betulinic acid [72]
22Capsaicin [6]
23Berberine [6,23,47,50]
24Naringin Inhibit the expression of GRP78 [77]
25Platycodon platycodon D Up-regulate expression of GRP78 [77]
26Diosmin [77]
27Isovitexin [77]
28Emodin [6]
29Curcumin DARTs, directly targeting GRP78 [6]
30Novolactone Destabilize HER2 and EGFR in cancer cells [75]
31Rifampicin [74]
32Puerarin [6]
33Hexachlorphene Induce apoptosis and block autophagy in melanoma cell lines [78]
34VER-155008 80 × 10−9 [72]
a KD values are presented as mean ± SD of three independent experiments (n = 3); b Untreated denotes that SPR running buffer contains no addition of either MgCl2 or EDTA. DSC: Differential scanning calorimetry; ITC: Isothermal titration calorimetry analysis; DHM: Dihydromyricetin; EGCG: Epigallocatechin gallate.

5.2. GRP78 Monomer/Heteromer and Conformational Changes

Over the past few decades, research attention has been paid to the targeted therapy, transcriptional activity, unfolded proteins response, ATP binding and hydrolysis activities of GRP78 (Figure 4) [72]. Subsequently, a great deal of scientific research has been devoted to clarifying the functions of GRP78 in both in vitro and in vivo. GRP78 was previously thought to exist as a monomer in eukaryotic cells and spontaneously dimerize/oligomerize to function. However, recent studies showed that GRP78 exists as a dynamic pool of monomer, dimer and oligomer, and dimer/oligomer transition from active monomer resulting in inactive GRP78 form [10]. EGCG, DHM and HA15 exert anti-obesity activity by inducing GRP78 conformational change [50]. Since ER stress-regulated by GRP78 is essential for virtually all cellular activities, either intracellular signaling or transcriptional regulation, and its association with several pathologies. Therapeutics targeting GRP78 via reducing its expression might have side effects. The potential of GRP78-targeted anti-tumor drugs has no effect on intracellular GRP78 expression but blocks its secretion. GRP78 conformation transition only works under specific pathological conditions, and allosteric modulation of GRP78 has better specificity. However, the mediation of GRP78 conformational transitions remains a gap, and the mechanism still is not clear.
Allosteric modulators are appealing sources of drug discovery for sensitivity, selectivity and security, which can be used in an ultra-low dose and do not fully close substrate channeling. Nevertheless, traditional regulators need higher doses to compete with substrates for the active site. However, GRP78 allosteric modulators still face challenges, such as no allosteric prediction models, lack of knowledge of the quaternary structure-activity relationship, low affinity, unknown certain binding sites, intracellular/extracellular distribution regulatory, incorporation polar and Log P median solubilization to enhance bioactivity.
Nevertheless, the great advantages of high selectivity and safety have raised a growing interest in the allosteric regulatory molecules of GRP78 in recent years. Although binding sites and crystal structures of allosteric regulatory molecules of GRP78 have been reported, there is a need for drug development and structural biology research. In addition to traditional experimental methods, computational biology and bioinformatics methods are powerful tools for the development of allosteric modulators targeting GRP78.
It is of great significance to explore the relationship between biological phase separation and disorder proteins in human diseases. Protein phase separation disorder can lead to protein transition from reversible liquid-liquid phase to irreversible liquid-solid phase and form irreversible amyloid-like aggregations.
It was first found that the chaperone GRP78 regulates liquid-liquid phase separation (LLPS) and liquid-solid phase transition through its own phase separation, which can maintain the LLPS homeostasis of transactive response DNA binding protein 43 (TDP-43) and inhibit spontaneous TDP-43 aggregation into amyloid fibrils in neurons under stress. Lipid droplet biogenesis is also driven by LLPS [79,80]. Phase separation is a basic organization in the cell membrane, and allosteric regulators may play a role by interfering with chaperones’ phase separation [79,80]. Although GRP78 is constitutively expressed, its synthesis can be induced by a variety of stresses, such as neurotoxicity, myocardial infarction, and arteriosclerosis. Its role is double-faceted, which cannot be clearly explained from the genome and transcription level. Protein post-translational modifications of GRP78 include phosphorylation, glycosylation, ubiquitination, allosteric modulation and acetylation [73], which can dynamically control the downstream proteins undergoing phase separation in multi-level. It warrants an exciting direction for future research.

6. Conclusions

The roles of GRP78 on obesity are clear, including promoting adipogenesis and lipogenesis, promoting de novo formation of lipid droplets, negatively regulating mitochondrial biosynthesis and energy balance, causing insulin resistance, eliminating liver lipotoxicity and then improving liver steatosis. While GRP78 shows regulatory functions against obesity, drug engagement needs further research. The majority of these natural ingredient studies have not yet been tested in clinical trials. Current in vitro and in vivo animal studies show promising anti-obesity effects. Molecules like ingredients of herbs described in this review may provide clues to future studies. Allosteric modulators with high efficacy at an ultralow dose and LLPS functions of GRP78 are probably ways to alleviate obesity.

Author Contributions

Conceptualization, D.P. and Q.X.L.; Literature search and first draft, D.P.; Funding acquisition and project administration, D.P., Z.T. and Q.X.L.; Writing—review and editing, D.P., A.N., Y.Y., Z.T. and Q.X.L. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the University-level Project of Guangxi University of Traditional Chinese Medicine (2018BS041, 2018ZD005-A04, 04B2101704 and 2021DXS43), the Research Basic Ability Improvement Project of Young and Middle-aged Teachers in Guangxi Universities (2020KY07043), and the USDA (HAW05020H).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data supporting the conclusion of this article will be available upon request from the corresponding authors.

Acknowledgments

Thanks to IPA analysis conducted by Ming Lv at Tianjin University of Chinese Medicine, and also thanks to Donghua Tang of The Second Affiliated Hospital, Guangxi Medical University for her contribution to this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

AMPK: adenosine monophosphate (AMP) activated protein kinase; Akt, an ubiquitous serine/threonine kinase, also known as protein kinase B (PKB) or RAS-alpha; ATF2, activating transcription factor-2; BAT, brown adipose tissue; BMI, body mass index; C/EBPα, CCAAT/enhancer-binding protein α (which CCAAT is a distinct pattern of nucleotides with GGCCAATCT consensus sequence that occurs upstream by 60–100 bases to the initial transcription site); CIMT, carotid intima media thickness; co-IP, co-immunoprecipitation; DARTS, drug affinity responsive target stability; EC50, half maximal effective concentration; EGCG, epigallocatechin gallate; ERS, endoplasmic reticulum stress; FRET, fluorescence resonance energy transfer assay; GLUT4, glucose transporter type 4; GRP78, 78 kDa glucose-regulated protein; HSPA5, human heat shock protein 5; HSP70, heat shock protein 70 family; HSL, hormone sensitive lipase; HSPA12A, heat shock protein family a (hsp70) member 12A; IR, insulin resistance; KD, dissociation constants; MST, microscale thermophoresis; mTOR, mammalian target of rapamycin; Nrf2/ARE, nuclear factor E2-related factor 2 (Nrf2)/ antioxidant response element; PGC1α, PPARγ coactivator 1α; PDK1, pyruvate dehydrogenase kinase 1; PI3K, phosphoinositide 3-kinases; PPARγ, peroxisome proliferator activated receptor γ; PZP, pregnancy zone protein, a novel hepatokine; PLIN, perilipin; p38 MAPK, p38 mitogen-activated protein kinases; p-Akt, phosphorylated Akt; ROS, reactive oxygen species; RTN3, reticular protein 3; SIRT1, sirtuin1; SPR, surface plasmon resonance; SREBP-1c, sterol regulatory element binding protein-1c; SREBPs, sterol regulatory element binding protein; SCAP, SREBP cleavage-activating protein; TG, triglyceride fatty acid; UCP1, uncoupling protein 1.

References

  1. WHO. About Diabetes; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  2. Available online: https://www.worldobesity.org/news/world-obesity-day-all-countries-significantly-off-track-to-meet-2025-who-targets-on-obesity (accessed on 30 September 2022).
  3. Available online: http://test.health-china.com/c/2021-09-22/803749.shtml (accessed on 30 March 2022).
  4. Available online: https://www.fortunebusinessinsights.com/anti-obesity-drugs-market-104783 (accessed on 30 September 2022).
  5. Martínez-Rodríguez, O.P.; Thompson-Bonilla, M.D.R.; Jaramillo-Flores, M.E. Association between obesity and breast cancer: Molecular bases and the effect of flavonoids in signaling pathways. Crit. Rev. Food Sci. Nutr. 2020, 60, 3770–3792. [Google Scholar] [CrossRef]
  6. Garza, A.; Milagro, F.I.; Boque, N.; Campión, J.; Martínez, J.A. Natural inhibitors of pancreatic lipase as new players in obesity treatment. Planta Med. 2011, 77, 773–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Marycz, K.; Kornicka, K.; Szlapka-Kosarzewska, J.; Weiss, C. Excessive endoplasmic reticulum stress correlates with impaired mitochondrial dynamics, mitophagy and apoptosis, in liver and adipose tissue, but not in muscles in ems horses. Int. J. Mol. Sci. 2018, 19, 165. [Google Scholar] [CrossRef] [Green Version]
  8. Deng, X.; Ye, Z.; Cao, H.; Bai, Y.; Che, Q.; Guo, J.; Su, Z. Chitosan oligosaccharide ameliorated obesity by reducing endoplasmic reticulum stress in diet-induced obese rats. Food Funct. 2020, 11, 6285–6296. [Google Scholar] [CrossRef]
  9. Contreras, C.; Fondevila, M.F.; López, M. Hypothalamic GRP78, a new target against obesity? Adipocyte 2018, 7, 63–66. [Google Scholar] [CrossRef] [PubMed]
  10. Takakuwa, J.; Nitika Knighton, L.; Truman, A. Oligomerization of Hsp70: Current perspectives on regulation and function. Front. Mol. Biosci. 2019, 6, 81. [Google Scholar] [CrossRef] [Green Version]
  11. Wieteska, L.; Shahidi, S.; Zhuravleva, A. Allosteric fine-tuning of the conformational equilibrium poises the chaperone BiP for post-translational regulation. eLife 2017, 6, e29430. [Google Scholar] [CrossRef] [PubMed]
  12. Kopp, M.C.; Larburu, N.; Durairaj, V.; Adams, C.J.; Ali, M.M.U. UPR proteins IRE1 and PERK switch BiP from chaperone to ER stress sensor. Nat. Struct. Mol. Biol. 2019, 26, 1053–1062. [Google Scholar] [CrossRef]
  13. Jiang, H.; He, J.; Pu, S.; Tang, C.; Xu, G. Heat shock protein 70 is translocated to lipid droplets in rat adipocytes upon heat stimulation. Biochim. Biophys. Acta 2007, 1771, 66–74. [Google Scholar] [CrossRef]
  14. Girona, J.; Rodríguez-Borjabad, C.; Ibarretxe, D.; Vallvé, J.C.; Ferré, R.; Heras, M.; Rodríguez-Calvo, R.; Guaita-Esteruelas, S.; Martínez-Micaelo, N.; Plana, N.; et al. The circulating GRP78/BiP is a marker of metabolic diseases and atherosclerosis: Bringing endoplasmic reticulum stress into the clinical scenario. J. Clin. Med. 2019, 8, 1793. [Google Scholar] [CrossRef]
  15. Khadir, A.; Kavalakatt, S.; Abubaker, J.; Cherian, P.; Madhu, D.; Al-Khairi, I.; Abu-Farha, M.; Warsame, S.; Elkum, N.; Dehbi, M.; et al. Physical exercise alleviates ER stress in obese humans through reduction in the expression and release of GRP78 chaperone. Metabolism 2016, 65, 1409–1420. [Google Scholar] [CrossRef] [PubMed]
  16. Girona, J.; Rodríguez-Borjabad, C.; Ibarretxe, D.; Vallvé, J.C.; Ferré, R.; Heras, M.; Rodríguez-Calvo, R.; Guaita-Esteruelas, S.; Martínez-Micaelo, N.; Plana, N.; et al. Circulating Grp78/Bip is increased in patients with obesity and related metabolic disorders and is associated with atherosclerosis. Atherosclerosis 2019, 287, e136. [Google Scholar] [CrossRef]
  17. Nourbakhsh, M.; Sharifi, R.; Heydari, N.; Nourbakhsh, M.; Ezzati-Mobasser, S.; Zarrinnahad, H. Circulating TRB3 and GRP78 levels in type 2 diabetes patients: Crosstalk between glucose homeostasis and endoplasmic reticulum stress. J. Endocrinol. Investig. 2022, 45, 649–655. [Google Scholar] [CrossRef]
  18. Zhu, G.; Ye, R.; Jung, D.Y.; Barron, E.; Friedline, R.H.; Benoit, V.M.; Hinton, D.R.; Kim, J.K.; Lee, A.S. GRP78 plays an essential role in adipogenesis and postnatal growth in mice. FASEB J. 2013, 27, 955–964. [Google Scholar] [CrossRef] [Green Version]
  19. Camacho, A.; Segoviano-Ramírez, J.C.; Sánchez-Garcia, A.; de Jesus Herrera-de la Rosa, J.; García-Juarez, J.; Hernandez-Puente, C.A.; Calvo-Anguiano, G.; Maltos-Uro, S.R.; Olguin, A.; Gojon-Romanillos, G.; et al. Tyrphostin AG17 inhibits adipocyte differentiation in vivo and in vitro. Lipids Health Dis. 2018, 17, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Perera, R.J.; Marcusson, E.G.; Koo, S.; Kang, X.; Kim, Y.; White, N.; Dean, N.M. Identification of novel PPARgamma target genes in primary human adipocytes. Gene 2006, 369, 90–99. [Google Scholar] [CrossRef]
  21. Nakachi, Y.; Yagi, K.; Nikaido, I.; Bono, H.; Tonouchi, M.; Schönbach, C.; Okazaki, Y. Identification of novel PPARgamma target genes by integrated analysis of ChIP-on-chip and microarray expression data during adipocyte differentiation. Biochem. Biophys. Res. Commun. 2008, 372, 362–366. [Google Scholar] [CrossRef]
  22. Zhang, X.; Chen, X.; Qi, T.; Kong, Q.; Cheng, H.; Cao, X.; Li, Y.; Li, C.; Liu, L.; Ding, Z. HSPA12A is required for adipocyte differentiation and diet-induced obesity through a positive feedback regulation with PPARγ. Cell Death Differ. 2019, 26, 2253–2267. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, L.; Zhou, M.; Lang, H.; Zhou, Y.; Mi, M. Dihydromyricetin enhances glucose uptake by inhibition of MEK/ERK pathway and consequent down-regulation of phosphorylation of PPARγ in 3T3-L1 cells. J. Cell. Mol. Med. 2018, 22, 1247–1256. [Google Scholar] [CrossRef] [Green Version]
  24. Sun, B.; Tan, D.; Pan, D.; Baker, M.R.; Liang, Z.; Wang, Z.; Lei, J.; Liu, S.; Hu, C.Y.; Li, Q.X. Dihydromyricetin imbues antiadipogenic effects on 3T3-L1 cells via direct interactions with 78-kDa glucose-regulated protein. J. Nutr. 2021, 151, 1717–1725. [Google Scholar] [CrossRef]
  25. Kammoun, H.L.; Chabanon, H.; Hainault, I.; Luquet, S.; Magnan, C.; Koike, T.; Ferré, P.; Foufelle, F. GRP78 expression inhibits insulin and ER stress-induced SREBP-1c activation and reduces hepatic steatosis in mice. J. Clin. Investig. 2009, 119, 1201–1215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Colgan, S.M.; Hashimi, A.A.; Austin, R.C. Endoplasmic reticulum stress and lipid dysregulation. Expert Rev. Mol. Med. 2011, 13, e4. [Google Scholar] [CrossRef]
  27. Lhoták, S.; Sood, S.; Brimble, E.; Carlisle, R.E.; Colgan, S.M.; Mazzetti, A.; Dickhout, J.G.; Ingram, A.J.; Austin, R.C. ER stress contributes to renal proximal tubule injury by increasing SREBP-2-mediated lipid accumulation and apoptotic cell death. Am. J. Physiol. Ren. Physiol. 2012, 303, F266–F278. [Google Scholar] [CrossRef] [Green Version]
  28. Lin, J.; Jiang, X.; Dong, M.; Liu, X.; Shen, Q.; Huang, Y.; Zhang, H.; Ye, R.; Zhou, H.; Yan, C.; et al. Hepatokine pregnancy zone protein governs the diet-induced thermogenesis through activating brown adipose tissue. Adv. Sci. 2021, 8, e2101991. [Google Scholar] [CrossRef]
  29. Smolič, T.; Zorec, R.; Vardjan, N. Pathophysiology of Lipid Droplets in Neuroglia. Antioxidants 2021, 11, 22. [Google Scholar] [CrossRef]
  30. Wang, W.; Wei, S.; Li, L.; Su, X.; Du, C.; Li, F.; Geng, B.; Liu, P.; Xu, G. Proteomic analysis of murine testes lipid droplets. Sci. Rep. 2015, 5, 12070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Xiang, R.; Fan, L.L.; Huang, H.; Chen, Y.Q.; He, W.; Guo, S.; Li, J.J.; Jin, J.Y.; Du, R.; Yan, R.; et al. Increased reticulon 3 (RTN3) leads to obesity and hypertriglyceridemia by interacting with heat shock protein family A (Hsp70) member 5 (HSPA5). Circulation 2018, 138, 1828–1838. [Google Scholar] [CrossRef]
  32. Sharma, A.; Anand, S.K.; Singh, N.; Dwivedi, U.N.; Kakkar, P. Berbamine induced AMPK activation regulates mTOR/SREBP-1c axis and Nrf2/ARE pathway to allay lipid accumulation and oxidative stress in steatotic HepG2 cells. Eur. J. Pharmacol. 2020, 882, 173244. [Google Scholar] [CrossRef]
  33. Prasad, M.; Pawlak, K.J.; Burak, W.E.; Perry, E.E.; Marshall, B.; Whittal, R.M.; Bose, H.S. Mitochondrial metabolic regulation by GRP78. Sci. Adv. 2017, 3, e1602038. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, X.; Li, X.; Fang, H.; Guo, F.; Li, F.; Chen, A.; Huang, S. Flavonoids as inducers of white adipose tissue browning and thermogenesis: Signalling pathways and molecular triggers. Nutr. Metab. 2019, 16, 47. [Google Scholar] [CrossRef]
  35. Zhang, Y.; Sowers, J.R.; Ren, J. Targeting autophagy in obesity: From pathophysiology to management. Nat. Rev. Endocrinol. 2018, 14, 356–376. [Google Scholar] [CrossRef] [PubMed]
  36. Yan, Q.; Li, X. Relationship between mitophagy and browning of white adipose. Int. J. Endocrinol. Metab. 2019, 39, 87–90. [Google Scholar]
  37. Alan, P.; Vandevoorde, K.R.; Joshi, B.; Cardoen, B.; Gao, G.; Mohammadzadeh, Y.; Hamarneh, G.; Nabi, I.R. Gp78-mediated basal mitophagy promotes mitochondrial health and limits mitochondrial ROS production. bioRxiv 2021. [Google Scholar] [CrossRef]
  38. Leiva-Rodríguez, T.; Romeo-Guitart, D.; Herrando-Grabulosa, M.; Muñoz-Guardiola, P.; Polo, M.; Bañuls, C.; Petegnief, V.; Bosch, A.; Lizcano, J.M.; Apostolova, N.; et al. GRP78 overexpression triggers PINK1-IP3R-mediated neuroprotective mitophagy. Biomedicines 2021, 9, 1039. [Google Scholar] [CrossRef]
  39. Kim, J.H.; Lee, E.; Friedline, R.H.; Suk, S.; Jung, D.Y.; Dagdeviren, S.; Hu, X.; Inashima, K.; Noh, H.L.; Kwon, J.Y.; et al. Endoplasmic reticulum chaperone GRP78 regulates macrophage function and insulin resistance in diet-induced obesity. FASEB J. 2018, 32, 2292–2304. [Google Scholar] [CrossRef] [PubMed]
  40. Yung, H.W.; Charnock-Jones, D.S.; Burton, G.J. Regulation of AKT phosphorylation at Ser473 and Thr308 by endoplasmic reticulum stress modulates substrate specificity in a severity dependent manner. PLoS ONE 2011, 6, e17894. [Google Scholar] [CrossRef] [PubMed]
  41. Angelini, G.; Salinari, S.; Bertuzzi, A.; Iaconelli, A.; Mingrone, G. Metabolic surgery improves insulin resistance through the reduction of gut-secreted heat shock proteins. Commun. Biol. 2018, 1, 69. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Zhang, L.; Lai, E.; Teodoro, T.; Volchuk, A. GRP78, but not protein-disulfide isomerase, partially reverses hyperglycemia-induced inhibition of insulin synthesis and secretion in pancreatic β-Cells. J. Biol. Chem. 2009, 284, 5289–5298. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Chen, W.T.; Zhu, G.; Pfaffenbach, K.; Kanel, G.; Stiles, B.; Lee, A.S. GRP78 as a regulator of liver steatosis and cancer progression mediated by loss of the tumor suppressor PTEN. Oncogene 2014, 33, 4997–5005. [Google Scholar] [CrossRef] [Green Version]
  44. Gentile, C.L.; Frye, M.; Pagliassotti, M.J. Endoplasmic reticulum stress and the unfolded protein response in nonalcoholic fatty liver disease. Antioxid. Redox Signal. 2011, 15, 505–521. [Google Scholar] [CrossRef] [Green Version]
  45. Chalasani, N.; Szabo, G. Alcoholic and Non Alcoholic Fatty Liver Disease Pathogenesis of NAFLD and NASH; Springer: Cham, Switzerland, 2016. [Google Scholar] [CrossRef]
  46. Teodoro-Morrison, T.; Schuiki, I.; Zhang, L.; Belsham, D.D.; Volchuk, A. GRP78 overproduction in pancreatic beta cells protects against high-fat-diet-induced diabetes in mice. Diabetologia 2013, 56, 1057–1067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Liu, K.; Tsung, K.; Attenello, F.J. Characterizing cell stress and GRP78 in glioma to enhance tumor treatment. Front. Oncol. 2020, 10, 608911. [Google Scholar] [CrossRef]
  48. Gray, M.J.; Mhawech-Fauceglia, P.; Yoo, E.; Yang, W.; Wu, E.; Lee, A.S.; Lin, Y.G. AKT inhibition mitigates GRP78 (glucose-regulated protein) expression and contribution to chemoresistance in endometrial cancers. Int. J. Cancer 2013, 133, 21–30. [Google Scholar] [CrossRef] [Green Version]
  49. Han, J.; Li, E.; Chen, L.; Zhang, Y.; Wei, F.; Liu, J.; Deng, H.; Wang, Y. The CREB coactivator CRTC2 controls hepatic lipid metabolism by regulating SREBP1. Nature 2015, 524, 243–246. [Google Scholar] [CrossRef]
  50. Qiao, Y.; Dsouza, C.; Matthews, A.A.; Jin, Y.; He, W.; Bao, J.; Jiang, F.; Chandna, R.; Ge, R.; Fu, L. Discovery of small molecules targeting GRP78 for antiangiogenic and anticancer therapy. Eur. J. Med. Chem. 2020, 193, 112228. [Google Scholar] [CrossRef] [PubMed]
  51. Chen, T.C.; Wang, W.; Golden, E.B.; Thomas, S.; Sivakumar, W.; Hofman, F.M.; Louie, S.G.; Schönthal, A.H. Green tea epigallocatechin gallate enhances therapeutic efficacy of temozolomide in orthotopic mouse glioblastoma models. Cancer Lett. 2011, 302, 100–108. [Google Scholar] [CrossRef]
  52. Gurusinghe, K.R.D.S.N.S.; Mishra, A.; Mishra, S. Glucose-regulated protein 78 substrate-binding domain alters its conformation upon EGCG inhibitor binding to nucleotide-binding domain: Molecular dynamics studies. Sci. Rep. 2018, 8, 5487. [Google Scholar] [CrossRef] [Green Version]
  53. Martin, S.; Lamb, H.K.; Brady, C.; Lefkove, B.; Bonner, M.Y.; Thompson, P.; Lovat, P.E.; Arbiser, J.L.; Hawkins, A.R.; Redfern, C.P. Inducing apoptosis of cancer cells using small-molecule plant compounds that bind to GRP78. Br. J. Cancer 2013, 109, 433–443. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Chio, C.C.; Chen, K.Y.; Chang, C.K.; Chuang, J.Y.; Liu, C.C.; Liu, S.H.; Chen, R.M. Improved effects of honokiol on temozolomide-induced autophagy and apoptosis of drug-sensitive and -tolerant glioma cells. BMC Cancer 2018, 18, 379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Chio, C.C.; Tai, Y.T.; Mohanraj, M.; Liu, S.H.; Yang, S.T.; Chen, R.M. Honokiol enhances temozolomide-induced apoptotic insults to malignant glioma cells via an intrinsic mitochondrion-dependent pathway. Phytomedicine 2018, 49, 41–51. [Google Scholar] [CrossRef]
  56. Booth, L.; Cazanave, S.C.; Hamed, H.A.; Yacoub, A.; Ogretmen, B.; Chen, C.S.; Grant, S.; Dent, P. OSU-03012 suppresses GRP78/BiP expression that causes PERK-dependent increases in tumor cell killing. Cancer Biol. Ther. 2012, 13, 224–236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kardosh, A.; Golden, E.B.; Pyrko, P.; Uddin, J.; Hofman, F.M.; Chen, T.C.; Louie, S.G.; Petasis, N.A.; Schönthal, A.H. Aggravated endoplasmic reticulum stress as a basis for enhanced glioblastoma cell killing by bortezomib in combination with celecoxib or its non-coxib analogue, 2,5-dimethyl-celecoxib. Cancer Res. 2008, 68, 843–851. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Fonseca, C.O.D.; Masini, M.; Futuro, D.; Caetano, R.; Gattass, C.R.; Quirico-Santos, T. Anaplastic oligodendroglioma responding favorably to intranasal delivery of perillyl alcohol: A case report and literature review. Surg. Neurol. 2006, 66, 611–615. [Google Scholar] [CrossRef] [PubMed]
  59. Cho, H.Y.; Wang, W.; Jhaveri, N.; Torres, S.; Tseng, J.; Leong, M.N.; Lee, D.J.; Goldkorn, A.; Xu, T.; Petasis, N.A.; et al. Perillyl alcohol for the treatment of temozolomide-resistant gliomas. Mol. Cancer Ther. 2012, 11, 2462–2472. [Google Scholar] [CrossRef] [Green Version]
  60. Marín-Ramos, N.I.; Pérez-Hernández, M.; Tam, A.; Swenson, S.D.; Cho, H.Y.; Thein, T.Z.; Hofman, F.M.; Chen, T.C. Inhibition of motility by NEO100 through the calpain-1/RhoA pathway. J. Neurosurg. 2019, 133, 1020–1031. [Google Scholar] [CrossRef]
  61. Cerezo, M.; Lehraiki, A.; Millet, A.; Rouaud, F.; Plaisant, M.; Jaune, E.; Botton, T.; Ronco, C.; Abbe, P.; Amdouni, H.; et al. Compounds triggering ER stress exert anti-melanoma effects and overcome BRAF inhibitor resistance. Cancer Cell 2016, 29, 805–819. [Google Scholar] [CrossRef] [Green Version]
  62. Pinkham, K.; Park, D.J.; Hashemiaghdam, A.; Kirov, A.B.; Adam, I.; Rosiak, K.; da Hora, C.C.; Teng, J.; Cheah, P.S.; Carvalho, L.; et al. Stearoyl CoA desaturase is essential for regulation of endoplasmic reticulum homeostasis and tumor growth in glioblastoma cancer stem cells. Stem Cell Rep. 2019, 12, 712–727. [Google Scholar] [CrossRef] [Green Version]
  63. Bakewell, S.J.; Rangel, D.F.; Ha, D.P.; Sethuraman, J.; Crouse, R.; Hadley, E.; Costich, T.L.; Zhou, X.; Nichols, P.; Lee, A.S. Suppression of stress induction of the 78-kilodalton glucose regulated protein (GRP78) in cancer by IT-139, an anti-tumor ruthenium small molecule inhibitor. Oncotarget 2018, 9, 29698–29714. [Google Scholar] [CrossRef] [Green Version]
  64. Prabhu, A.; Sarcar, B.; Kahali, S.; Shan, Y.; Chinnaiyan, P. Targeting the unfolded protein response in glioblastoma cells with the fusion protein EGF-SubA. PLoS ONE 2012, 7, e52265. [Google Scholar] [CrossRef] [Green Version]
  65. Dadey, D.Y.A.; Kapoor, V.; Hoye, K.; Khudanyan, A.; Collins, A.; Thotala, D.; Hallahan, D.E. Antibody targeting GRP78 enhances the efficacy of radiation therapy in human glioblastoma and non-small cell lung cancer cell lines and tumor models. Clin. Cancer Res. 2017, 23, 2556–2564. [Google Scholar] [CrossRef]
  66. Kia, A.; Przystal, J.M.; Nianiaris, N.; Mazarakis, N.D.; Mintz, P.J.; Hajitou, A. Dual systemic tumor targeting with ligand-directed phage and Grp78 promoter induces tumor regression. Mol. Cancer Ther. 2012, 11, 2566–2577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Przystal, J.M.; Waramit, S.; Pranjol, M.Z.I.; Yan, W.; Chu, G.; Chongchai, A.; Samarth, G.; Olaciregui, N.G.; Tabatabai, G.; Carcaboso, A.M.; et al. Efficacy of systemic temozolomide-activated phage-targeted gene therapy in human glioblastoma. EMBO Mol. Med. 2019, 11, e8492. [Google Scholar] [CrossRef]
  68. Kaliberov, S.A.; Kaliberova, L.N.; Yan, H.; Kapoor, V.; Hallahan, D.E. Retargeted adenoviruses for radiation-guided gene delivery. Cancer Gene Ther. 2016, 23, 303–314. [Google Scholar] [CrossRef] [Green Version]
  69. Hughes, S.J.; Antoshchenko, T.; Chen, Y.; Lu, H.; Pizarro, J.C.; Park, H.W. Probing the ATP site of GRP78 with nucleotide triphosphate analogs. PLoS ONE 2016, 11, e0158256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Wang, T.E.; Lai, Y.H.; Yang, K.C.; Lin, S.J.; Chen, C.L.; Tsai, P.S. Counteracting cisplatin-induced testicular damages by natural polyphenol constituent honokiol. Antioxidants 2020, 9, 723. [Google Scholar] [CrossRef] [PubMed]
  71. Wang, J.; Nie, Y.; Li, Y.; Hou, Y.; Zhao, W.; Deng, J.; Wang, P.G.; Bai, G. Identification of target proteins of mangiferin in mice with acute lung injury using functionalized magnetic microspheres based on click chemistry. J. Agric. Food Chem. 2015, 63, 10013–10021. [Google Scholar] [CrossRef]
  72. Bailly, C.; Waring, M.J. Pharmacological effectors of GRP78 chaperone in cancers. Biochem. Pharmacol. 2019, 163, 269–278. [Google Scholar] [CrossRef] [PubMed]
  73. Bhattacharjee, R.; Devi, A.; Mishra, S. Molecular docking and molecular dynamics studies reveal structural basis of inhibition and selectivity of inhibitors EGCG and OSU-03012 toward glucose regulated protein-78 (GRP78) overexpressed in glioblastoma. J. Mol. Model. 2015, 21, 272. [Google Scholar] [CrossRef]
  74. Enogieru, A.B.; Omoruyi, S.I.; Hiss, D.C.; Ekpo, O.E. GRP78/BIP/HSPA5 as a therapeutic target in models of parkinson’s disease: A mini review. Adv. Pharmacol. Sci. 2019, 2019, 2706783. [Google Scholar] [CrossRef] [Green Version]
  75. Ambrose, A.J.; Chapman, E. Function, therapeutic potential, and inhibition of Hsp70 chaperones. J. Med. Chem. 2021, 64, 7060–7082. [Google Scholar] [CrossRef]
  76. Ermakova, S.P.; Kang, B.S.; Choi, B.Y.; Choi, H.S.; Schuster, T.F.; Ma, W.Y.; Bode, A.M.; Dong, Z. (−)-Epigallocatechin gallate overcomes resistance to etoposide-induced cell death by targeting the molecular chaperone glucose-regulated protein 78. Cancer Res. 2006, 66, 9260–9269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Yang, Y. Screening and Mechanism of Drugs for Intervention of Tumor Secretory GRP78. Master’s Thesis, Shanxi University, Taiyuan, China, 2019. [Google Scholar]
  78. Ambrose, A.J.; Zerio, C.J.; Sivinski, J.; Schmidlin, C.J.; Shi, T.; Ross, A.B.; Widrick, K.J.; Johnson, S.M.; Zhang, D.D.; Chapman, E. A high throughput substrate binding assay reveals hexachlorophene as an inhibitor of the ER-resident HSP70 chaperone GRP78. Bioorg. Med. Chem. Lett. 2019, 29, 1689–1693. [Google Scholar] [CrossRef] [PubMed]
  79. Zoni, V.; Khaddaj, R.; Campomanes, P.; Thiam, R.; Schneiter, R.; Vanni, S. Lipid droplet biogenesis is driven by liquid-liquid phase separation. eLife 2020. [Google Scholar] [CrossRef]
  80. Shao, H.; Li, X.; Moses, M.A.; Gilbert, L.A.; Kalyanaraman, C.; Young, Z.T.; Chernova, M.; Journey, S.N.; Weissman, J.S.; Hann, B.; et al. Exploration of benzothiazole rhodacyanines as allosteric inhibitors of protein-protein interactions with heat shock protein 70 (Hsp70). J. Med. Chem. 2018, 61, 6163–6177. [Google Scholar] [CrossRef]
Figure 1. Anti-obesity role of GRP78 and downstream molecular functions associated with obesity. Roles and downstream pathways of GRP78 relevant to obesity initiated by binding of α2-macroglobulin-GRP78 that triggers the expression of IP3, RAS and MAPK, PAk2, PI3K, PLD, COX2, and cPLA2 and increases concentrations of cAMP, Ca2+ and pH value (left), which can activate the downstream pathways (right). Green arrows indicate up-expression. AMPK, adenosine monophosphate (AMP) activated protein kinase; Akt, an ubiquitous serine/threonine kinase, also known as protein kinase B (PKB) or RAS-alpha; ATF2, activating transcription factor-2; cAMP, cyclic adenosine monophosphate; C/EBPα, CCAAT/enhancer-binding protein α (which CCAAT is a distinct pattern of nucleotides with GGCCAATCT consensus sequence that occurs upstream by 60–100 bases to the initial transcription site); COX2, cicloxigenases; cPLA2, cytosolic phospholipase A 2; ERS, endoplasmic reticulum stress; GLUT4, glucose transporter type 4; GRP78, 78 kDa glucose-regulated protein; HSP70, heat shock protein 70 family; HSL, hormone sensitive lipase; HSPA12A, heat shock protein family a (hsp70) member 12A; IP3, inositol triphosphate; IR, insulin resistance; mTOR, mammalian target of rapamycin; PAk2, p21-activated protein kinase; PGC1α, PPARγ coactivator 1α; PDK1, pyruvate dehydrogenase kinase 1; PLD, phospholipase D; PI3K, phosphoinositide 3-kinases; PPARγ, peroxisome proliferator activated receptor γ; PLIN, perilipin; p38 MAPK, p38 mitogen-activated protein kinases; p-Akt, phosphorylated Akt; RAS, renin-angiotensin system; RTN3, reticular protein 3; SIRT1, sirtuin1; SREBP-1c, sterol regulatory element binding protein-1c; SREBPs, sterol regulatory element binding protein; SCAP, SREBP cleavage-activating protein; TG, triglyceride fatty acid; UCP1, uncoupling protein 1.
Figure 1. Anti-obesity role of GRP78 and downstream molecular functions associated with obesity. Roles and downstream pathways of GRP78 relevant to obesity initiated by binding of α2-macroglobulin-GRP78 that triggers the expression of IP3, RAS and MAPK, PAk2, PI3K, PLD, COX2, and cPLA2 and increases concentrations of cAMP, Ca2+ and pH value (left), which can activate the downstream pathways (right). Green arrows indicate up-expression. AMPK, adenosine monophosphate (AMP) activated protein kinase; Akt, an ubiquitous serine/threonine kinase, also known as protein kinase B (PKB) or RAS-alpha; ATF2, activating transcription factor-2; cAMP, cyclic adenosine monophosphate; C/EBPα, CCAAT/enhancer-binding protein α (which CCAAT is a distinct pattern of nucleotides with GGCCAATCT consensus sequence that occurs upstream by 60–100 bases to the initial transcription site); COX2, cicloxigenases; cPLA2, cytosolic phospholipase A 2; ERS, endoplasmic reticulum stress; GLUT4, glucose transporter type 4; GRP78, 78 kDa glucose-regulated protein; HSP70, heat shock protein 70 family; HSL, hormone sensitive lipase; HSPA12A, heat shock protein family a (hsp70) member 12A; IP3, inositol triphosphate; IR, insulin resistance; mTOR, mammalian target of rapamycin; PAk2, p21-activated protein kinase; PGC1α, PPARγ coactivator 1α; PDK1, pyruvate dehydrogenase kinase 1; PLD, phospholipase D; PI3K, phosphoinositide 3-kinases; PPARγ, peroxisome proliferator activated receptor γ; PLIN, perilipin; p38 MAPK, p38 mitogen-activated protein kinases; p-Akt, phosphorylated Akt; RAS, renin-angiotensin system; RTN3, reticular protein 3; SIRT1, sirtuin1; SREBP-1c, sterol regulatory element binding protein-1c; SREBPs, sterol regulatory element binding protein; SCAP, SREBP cleavage-activating protein; TG, triglyceride fatty acid; UCP1, uncoupling protein 1.
Ijerph 19 15965 g001
Figure 4. Regulatory functions of GRP78 and downstream molecular functions relevant to anti-obesity. Domain structures (top) and 3D structural model (middle) of GRP78, molecules interacting with either the ATPase domain (in magenta), β sandwich subdomain (in red) or the helix subdomain (in cyan) of GRP78, and chemical structures of nucleosides (bottom). The cited small molecules can up (+) or down (−) regulate GRP78. Modified from [72].
Figure 4. Regulatory functions of GRP78 and downstream molecular functions relevant to anti-obesity. Domain structures (top) and 3D structural model (middle) of GRP78, molecules interacting with either the ATPase domain (in magenta), β sandwich subdomain (in red) or the helix subdomain (in cyan) of GRP78, and chemical structures of nucleosides (bottom). The cited small molecules can up (+) or down (−) regulate GRP78. Modified from [72].
Ijerph 19 15965 g004
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pan, D.; Yang, Y.; Nong, A.; Tang, Z.; Li, Q.X. GRP78 Activity Moderation as a Therapeutic Treatment against Obesity. Int. J. Environ. Res. Public Health 2022, 19, 15965. https://doi.org/10.3390/ijerph192315965

AMA Style

Pan D, Yang Y, Nong A, Tang Z, Li QX. GRP78 Activity Moderation as a Therapeutic Treatment against Obesity. International Journal of Environmental Research and Public Health. 2022; 19(23):15965. https://doi.org/10.3390/ijerph192315965

Chicago/Turabian Style

Pan, Dongjin, Yunzhu Yang, Aihua Nong, Zhenzhou Tang, and Qing X. Li. 2022. "GRP78 Activity Moderation as a Therapeutic Treatment against Obesity" International Journal of Environmental Research and Public Health 19, no. 23: 15965. https://doi.org/10.3390/ijerph192315965

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop