Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Nat Rev Cancer. Author manuscript; available in PMC 2014 Oct 1.
Published in final edited form as:
PMCID: PMC4158750
NIHMSID: NIHMS621506
PMID: 24658275

Glucose regulated proteins in cancer: molecular mechanisms and therapeutic potential

Associated Data

Supplementary Materials

Preface

The glucose regulated proteins (GRPs) are stress inducible chaperones majorly residing in the endoplasmic reticulum (ER) and the mitochondria. Recent advances reveal that the GRPs serve distinct functions from the related heat shock proteins (HSPs), and they can be actively translocated to other cellular locations and assume novel functions controlling signaling, proliferation, invasion, apoptosis, inflammation and immunity. Mouse models further identified their specific roles in development, tumorigenesis, metastasis and angiogenesis. This Review describes their discovery, regulation and their biological functions in cancer. Promising agents using or targeting the GRPs are being developed, and their efficacy as anti-cancer therapeutics is also discussed.

Introduction

Glucose regulated proteins, GRP78 (also known as BiP and HSPA5), GRP94 (also known as gp96 and HSP90B1), GRP170 (also known as ORP150 and HYOU1) and GRP75 (also known as mortalin and HSPA9) are stress-inducible molecular chaperones belonging to the heat shock protein (HSP) family (Box 1). Unlike the majority of the HSPs, which reside mainly in the cytosol and nucleus, these GRPs are found in the endoplasmic reticulum (ER) and the mitochondria, which are key organelles regulating protein quality control and metabolic balance1-4. In their traditional chaperone roles, these GRPs facilitate protein folding and assembly and the export of misfolded proteins for degradation. Coupled with their Ca2+ binding functions, they maintain the integrity and homeostasis of the ER and the mitochondria under physiological and pathological conditions.

Box 1

Discovery of the GRPs

The GRPs were discovered in the mid-1970s as constitutively expressed cellular proteins induced by glucose starvation or a block in protein glycosylation, hence they were named glucose regulated proteins169-171. GRP78, encoded in humans by HSPA5, shares 60% amino acid homology with HSP70, including the ATP binding domain required for their ATPase catalytic activity (see figure) and is a HSP70 analogue in the ER. GRP78 is identical to BiP, originally discovered as an Ig heavy chain binding protein172-174. This led to the designation of GRP78 as an ER molecular chaperone, and it is now established as a ubiquitous protein essential for processing a wide repertoire of client proteins and maintaining the structural integrity of the ER1,32,175.

Following the discovery of hamster GRP94 in 1984176, GRP94 has been identified as endoplasmin (discovered as a Ca2+ binding protein)177, ERp99 (discovered as a major ER glycoprotein)178, and as the tumour rejection antigen gp96179. GRP94 encoded in humans by HSP90B1, shares 50% amino acid homology with HSP90 and is one of four HSP90 isoforms180. As well as being an ER chaperone, GRP94 is also a regulator of innate and adaptive immunity1,4,181.

GRP75 encoded in humans by HSPA9 was first identified as a 66 kDa protein (p66mot-1) linked to mortality with anti-proliferative properties182. cDNA cloning and a homology search revealed 80% homology to yeast mitochondrial HSP and 70% homology with mouse HSP70 (HSPA1A). Although GRP75 can localize to multiple subcellular sites, its primary location is in the mitochondria, as directed by its N-terminal leader sequence3 (see figure).

Studying proteins induced by glucose starvation led to the discovery of a 150 kDa protein, GRP170183. GRP170, encoded in humans by HYOU1, is a large HSP70/HSP110-like protein in the ER184 that is induced by hypoxia185,186 (see figure).

Therefore, all of the GRPs can function as chaperones that can be induced during cellular stress.

An external file that holds a picture, illustration, etc.
Object name is nihms621506f4.jpg
Box 1 Figure Legend (Optional). Functional domains of the GRPs

The locations of the signal sequence targeting the proteins into the ER (GRP78, GRP94 and GRP170) or the mitochondria (GRP75) are shown. The ATPase and substrate binding domains are indicated for all the GRPs. The location and the ER retention motifs for GRP78, GRP94 and GRP170 are shown. The Ca2+ binding, receptor binding and dimerization domains for GRP94 are denoted.

GRP overexpression is widely reported in cancer cell lines, associating with aggressive growth and invasive properties5,6 (Supplemental Table 1). During the past decade, exciting discoveries have been made in identifying common and distinctive functions of these GRPs in cancer. In sustaining ER protein folding capacity and maintaining ER stress sensors and ER associated pro-apoptotic machineries in their inactive state, GRP78 regulates the balance between cancer cell viability and apoptosis7. GRP94 is essential for the processing of proteins that have been implicated in tumorigenesis, such as insulin-like growth factor 1 (IGF-1), Toll-like receptors (TLRs) and integrins4. GRP170, which has ADP-ATP exchange function, is both a co-chaperone for GRP78 and an independent chaperone, and is critical for vascular endothelial growth factor A (VEGFA) processing and maturation2,8,9. GRP75 interacts with the tumour suppressor p53, inactivating the capacity of p53 to function as a transcription factor and inducing apoptosis10. Furthermore, these GRPs, traditionally regarded to reside exclusively in the ER lumen, can be actively translocated to other cellular locations and secreted, and have additional functions that control signaling, proliferation, invasion, apoptosis, inflammation and immunity11-14. ER stress, as well as development of therapeutic resistance, actively promotes cell surface expression of GRP78, which serves as an upstream regulator of the PI3K-AKT oncogenic signaling pathway15-17. GRP78 is also a downstream target of AKT activation18,19. At the cell surface, GRP94 and GRP170 function in antigen presentation, and their secreted forms have the ability to elicit innate and adaptive immune responses, which could be useful in the development of cancer vaccines1,2,20.

Through the use of cancer cell lines, xenografts and conditional knockout mouse models, the important roles of these GRPs in cancer are being established5,20,21. Promising therapeutics specifically directed against the GRPs, including conjugated peptides and toxins, antibodies, small molecules and microRNAs, are being developed5,20,22. Thus, these GRPs represent novel prognostic markers and targets5, as well as mediators or vaccines for anti-cancer therapy2,23 that warrant vigorous investigation.

GRPs in the stress response

The GRPs are ubiquitous chaperones that are constitutively expressed at basal level and that sustain organ homeostasis through different mechanisms (Supplemental Table 2). The induction of the GRPs is widely used as an indicator for the onset of ER stress and studies into their transcriptional activation mechanism (Box 2) have facilitated the discovery of novel intracellular signaling pathways whereby stress from the ER can be communicated to the nucleus to initiate transcription of the unfolded protein response (UPR)-associated genes20,24,25. Cancer cells are subjected to ER stress triggered by both intrinsic and extrinsic factors, such as altered cell metabolism, hyperproliferation, hypoglycemia, hypoxia, acidosis, viral infection and genetic lesions leading to the production of mutated proteins that misfold20,26. These adverse conditions impinge on proper protein folding in the ER, creating ER stress. GRP78 regulates the UPR by binding to and inactivating all three ER stress transducers [PRKR-like endoplasmic reticulum kinase (PERK), inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6)]7,20 under non-stressed conditions. When misfolded proteins accumulate in the ER, GRP78 binds to them, thereby releasing the UPR sensors and leading to the activation of the UPR pathways27-31 (Figure 1). Conversely, when GRP78 is depleted or inactivated, the UPR can be triggered spontaneously, with diverse physiological consequences32-35 (Supplemental Table 2). Nonetheless, GRP induction is not limited to ER stress. For example, autophagy-defective tumor cells upregulate ER chaperones in response to metabolic stress36, and histone deacetylase inhibitors activate GRP78 transcription without concomittantly inducing stress response in general37 (Box 2).

An external file that holds a picture, illustration, etc.
Object name is nihms621506f1.jpg
GRPs in UPR and the stress response

ER luminal GRP78 acts as a UPR signaling regulator by binding to and maintaining the ER stress sensors (PERK, ATF6 and IRE1) in inactive forms. It also binds to and suppresses the activation of ER associated caspase-7 and -12 (C7/12). Upon ER stress, GRP78 is titrated away through binding to mal-folded proteins. This triggers the UPR, as exemplified by dimerization of PERK and IRE1, and activation of their downstream signaling pathways, leading to arrest of translation and ER associated protein degradation (ERAD). The UPR also generates the active nuclear form of ATF6, as well as ATF4, and the spliced form of XBP (XBP-1s), which act in concert with other transcriptional factors including YY1, NF-Y and TFII-I and chromatin modifiers, to activate the ER stress response element (ERSE) present on the promoters of ER stress responsive genes. A major UPR response is to induce the transcription of ER folding proteins such as the GRPs to booster the ER protein folding capacity, as well as the mitochondrial (Mito) chaperone GRP75. Stressed cells actively promote re-localization of GRP78 and GRP94 to the plasma membrane (PM), and in some instances, their secretion, and generate a cytosolic isoform of GRP78 (78va) through alternative splicing. Nonetheless, UPR can also induce transcription of the pro-apoptotic transcription factor CHOP, and following release from GRP78, caspase-7 and caspase-12 are activated, triggering apoptosis. Thus, the UPR regulates the balance between survival and cell death in stressed cells, and the up-regulation of the GRPs represents a major adaptive, protective measure through maintenance of cellular homeostasis.

Box 2

Transcriptional activation of GRP promoters

The GRP78 promoter contains three ER stress response elements (ERSE) located upstream of the TATA element187,188. In non-stressed cells, NF-Y (also known as CBF), SP1 and histone deacetylase 1 (HDAC1) bind to the ERSEs and maintain GRP78 at a low basal transcriptional level189. Upon ER stress, ATF6 dissociates from GRP78 in the ER and translocates to the Golgi where it is cleaved to generate a form of ATF6 that can enter the nucleus [ATF6(N)] (see Figure 1). ATF6(N), which binds to the ERSE through binding to NF-Y, also associates with YY1 and increases the binding of YY1 to the GRP78 promoter189. YY1 associating protein arginine methyltransferase (PRMT) and the histone transacetylase p300 are also recruited to the GRP78 promoter, concurrent with histone 4 acetylation and arginine 3 methylation known to activate transcription189. Together, these transcription factors and chromatin modifiers form an ERSE binding complex, which can include the transcription factor TFII-I that functions as a scaffold protein190. While the IRE1/XBP-1 is also an important branch of ER stress, MEFs devoid of XBP-1 showed only modest effect on ER stress induction of GRP78 and no effect on GRP94191. Thus the XBP-1 may contribute to GRP transcription but is not obligatory. ER stress induction of GRP78 might also be partly attributed to ERSE-independent pathways, mediated by ATF4, a bZIP transcription factor, in complex with cAMP-responsive element-binding protein 1 (CREB1)30. Promoters of GRP94, GRP75 and GRP170 contain the ERSE consensus sequence and are similarly regulated4,25,125,192.

Biological functions of the GRPs in cancer

As summarized in this section, the GRPs, in both UPR-dependent and UPR-independent functions, have important roles in regulating a variety of processes that are essential for tumorigenesis at multiple cellular locations (Figure 2).

An external file that holds a picture, illustration, etc.
Object name is nihms621506f2.jpg
GRPs in survival and immunity

The majority of GRP78, GRP94 and GRP170 are located in the ER lumen serving as ER folding proteins, and GRP75 is primarily a mitochondrial chaperone. Under ER stress or pathological stress conditions, a subfraction of GRP78 and GRP94 translocate to the cell surface and their secreted forms can be detected. Cell surface GRPs control critical growth and apoptotic signaling functions, as well as immune functions notably antigen presentation. ER stress also induces GRP78 translocation into the nucleus and mitochondria, and alternative splicing of GRP78 mRNA leading to the generation of a cytosolic isoform. The pro-survival functions of the GRPs in the various subcellular locations are indicated in blue, and the immune functions in red.

Proliferation

GRP78 expression levels correlated with proliferative rates of human glioma cell lines and knockdown of GRP78 by small interfering RNA (siRNA) suppresses their growth38. In a mouse mammary tumour virus (MMTV)-Polyoma middle T (PyT) mammary tumour model, Grp78 heterozygosity was sufficient to prolong the latency period and impede cancer growth in part through suppression of tumour cell proliferation39 (Table 1). How might GRP78 facilitate proliferation? As an ER chaperone, GRP78 controls processing and maturation of a wide variety of cell surface receptors and secretory proteins crucial for the ability of cancer cells to respond to extrinsic proliferative signals20. GRP78 is also a regulator of Wnt-β-catenin proliferative signaling through its stabilization of Wnt in the ER. When GRP78 was dissociated from Wnt under hypoxic conditions, Wnt was not properly processed, leading to its proteasomal degradation and reduced Wnt secretion40.

Table 1

Role of GRP78 and GRP94 in mouse models of cancer
Cancer TypeMouse GenotypeKOKey PhenotypesRefs.
Breast carcinomaGrp78+/-; MMTV PyVT+/-Prolongs latency period, impedes tumour growth Tumour cell proliferation↓, angiogenesis↓, apoptosis↑39
Prostate adenocarcinomaGrp78f/+ or Grp78f/f; Ptenf/f; probasin Cre+/- or -/-Suppresses tumorigenesis AKT activation↓47
LeukemiaGrp78f/+; Ptenf/f; Mx1-Cre+/-Suppresses leukemic blast cell expansion PI3K/AKT signaling↓ Normal hematopoietic phenotype48
Liver cancerGrp78f/f; Ptenf/f; Alb-Cre-/-Exacerbates steatosis and liver injury Liver progenitor cell activation, accelerates tumorigenesis Strong GRP78 re-expression in cancer lesions111
Grp94f/f; Ptenf/f; Alb-Cre-/-Minor liver injury, disrupts cell adhesion protein organization Liver progenitor cell proliferation↑ ERK activation, accelerates tumorigenesis113
Multiple myelomaHsp90b1f/f; CD19-Cre; XBP1-sTg-/-Suppresses tumour growth Inhibits Wnt/β-catenin signaling53
Colorectal cancerHsp90b1f/f; LysM-Cre-/-Reduction in number and size of colitis-associated colon cancer Colonic epithelial β-catenin mutation↓,Wnt signaling↓54

GRP78 might also promote cell proliferation from the cell surface. ER stress or ectopic expression of GRP78 leads to localization of a subfraction of GRP78 on the cell surface15. Specific proteins have been reported to transport GRP78 to the cell surface in different cell types, such as the carrier protein MTJ-1 in macrophages and the tumour suppressor prostate apoptosis response 4 (PAR-4; also known as PAWR) in the prostate cancer cell line PC-341,42.

Cell surface GRP78 acts as a multifunctional receptor impacting both cell proliferation and viability11-14. For example, in prostate cancer cells, cell surface GRP78 serves as receptor for the activated form of the plasma proteinase inhibitor α2-macroglobulin (α2-M*)43, triggering ERK and AKT activation and increased DNA and protein synthesis44. AKT signaling, which promotes proliferation and inhibits apoptosis, is also triggered by autoantibodies against the N-terminus of GRP78 that are found in cancer patients45. How might GRP78 regulate AKT activation? Cell surface GRP78 co-localizes with PI3K, an activator of AKT, and co-immunoprecipitates with PI3K subunits16,17. Furthermore, in cell culture model systems, overexpression of GRP78 leads to increased PIP3 production (a signaling molecule downstream of PI3K) and mutation of the N-terminal region of GRP78 reduced both the binding of cell surface GRP78 to PI3K and PIP3 production16. A requirement for GRP78 in a serum-stimulated increase in PIP3 production has also been reported in human leukemic cells34.

PTEN (phosphatase and tension homolog deleted in chromosome 10), which encodes a plasma membrane lipid phosphatase that antagonizes the PI3K signaling pathway, is a major tumour suppressor gene in human cancer46. A biallelic conditional knockout mouse model of Grp78 and Pten in the prostate epithelium or bone marrow showed that GRP78 deficiency reduces PI3K-AKT activation, which normally occurs as a result of PTEN loss in these cells, and potently inhibits prostate tumorigenesis47 and leukemogenesis48, respectively (Table 1). Although cell surface GRP78 has been shown to regulate PI3K signaling, further studies are required to determine whether GRP78 in the ER or other cellular locations might also regulate PI3K-AKT signaling. Recent studies revealed GRP78 is a downstream target of the IGF-1R-PI3K signaling pathway in mouse embryo fibroblasts, as well as in cancer cell lines18,19, and this could represent a feedback regulatory mechanism that balances GRP78 expression and cancer cell proliferation.

Another pro-proliferative mechanism of GRP78 is the interaction of cell surface GRP78 with Cripto-1 (also known as teratocarcinoma-derived growth factor 1), a glycosylphosphatidylinositol (GPI)-anchored, developmentally regulated, oncoprotein.49 Disruption of cell surface GRP78 and Cripto complex blocked Cripto activation of MAPK and PI3K pathways and modulation of activin-A, activin-B, nodal and transforming growth factor-β1 signaling50. Thus, cell surface GRP78 is a necessary mediator of Cripto proliferative signaling in human cancer.

GRP94 controls the maturation and secretion of IGFs, which are important mitogenic factors51, and binding of IGF-1 or IGF-2 to the IGF-1R leads to PI3K-AKT activation. GRP94 regulates the processing of the low density lipoprotein receptor-related protein 6 (LRP6), a Wnt co-receptor52. Without GRP94, LRP6 is not exported from the ER to the cell surface, leading to attenuation of the pro-proliferative and pro-survival Wnt-β-catenin signaling pathway. This is the proposed mechanism for the attenuation of multiple myeloma and inflammatory colorectal cancer in mouse models where Grp94 is deleted in B-cells53 and macrophages54, respectively (Table 1). In breast cancer cells that are able to proliferate under chronic exposure to reactive oxygen species (ROS) in vitro, the expression of GRP94, but not HSP90 or GRP78, is increased55. ROS are counteracted by the production of antioxidants and the formation of disulphide bonds in proteins in the ER, which is promoted by GRP94.

Overexpression of GRP75 in mouse fibroblasts leads to anchorage-independent growth, and formation of tumours when transplanted into nude mice56. Contributing factors might include the role of GRP75 as a mitochondrial protein importer and its ability to retain p53 in the cytoplasm, leading to down-regulation of p53 target genes such as Cdkn1a and Mdm2. This effect on p53 has been shown in a subset of neuroblastomas57. Another client protein of GRP75 is fibroblast growth factor-1 (FGF-1) which possesses broad mitogenic activities and functions as a modifier of endothelial cell migration and proliferation and is therefore pro-angiogenic58.

Apoptosis

In general, the GRPs are suppressors of apoptosis20. Caspase-7, an executioner caspase that is associated with the ER, can be activated by the chemotherapeutic agent etoposide, and GRP78, in a manner dependent on its ATP binding activity, forms complex with caspase-7 (Figure 1) and protects cells from apoptosis induced by etoposide59,60. Recently, a functional relationship was uncovered at the outer surface of the ER between GRP78, the pro-apoptotic protein BIK and the anti-apoptotic protein BCL-261,62. GRP78 and BCL-2 form separate complexes with different domains of BIK. BIK sequestration of BCL-2 reduces BCL-2 interaction with the ER, leading to ER Ca2+ release, translocation of the pro-apoptotic protein BAX to the mitochondria and the release of cytochrome c to the cytosol, which initiates apoptosis. However, high levels of GRP78 sequester BIK, which releases the inhibition of BCL‐2, thereby suppressing apoptosis62.

These observations, however, raise the important question of how GRP78 as an ER lumen protein, can interact with cytosolic proteins that associate with the outer ER membrane. Intriguingly, two independent studies showed that a subpopulation of GRP78 from isolated microsomes was resistant to sodium carbonate extraction and existed as a partially protease resistant (presumably transmembrane) protein59,60. However, despite the presence of some weak hydrophobic motifs supporting this possibility, the primary amino sequence of GRP78 does not predict a traditional transmembrane configuration under normal physiological conditions. Thus, the interaction between GRP78 and cytosolic proteins will have to be mediated either by an unconventional form of GRP78 that spans the ER membrane or lumenal GRP78 in complex with other ER transmembrane proteins; this issue remains to be resolved.

ER stress induces alternative splicing of GRP78, generating a cytosolic isoform (GRP78va) that regulates PERK signaling and enhances leukemia cell survival63. ER stress also promotes GRP78 localization to the mitochondria, which are physically and functionally interconnected with the ER (Box 3). Mitochondria-associated GRP78 can bind to RAF1 and this interaction is involved in the maintenance of mitochondrial permeability and thus protective against ER-stress-induced apoptosis64. In support of the anti-apoptotic functions of GRP78, knockout of Grp78 in various tissues led to caspase activation and tissue atrophy33,34,65,66 (Supplemental Table 2). In breast, prostate and leukemic cancer models, heterozygous and/or homozygous knockout of Grp78 increased tumour apoptosis and impeded tumour progression39,47,48 (Table 1).

Box 3

GRP interconnectivity in the ER and mitochondria

In addition to protein folding and secretion, the ER is central to Ca2+ homeostasis and the regulation of apoptosis. As low affinity, high capacity Ca2+ binding proteins, GRP78 and GRP94 help maintain ER Ca2+ balance1. Mitochondria are the site of oxidative phosphorylation-dependent ATP generation that is critical for maintaining energy homeostasis, and they also integrate and transduce apoptotic signals, and participate in the regulation of intracellular Ca2+. Structural and functional analyses reveal zones of close contact between the ER and mitochondria, referred to as mitochondria associated membranes (MAMs)193. MAMs enable the efficient transmission of Ca2+ from the ER to mitochondria and molecular chaperones such as GRP75, calnexin, calreticulin, ERp44, ERp57 and the sigma-1 receptor coexist at MAMs194. Signaling from the ER to mitochondria can be critical in the induction of mitochondrial dependent cell death pathways61,195,196. The UPR promotes GRP78 localization to the mitochondria64,197 and slows the increase of Ca2+ in mitochondria after stress and reduces free radical generation, associating with protection against ischemic injury196. For cancer cells, over-expression of GRP78 might protect them against damage resulting from potentially carcinogenic free radicals generated from endogenous or exogenous source. GRP78 might also regulate mitochondria energy balance through modulation of GRP75 and peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) expression198. In specific tissues, GRP78 haploinsufficiency leads to compensatory upregulation of GRP75, GRP94 and other ER chaperones33,198; conversely, GRP94 deficiency triggers GRP78 upregulation199.

Collectively, these findings demonstrate the interconnectivity of the GRPs and adaptive responses in cells to maintain functional Ca2+ and ATP homeostasis in the ER and mitochondria. These mechanisms might result in a survival advantage to cancer cells in the face of the variety of metabolic and environmental perturbations that exist in growing tumours.

Through interaction with α2-M*, cell surface GRP78 promotes 1-LN prostate cancer cell survival by activating the AKT and NF-κB signaling cascades67. In hypoxic HT1080 fibrosarcoma cells, cell surface GRP78 serves as a receptor for Kringle 5, a human plasminogen factor, which upon internalization, competes with procaspase-7 binding to the ATP binding domain of ER GRP78, leading to caspase-7 activation and tumor cell apoptosis68. However, one study indicates that cell surface GRP78, along with PAR4 has a pro-apoptotic function through mediating TNF-related apoptosis-inducing ligand (TRAIL; also known as TNFSF10) activation, triggering the extrinsic apoptotic pathway in PC3, HeLa and H460 cells42. Nonetheless, in MCF-10A and MDA-MB-231 cells, GRP78 prevents TRAIL-induced apoptosis and is therefore a pro-survival factor69. Thus, the effect of GRP78 on TRAIL-induced apoptosis may be context dependent. Besides modulating apoptosis, GRP78 has been implicated in protective autophagy through maintenance of ER structural integrity32, and modulation of mTOR signaling in estrogen resistant breast cancer cells70.

GRP94 maintains ER Ca2+ homeostasis and protects cancer cells from apoptosis71. GRP94 deficiency in human multiple myeloma cells resulted in apoptosis through inhibition of the Wnt-survivin pathway53. In the SkBr3 breast cancer cell line that abundantly expressed cell surface HER2 (also known as ERBB2), pharmacological inactivation of GRP94 destabilized HER2 and inhibited RAF1-MAPK survival signaling at the cell membrane72. In cancer cells, GRP170 is upregulated by hypoxia and by drugs such as celecoxib and proteasome inhibitors, and knockdown of GRP170 activated the expression of the UPR pro-apoptotic factor CHOP and stimulated apoptosis73,74. GRP170 may also protect cancer cells against cell death through blocking ER Ca2+ release or delaying the onset of UPR by binding to the ER stress sensors75,76.

Angiogenesis

Eliminating the tumour vasculature, which supplies nutrients and oxygen within the tumour, is a key strategy for anti-cancer therapy. Tumour-associated endothelial cells are physiologically and functionally different from endothelial cells derived from normal tissues, and express high levels of GRP78 compared with normal organs38,77,78. Grp78+/- mice, as well as mice with conditional heterozygous knockdown of Grp78 in the host endothelial cells, showed a dramatic reduction in tumour microvessel density (MVD) while having no effect on the MVD of normal organs39 (Table 1). Grp78 knockdown impairs immortalized endothelial cell proliferation, survival and migration in vitro, supporting its requirement for neoangiogenesis in primary tumour and metastatic growth39.

GRP78 is expressed on the surface of proliferating endothelial cells68,79. Cell surface GRP78 assoicates with the GPI (glycosylphosphatidylinositol) anchored T-cadherin and mediates T-cadherin-dependent endothelial cell survival80. In exploiting cell surface GRP78 for anti-angiogeneis therapy, plasminogen Kringle 5 is reported to bind GRP78 in glioma endothelial cells for induction of apoptosis68, which can be sensitized by radiation dependent on internalization of cell surface GRP78 by the low-density receptor-related protein and activation of p38 MAK78. Other studies however suggest that the apoptotic effect of Kringle 5 in proliferating endothelial cells and 1-LN prostate cancer cells was mediated by the cell surface voltage-dependent anion channel which co-localizes with GRP78 and is regulated by GRP7881. VEGF can induce cell surface expression of GRP78 in endothelial cells and knockdown of GRP78 suppressed VEGF-mediated MAPK signaling and endothelial cell proliferation82. On the other hand, knockdown of GRP170, while having no effect on GRP78 and GRP94 expression, resulted in retention of VEGF in the ER and blocked its secretion83. Collectively, these studies suggest that targeting GRP78 and GRP170 could achieve a dual effect in suppressing tumour growth as well as tumour angiogenesis.

Invasion and metastasis

Tumour metastasis is a multistep process and requires enhancement of specific tumour cell properties including degradation of the extracellular matrix (ECM), migration, invasion, angiogenesis and survival. The level of intracellular GRP78, as well as cell surface GRP78, is increased in metastatic cancer cell lines, lymph node metastases and human metastatic lesions6,84-86. Knockdown of GRP78 suppresses tumour cell invasion in vitro and suppresses metastatic growth in xenograft and syngeneic tumour models87-89. In addition to protecting metastatic tumour cells from the adverse host environment and promoting angiogenesis, GRP78 has been shown to promote cell motility. One mechanism is through cell surface GRP78 acting as co-receptor for ligands signaling the activation of kinases known to enhance migration, such as AKT, focal adhesion kinase (FAK) and p21-activated kinase 2 (PAK2)88,90. It has also been proposed that cell surface GRP78 acts as a bridge protein for the urokinase-type plasminogen activator (uPA-uPAR) protease system, which can mediate degradation of the ECM and promote invasion88. Like GRP78, GRP94 overexpression is associated with lymph node metastasis and carcinoma recurrence, and silencing of GRP94 inhibits migration and proliferation of MDA-MB-231 breast cancer cells in vitro55,91. GRP94 client proteins include cell interaction and cell matrix component, such as integrins, which might explain its influence on cell invasion. Recently, it was demonstrated that a cell-permeable peptide that competitively inhibited the interaction between GRP94 and integrins blocked cell invasion92. GRP75 overexpression is associated with liver cancer metastasis93, and GRP170 upregulation is observed in invasive breast cancer94 (Supplemental Table 1). Thus, the GRPs are novel protein targets for the inhibition of cancer metastasis.

Inflammation and immunity

ER stress can drive a pro-inflammatory program in tumour cells and macrophages that facilitates tumour progression. Additionally, stressed tumour cells secrete mediators that stimulate macrophages to produce pro-inflammatory cytokines, further amplifying the pro-inflammatory response of tumour cells20,95. On the other hand, cancer cell survival requires resistance against host immune defenses. GRP78 regulates inflammation and immunity through multiple mechanisms20,96. As a major ER chaperone, GRP78 facilitates the processing and secretion of cytokines and chemokines96,97. Acute ablation of Grp78 in adult mice results in alteration of their chemokine and cytokine profile34 (Supplemental Table 2). In terms of immune evasion, GRP78 protects fibrosarcoma cells from lysis by cytotoxic T lymphocytes (CTL) and tumour necrosis factor in vitro, and when fibrosarcoma cells incapable of inducing GRP78 were injected into mice, tumours were either not formed or rapidly regressed with evidence of cytotoxic T cell response98.

GRP78 is an obligatory binding partner for cell surface major histocompatibility complex (MHC) class I molecules99. Acting as the α2M* cell surface signaling receptor, GRP78 regulates the Gs-mediated cAMP production and the pro-inflammatory COX-2-PGE-cAMP signaling cascade100,101. Regulatory T cells (Tregs) are a subpopulation of T cells that drive immune suppression. In some cancers, increased numbers of Tregs promote cancer progression by active suppression of the immune responses against the tumour. Cell surface GRP78 in T cells forms a complex with and confers stabilization to cell surface TGF-β which is an immune regulator and inducer of Tregs102. Some cancer cells secrete GRP78, which modulates human monocyte differentiation into mature dendritic cells and subsequent recruitment of T cells leads to generation of Tregs103.

GRP94 has an important role in immunity by facilitating MHC class I molecule-mediated antigen presentation; by inducing the maturation and activation of various cells involved in innate and adaptive immune responses; and by secretion of proinflammatory cytokines20. GRP94 is the unique and obligatory chaperone of Toll-like receptors (TLRs), facilitating their maturation and translocation to the cell surface. Macrophage-specific knockout of Grp94 resulted in lack of response to TLR ligands and loss of innate immune function104 (Supplemental Table 2), and exhibited reduced colitis and inflammation-associated colon tumorigenesis54. Thus, GRPs regulate inflammation and immunity in both tumour cells and through interactions with the tumour microenvironment (Figure 2).

Stem cell regulation

The notion that cancers are perpetuated by a small population of tumour initiating cells (TICs) that exhibit stem cell-like properties suggests a link between deregulated stem cell activation and cancer development. Initially identified for leukemia, TICs have also been implicated in solid tumours. Hematopoietic stem cells (HSCs) must maintain a balance between quiescence and activation to respond to demands for hematopoiesis yet sustaining long-term stem cell maintenance. Consistent with the pro-survival properties of GRP78, acute inducible ablation of GRP78 in the adult hematopoietic system resulted in intrinsic reduction of the HSC pool through increased apoptosis34 (Supplemental Table 2). Inactivation of PTEN in bone marrow HSCs led to activation of the PI3K-AKT pathway, expansion of the HSC population, development of a myeloproliferative disorder and eventually leukemia105. Strikingly, heterozygous knockdown of Grp78 in Pten-null mice was sufficient to inhibit PI3K-AKT activation, restore the HSC population to a normal level and suppress leukemic blast expansion48 (Table 1). This effect is mediated at least in part by GRP78 at the cell surface, as treatment of the Pten-null mice with a GRP78 targeted antibody also suppressed AKT activation and leukemic blast formation17. Despite their similarity as ER chaperones, acute loss of GRP94 in the bone marrow led to AKT activation and expansion of HSCs, corresponding with loss of surface integrin β4 expression and HSC niche attachment106,107 (Supplemental Table 2). These findings provide the first evidence that GRPs regulate HSC homeostasis through distinct pathways with different outcomes.

In head and neck TICs, expression of GRP78 at the cell surface is associated with self-renewal, suppression of differentiation and radioresistance108,109. RNAi-mediated silencing of GRP78 suppressed the growth of head and neck TICs in a mouse xenograft model, suggesting that cell surface GRP78 is a novel biomarker of TICs and a potential therapeutic target108,109. PTEN inactivation, which occurs in about half of all cases of human liver cancer, results in steatosis, liver injury and inflammation, which lead to liver progenitor cell (LPC) proliferation and the development of liver cancer110. Reduction of GRP78 to less than 25% by genetic knockout in the mouse liver resulted in steatosis, but did not trigger LPC activation or malignancies111,112 (Supplemental Table 2). However, a similar reduction in GRP78 expression in the Pten-null liver model increased steatosis and liver injury and accelerated hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC) formation111 (Table 1). Strikingly, intense GRP78 re-expression was observed in the cancer lesions, and GRP78 expression in the surrounding liver tissue also reverted back to the wild-type level111, suggesting repopulation of the liver by GRP78 positive, unrecombined cells conferring survival advantage, as reported for other tissues35. Knocking out Grp94 in the liver caused only mild injury, but in the Pten-null mice loss of Grp94 perturbed cell adhesion, stimulated LPC proliferation and accelerated HCC and CC progression113 (Table 1). In human liver cancer, as well as in the PTEN-null mouse model, the levels of GRPs are upregulated and correlate with poor prognosis (Supplemental Table 1). So how can these observations be reconciled? One plausible explanation is that in organs where loss of the GRPs leads to progenitor cell activation, when coupled with other carcinogenic events, tumorigenesis may be accelerated. However, there is generally a gain rather than a loss of GRP function in cancer due to stress-induced expression of GRPs. Under these conditions, the GRPs, with their pro-proliferation and anti-apoptotic functions, protect tumour cells from the host defense systems and promote tumour progression and resistance.

GRPs in therapeutic resistance

The expression of GRPs, in both tumour cells and the stromal cells, as an adaptive response to stress induced by cancer treatments, could represent a major obstacle to therapeutic efficacy5,26,77,114. GRP78 has been extensively documented to confer resistance against a wide range of therapies including chemotoxic drugs, anti-hormonal agents, DNA damaging agents, anti-angiogenesis drugs and chromatin-modifying drugs, as well as radiation therapy20,21,115-117. The effects are observed in proliferating and dormant cancer cells, TICs, as well as in tumour associated endothelial cells, and involve not only the ER form of GRP78, but also the stressed-induced cytosolic isoform63, the secreted form118 and the cell surface form of GRP78108. Although less well-studied, GRP94 and GRP170 have been linked with chemoresistance in various tumours71,84, and GRP75 has been linked with resistance to cisplatin in ovarian cancer119.

Targeting GRPs

As the GRPs are crucial factors in the multiple steps of tumorigenesis and often induced in tumours that have developed resistance against conventional therapy, they are attractive targets for drug and vaccine development to combat cancer progression and recurrence. Recently, GRP inhibitors acting at multiple levels have been identified (Figure 3). Importantly, cell surface expression of GRPs primarily in malignant but not normal cells in vivo offers the opportunity for cancer-specific therapy and drug delivery without harming the normal organs. Additionally, because GRPs are upregulated in the cancer microenvironment, their promoters could be of use in the development of gene therapies for the treatment of cancer.

An external file that holds a picture, illustration, etc.
Object name is nihms621506f3.jpg
Summary of agents targeting the GRPs

Stress-induced GRP expression can be suppressed at the transcriptional level by inhibiting transcription factors required for the stress induction of the GRP promoter. Several microRNAs have been identified that suppresses the translation of GRP78 mRNA in cancer cells. Since chaperone function of the GRPs depends on the ATPase catalytic activity, compounds or peptides that bind to their ATP binding domains or alter their ATPase activity are effective in suppressing GRP function. A BAG1 peptide binds to the GRP78 substrate binding domain and inhibits its protein refolding activity. GRP78 can be cleaved specifically by the bacterial toxin SubAB, which renders it non-functional. Cell surface GRP78 can be effectively targeted by specific peptides in conjugated or non-conjugated forms and the human plasminogen factor Kringle 5 (K5). Antibodies against cell surface GRP78 are able to suppress GRP78-mediated oncogenic signaling and induce cancer cell death by multiple mechanisms. On the other hand, the inducible GRP promoter containing the ER stress responsive elements (ERSEs) can be used to direct cytotoxic gene expression in cancer cells. The GRP inhibitors listed have shown a wide range of anti-cancer effects in vitro and in vivo.

Inhibitors of the GRPs

In principle, agents that inhibit the synthesis, stability or activity of the GRPs can simultaneously suppress their function at various cellular locations. The challenge is to minimize toxicity to normal organs. Various heterozygous knockout mouse models revealed that a 50% decrease in GRP78 expression has no effect on normal organs but significantly impedes tumour growth and angiogenesis89 (Table 1). This implies that agents that selectively block the stress induction of GRP78 will affect tumours that require a high level of GRP78 and spare normal organs. Natural compounds with anti-cancer properties that suppress GRP78 induction have been reported, however, they exert pleiotrophic effects5,120. Specific cancers also express microRNAs that can act cooperatively to suppress GRP78 translation (Figure 3) and reverse chemoresistance121. However, specific inhibitors of GRP78 stress induction remain to be identified.

Alternatively, as the GRP78 promoter is highly active in aggressive solid tumours, this offers the opportunity to use the GRP78 promoter to direct expression of suicide genes, immunosuppressors and tumour suppressors in anti-cancer therapy. As proof-of-principle, the Grp78 promoter driving the expression of the herpes simplex kinase suicide gene in a retroviral system results in eradication of sizable tumours122,123. Recently, systemic administration of a dual tumour-targeted phage containing the RGD tumour homing ligand and the Grp78 promoter, showed persistent transgene expression in vitro and significant killing of therapy resistant tumours in vivo124. Likewise, cancer-inducible transgene expression can be directed by the Grp94 promoter in tumours of various origins and cancer-associated macrophages125.

Selective destruction of GRP78 at the protein level might be possible owing to the discovery of a bacterial toxin SubAB which cleaves GRP78 at a single site (L416-L417) in the hinge region connecting the ATPase and the substrate binding domain of the molecule, thereby inactivating it126. Systemic delivery of an engineered fusion protein combining epidermal growth factor (EGF) and SubAB was toxic to EGFR expressing cancer cells in vitro and caused a delay in the growth of human breast, prostate and glioblastoma xenografts in mice127,128. One of these studies128 also showed that modest cleavage of GRP78 in normal mouse liver cells due to EGFR expression did not lead to weight loss, consistent with the findings in genetic models that normal organs including the liver can tolerate partial decrease in GRP78 levels39.

Since the ATPase catalytic activity of GRP78 is necessary for its anti-apoptotic function60, targeting its ATP binding domain can effectively inactivate GRP78 in cancer. Several plant compounds including (-)epigallocatechin gallate (EGCG), honokiol and aspirin (also known as salicylate) directly bind to this domain and inhibit the ATPase activity (Figure 3), and this is associated with the sensitization of cancer cells to chemotoxic agents129-131. Furthermore, an unconjugated peptide derived from the co-chaperone BAG1 binds to the substrate binding domain of GRP78 and inhibits its protein refolding activity, and prostate cancer cells stably expressing this peptide showed reduced growth and apoptosis in xenograft models in a manner dependent on binding to GRP78132. In glioblastoma cells, GRP78 can also be inactivated via acetylation by vorinostat, a deacetylase inhibitor with anti-tumour activity133.

Recently, specific inhibitors against GRP94 function have been identified based on its unique secondary nucleotide binding pocket72,134. One of the compounds, PU-WS13, has been shown to reduce the viability of breast cancer cells expressing high level of cell surface ERBB272 and human multiple myeloma cells53 in vitro. Interestingly, honokiol induces calpain-mediated cleavage GRP94 in human gastric cancer cells, associating with apoptosis and reduction in tumour growth135. GPM1, a chemical that can bind GRP94, suppresses its surface presentation through increased ER retention. This chemical was shown to compromise the immune functions of GRP94 in vivo; however, its efficacy in cancer is not known136. MKT-077, a cationic rhodacyanine dye, binds the nucleotide-binding domain of GRP75, abrogates its interaction with p53 and reactivates the transcriptional and pro-apoptotic activities of p53 in cancer cells, but not in normal cells in vitro137-139. Additionally, virtual screening of a drug database has revealed several small molecule inhibitors that are able to interrupt the p53-GRP75 complex140.

Collectively, these proof-of-concept studies demonstrate that GRP inhibitors can selectively confer toxicity to cancer cells in vitro and in vivo, warranting further development and validation.

Cytotoxic agents targeting cell surface GRP

Preferential expression of GRP78 on the surface of tumour cells in vivo enables specific tumour targeting with minimal harmful effects on normal cells11,13,141. As cell surface GRP78 expression is further detected in some TICs and increased in metastatic and drug resistant tumours6,16,117 and in hypoxic endothelial cells supporting tumour growth68,78, cytotoxic agents against cell surface GRP78 have the potential to target these cells in addition to the primary tumour.

Several synthetic peptides composed of GRP78 binding motifs fused to cell-death inducing peptides or cytotoxic drugs are able to promote apoptosis in cancer cells in vitro, including human prostate, and breast cancer cells, human melanoma, chemotherapy-resistant B lineage acute lymphoblastic leukemia cells and multidrug resistant gastric cells6,142-148 (Figure 3). Furthermore, xenograft and isogenic mouse models were used to validate the efficacy of the peptides in suppressing tumour growth of human prostate and breast cancer, melanoma, as well as bone metastasis with no apparent toxicity6,142,145. The GRP78 binding peptides have been conjugated to nanoparticles or liposomes for more efficient drug delivery145,149, and such agents are able to home to endothelial cells in tumours, suppressing growth and prolonged survival of colon carcinoma bearing mice82. Furthermore, a reconstructed protein containing GRP78 binding peptide and mung bean trypsin inhibitor displays targeted anti-cancer effects both in vitro and in vivo in colorectal cancer150.

Recently, MAb159, a high affinity GRP78 specific mouse monoclonal IgG antibody was identified that triggers endocytosis and degradation of cell surface GRP78, and activates both intrinsic and extrinsic apoptosis17. MAb159 causes cancer cell death and suppresses the growth of colon and lung xenografts, the metastatic growth of human breast and melanoma xenografts and the growth of prostate cancer and leukemia in genetically engineered mouse models, at least in part through inhibition of the PI3K signaling pathway17. MAb159 also synergizes with Irinotecan, a topoisomerase I inhibitor, in suppressing human colon cancer xenograft growth. A humanized MAb159 retains anti-tumour activity with no toxicity in mice and exhibits favorable pharmacokinetics17. In principle, this antibody can also be used as an in vivo imaging agent for selection of patients expressing cell surface GRP78 and to determine whether that predicts disease progression and response to therapy. Another screen yielded a mouse monoclonal IgG antibody targeting the carboxyl-terminal domain of GRP78, C107, which is capable of inducing apoptosis in melanoma cells in vitro and slowing their growth as xenografts in mice151. A human monoclonal IgM antibody (PAT-SM6) isolated from a patient with gastric cancer that can simultaneously bind low density lipoproteins and multiple GRP78 molecules on the surface of tumour cells, induces lipid accumulation and apoptosis in human multiple myeloma cells152,153, and suppresses human melanoma growth both in vitro and in xenografts154. Based on favorable safety profiles in phase I studies, the efficacy of PAT-SM6 is being tested in clinical trials154. It has also been reported that autoantibodies against GRP78 from ovarian cancer patients promote apoptosis and decrease the invasiveness of ovarian cancer cells155. In another study, autoantibodies against GRP78 from prostate cancer patients trigger ER Ca2+ release in human bladder carcinoma cells and increase tissue factor procoagulant activity, implying that blocking cell surface GRP78 signaling could potentially reduce the risk of cancer-related thrombotic events156. As proof-of-principle that cell surface GRP75 may also be amendable to therapy, intra-tumoural and intra-peritoneal injections of an anti-GRP75 antibody results in tumour growth suppression22.

Vaccination strategies

Molecular chaperone preparations from tumours carrying tumour antigens offer a personalized, polyvalent vaccine therapy2,23,157. Although vaccination of lethally irradiated cancer cells expressing autologous secretory GRP94 fusion proteins protected mice from primary tumour growth and metastasis158, vitespen, a GRP94-peptide complex that was purified ex vivo from individual patient's tumour cells showed variable immunogenicity and overall limited efficacy in clinical trials, with clinical responses only in certain patient subsets159-161. Recent studies showed that low dose of GRP94 immunization activated cytotoxic T lymphocytes with some tumour suppression in mice whereas high dose induced Treg proliferation and immune suppression, in a manner dependent on TLR-mediated NF-κB activation162. The use of GRP94 fusion proteins with tumour antigens, depletion of Treg cells, and pooled GRP94 vaccines have all been proposed to enhance the anti-tumour activity of GRP94 immunization163, however challenges remain with these approaches.

GRP170 can form complexes with full-length protein antigens, such as gp100, and increase their presentation to immune cells, thereby augmenting multivalent T-cell-mediated anti-tumour immune responses164. Genetic modification of various poorly immunogenic melanomas to express a secretable form of GRP170 significantly suppressed tumour growth in vivo, and this was associated with increased tumour-infiltrating CD8+ T cells and stimulation of dendritic cells165. GRP170-secreting tumour cells used as a cell-based vaccine is effective in treating established mouse prostate tumours166. Incorporation of a pathogen-associated molecule such as the NF-κB-activating domain of the bacterial flagellin into GRP170 maintains high efficient antigen-holding ability and through pathogen sensing TLR signaling, additionally activates dendritic cells, mounting a superior anti-tumour immune response against the primary tumour and distant metastasis167. The tumour derived secreted form of GRP78 is also able to elicit an anti-tumour immune response in mouse models as a result of activation of cytotoxic T cells168.

Conclusions and perspectives

The GRPs possess functions that are distinct from the HSPs, impacting both the tumour cells and the tumour microenvironment. As the stress induction of GRPs could be a major contributor for tumorigenesis and therapeutic resistance, their specific inhibitors and targeting agents hold great therapeutic promise. Their clinical efficacies, as well as large GRPs as vaccines, warrant vigorous testing in the clinical setting. However, answers to key issues on basic mechanisms, such as how the stress-induced relocalization of the GRPs from the ER to the cell surface and other organelles occurs, what their interactive partners are and the mechanisms of signaling, as well as the utility of GRPs as prognostic markers and companion imaging agents for precision cancer care will greatly advance the understanding of GRP biology and their applications in cancer.

Key points NRC-12-276V4

  1. The glucose regulated proteins (GRP78, GRP94, GRP170 and GRP75) are members of the heat shock protein family primarily residing in the endoplasmic reticulum (ER) or the mitochondria and they are induced at the transcriptional level upon ER stress.
  2. As molecular chaperones, the GRPs regulate protein quality control and degradation, with GRP78 serving additionally as a pivotal regulator of the unfolded protein response and the apoptotic machinery associated with the ER.
  3. The GRPs can be actively translocated to other cellular locations and secreted, and assume additional functions that control cellular signaling, proliferation, invasion, apoptosis, inflammation and immunity, which have major implications in cancer progression and therapeutic resistance.
  4. Specific roles of GRPs in development, tumorigenesis, metastasis and angiogenesis have been identified in vitro and validated in genetically engineered mouse models.
  5. GRP overexpression is widely reported in many human cancers and associated with aggressive properties, suggesting potential prognostic value and that interfering with their production or activities in those tumors might provide new approaches for anti-cancer treatment.
  6. The discovery that cell surface GRP78 is preferably expressed in cancer and stressed endothelial cells leads to the development of therapeutic agents specifically targeting cell surface GRP78 capable of inducing cancer cell apoptosis and suppressing tumorigenesis with minimal toxicity.
  7. While the GRPs are attractive targets for drug development, they can also serve as mediators for cancer specific drug delivery, transcriptional targeting of cancer and vaccine development.
  8. The large chaperone GRP170 with superior property in presentation of full-length protein antigens opens up a new vaccine platform to augment anti-tumor immune responses.

Supplementary Material

Suppl Table 1

Suppl Table 2

Acknowledgments

This work is supported in part by NIH grants CA027607 and AG034906. I thank Drs. Larry Hightower, Zihai Li, John Subjeck, Xiang-Yang Wang, and John Luk for helpful discussions, Lee lab members in particular, Jieli Shen and Yi Zhang, and Esther and Sarah Olivo for assistance.

Footnotes

Competing Interests Statement: The author declares that she has no competing financial interests.

References

1. Ni M, Lee AS. ER chaperones in mammalian development and human diseases. FEBS Lett. 2007;581:3641–3651. [PMC free article] [PubMed] [Google Scholar]
2. Wang XY, Subjeck JR. High molecular weight stress proteins: Identification, cloning and utilisation in cancer immunotherapy. Int J Hyperthermia. 2013;29:364–375. [PMC free article] [PubMed] [Google Scholar]
3. Wadhwa R, Taira K, Kaul SC. An Hsp70 family chaperone, mortalin/mthsp70/PBP74/Grp75: what, when, and where? Cell Stress Chaperones. 2002;7:309–316. [PMC free article] [PubMed] [Google Scholar]
4. Marzec M, Eletto D, Argon Y. GRP94: An HSP90-like protein specialized for protein folding and quality control in the endoplasmic reticulum. Biochimica et Biophysica Acta (BBA) - Mol Cell Res. 2012;1823:774–787. [PMC free article] [PubMed] [Google Scholar]
5. Lee AS. GRP78 induction in cancer: therapeutic and prognostic implications. Cancer Res. 2007;67:3496–3499. [PubMed] [Google Scholar]
6. Miao YR, et al. Inhibition of established micrometastases by targeted drug delivery via cell surface-associated GRP78. Clin Cancer Res. 2013;19:2107–2116. [PMC free article] [PubMed] [Google Scholar]
7. Wang M, Wey S, Zhang Y, Ye R, Lee AS. Role of the unfolded protein response regulator GRP78/BiP in development, cancer, and neurological disorders. Antioxid Redox Signal. 2009;11:2307–2316. [PMC free article] [PubMed] [Google Scholar]
8. Kusaczuk M, Cechowska-Pasko M. Molecular chaperone ORP150 in ER stress-related diseases. Curr Pharm Des. 2013;19:2807–2818. [PubMed] [Google Scholar]
9. Behnke J, Hendershot LM. The large Hsp70 Grp170 binds to unfolded protein substrates in vivo with a regulation distinct from conventional Hsp70s. J Biol Chem. 2013 Epub ahead of print. [PMC free article] [PubMed] [Google Scholar]
10. Wadhwa R, et al. Hsp70 family member, mot-2/mthsp70/GRP75, binds to the cytoplasmic sequestration domain of the p53 protein. Exp Cell Res. 2002;274:246–253. [PubMed] [Google Scholar]
11. Ni M, Zhang Y, Lee AS. Beyond the endoplasmic reticulum: atypical GRP78 in cell viability, signalling and therapeutic targeting. Biochem J. 2011;434:181–188. [PMC free article] [PubMed] [Google Scholar]
12. Gonzalez-Gronow M, Selim MA, Papalas J, Pizzo SV. GRP78: a multifunctional receptor on the cell surface. Antioxid Redox Signal. 2009;11:2299–2306. [PubMed] [Google Scholar]
13. Sato M, Yao VJ, Arap W, Pasqualini R. GRP78 signaling hub a receptor for targeted tumor therapy. Adv Genet. 2010;69:97–114. [PubMed] [Google Scholar]
14. Gray PC, Vale W. Cripto/GRP78 modulation of the TGF-beta pathway in development and oncogenesis. FEBS Lett. 2012;586:1836–1845. [PMC free article] [PubMed] [Google Scholar]
15. Zhang Y, Liu R, Ni M, Gill P, Lee AS. Cell surface relocalization of the endoplasmic reticulum chaperone and unfolded protein response regulator GRP78/BiP. J Biol Chem. 2010;285:15065–15075. [PMC free article] [PubMed] [Google Scholar]
16. Zhang Y, et al. Cancer cells resistant to therapy promote cell surface relocalization of GRP78 which complexes with PI3K and enhances PI(3,4,5)P3 production. PLoS One. 2013;8:e80071. [PMC free article] [PubMed] [Google Scholar]
17. Liu R, et al. Monoclonal antibody against cell surface GRP78 as a novel agent in suppressing PI3K/AKT signaling, tumor growth and metastasis. Clin Cancer Res. 2013;19:6802–6811. [PMC free article] [PubMed] [Google Scholar]
18. Pfaffenbach KT, et al. GRP78/BiP is a novel downstream target of IGF-1 receptor mediated signaling. J Cell Physiol. 2012;227:3803–3811. [PMC free article] [PubMed] [Google Scholar]
19. Gray MJ, et al. AKT inhibition mitigates GRP78 (glucose-regulated protein) expression and contribution to chemoresistance in endometrial cancers. Int J Cancer. 2013;133:21–30. [PMC free article] [PubMed] [Google Scholar]
20. Luo B, Lee AS. The critical roles of endoplasmic reticulum chaperones and unfolded protein response in tumorigenesis and anticancer therapies. Oncogene. 2013;32:805–818. [PMC free article] [PubMed] [Google Scholar]
21. Li J, Lee AS. Stress induction of GRP78/BiP and its role in cancer. Curr Mol Med. 2006;6:45–54. [PubMed] [Google Scholar]
22. Yun CO, Wadhwa R. In: Mortalin Biology: Life, Stress and Death: Life, Stress and Death. Kaul SC, Wadhwa R, editors. Chapter 18. Springer; Heidelberg: 2012. [Google Scholar]
23. Srivastava PK. Identification of chaperones as essential components of the tumor rejection moieties of cancers. Cancer Immun. 2012;12:5. [PMC free article] [PubMed] [Google Scholar]
24. Mori K. Tripartite management of unfolded proteins in the endoplasmic reticulum. Cell. 2000;101:451–454. [PubMed] [Google Scholar]
25. Chang SC, Erwin AE, Lee AS. Glucose-regulated protein (GRP94 and GRP78) genes share common regulatory domains and are coordinately regulated by common trans-acting factors. Mol Cell Biol. 1989;9:2153–2162. [PMC free article] [PubMed] [Google Scholar]
26. Ma Y, Hendershot LM. The role of the unfolded protein response in tumour development: friend or foe? Nat Rev Cancer. 2004;4:966–977. [PubMed] [Google Scholar]
27. Wu J, Kaufman RJ. From acute ER stress to physiological roles of the unfolded protein response. Cell Death Differ. 2006;13:374–384. [PubMed] [Google Scholar]
28. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007;8:519–529. [PubMed] [Google Scholar]
29. Hetz C. The unfolded protein response: controlling cell fate decisions under ER stress and beyond. Nat Rev Mol Cell Biol. 2012;13:89–102. [PubMed] [Google Scholar]
30. Luo S, Baumeister P, Yang S, Abcouwer SF, Lee AS. Induction of Grp78/BiP by translational block: activation of the Grp78 promoter by ATF4 through an upstream ATF/CRE site independent of the endoplasmic reticulum stress elements. J Biol Chem. 2003;278:37375–37385. [PubMed] [Google Scholar]
31. Lee K, et al. IRE1-mediated unconventional mRNA splicing and S2P-mediated ATF6 cleavage merge to regulate XBP1 in signaling the unfolded protein response. Genes Dev. 2002;16:452–466. [PMC free article] [PubMed] [Google Scholar]
32. Li J, et al. The unfolded protein response regulator GRP78/BiP is required for endoplasmic reticulum integrity and stress-induced autophagy in mammalian cells. Cell Death Differ. 2008;15:1460–1471. [PMC free article] [PubMed] [Google Scholar]
33. Wang M, et al. Essential role of the unfolded protein response regulator GRP78/BiP in protection from neuronal apoptosis. Cell Death Differ. 2010;17:488–498. [PMC free article] [PubMed] [Google Scholar]
34. Wey S, Luo B, Lee AS. Acute inducible ablation of GRP78 reveals its role in hematopoietic stem cell survival, lymphogenesis and regulation of stress signaling. PLoS One. 2012;7:e39047. [PMC free article] [PubMed] [Google Scholar]
35. Heijmans J, et al. ER stress causes rapid loss of intestinal epithelial stemness through activation of the unfolded protein response. Cell Rep. 2013;3:1128–1139. [PubMed] [Google Scholar]
36. Mathew R, et al. Autophagy suppresses tumorigenesis through elimination of p62. Cell. 2009;137:1062–1075. [PMC free article] [PubMed] [Google Scholar]
37. Baumeister P, Dong D, Fu Y, Lee AS. Transcriptional induction of GRP78/BiP by histone deacetylase inhibitors and resistance to histone deacetylase inhibitor-induced apoptosis. Mol Cancer Ther. 2009;8:1086–1094. [PMC free article] [PubMed] [Google Scholar]
38. Pyrko P, Schonthal AH, Hofman FM, Chen TC, Lee AS. The unfolded protein response regulator GRP78/BiP as a novel target for increasing chemosensitivity in malignant gliomas. Cancer Res. 2007;67:9809–9816. [PubMed] [Google Scholar]
39. Dong D, et al. Critical role of the stress chaperone GRP78/BiP in tumor proliferation, survival, and tumor angiogenesis in transgene-induced mammary tumor development. Cancer Res. 2008;68:498–505. [PubMed] [Google Scholar]
40. Verras M, Papandreou I, Lim AL, Denko NC. Tumor hypoxia blocks Wnt processing and secretion through the induction of endoplasmic reticulum stress. Mol Cell Biol. 2008;28:7212–7224. [PMC free article] [PubMed] [Google Scholar]
41. Misra UK, Gonzalez-Gronow M, Gawdi G, Pizzo SV. The role of MTJ-1 in cell surface translocation of GRP78, a receptor for alpha 2-macroglobulin-dependent signaling. J Immunol. 2005;174:2092–2097. [PubMed] [Google Scholar]
42. Burikhanov R, et al. The tumor suppressor Par-4 activates an extrinsic pathway for apoptosis. Cell. 2009;138:377–388. [PMC free article] [PubMed] [Google Scholar]
43. Misra UK, et al. The role of Grp 78 in alpha 2-macroglobulin-induced signal transduction. Evidence from RNA interference that the low density lipoprotein receptor-related protein is associated with, but not necessary for, GRP 78-mediated signal transduction. J Biol Chem. 2002;277:42082–42087. [PubMed] [Google Scholar]
44. Misra UK, Pizzo SV. Receptor-recognized alpha(2)-macroglobulin binds to cell surface-associated GRP78 and activates mTORC1 and mTORC2 signaling in prostate cancer cells. PLoS One. 2012;7:e51735. [PMC free article] [PubMed] [Google Scholar]
45. de Ridder GG, Gonzalez-Gronow M, Ray R, Pizzo SV. Autoantibodies against cell surface GRP78 promote tumor growth in a murine model of melanoma. Melanoma Res. 2011;21:35–43. [PMC free article] [PubMed] [Google Scholar]
46. Salmena L, Carracedo A, Pandolfi PP. Tenets of PTEN tumor suppression. Cell. 2008;133:403–414. [PubMed] [Google Scholar]
47. Fu Y, et al. Pten null prostate tumorigenesis and AKT activation are blocked by targeted knockout of ER chaperone GRP78/BiP in prostate epithelium. Proc Natl Acad Sci USA. 2008;105:19444–19449. [PMC free article] [PubMed] [Google Scholar]
48. Wey S, et al. Inducible knockout of GRP78/BiP in the hematopoietic system suppresses Pten-null leukemogenesis and AKT oncogenic signaling. Blood. 2012;119:817–825. [PMC free article] [PubMed] [Google Scholar]
49. Shani G, et al. GRP78 and Cripto form a complex at the cell surface and collaborate to inhibit transforming growth factor beta signaling and enhance cell growth. Mol Cell Biol. 2008;28:666–677. [PMC free article] [PubMed] [Google Scholar]
50. Kelber JA, et al. Blockade of Cripto binding to cell surface GRP78 inhibits oncogenic Cripto signaling via MAPK/PI3K and Smad2/3 pathways. Oncogene. 2009;28:2324–2336. [PMC free article] [PubMed] [Google Scholar]
51. Wanderling S, et al. GRP94 is essential for mesoderm induction and muscle development because it regulates insulin-like growth factor secretion. Mol Biol Cell. 2007;18:3764–3775. [PMC free article] [PubMed] [Google Scholar]
52. Liu B, et al. Essential roles of grp94 in gut homeostasis via chaperoning canonical Wnt pathway. Proc Natl Acad Sci USA. 2013;110:6877–6882. [PMC free article] [PubMed] [Google Scholar]
53. Hua Y, et al. Molecular chaperone gp96 is a novel therapeutic target of multiple myeloma. Clin Cancer Res. 2013;19:6242–6251. [PMC free article] [PubMed] [Google Scholar]
54. Morales C, et al. Immune chaperone gp96 drives the contributions of macrophages to inflammatory colon tumorigenesis. Cancer Res. 2014;74:446–459. [PMC free article] [PubMed] [Google Scholar]
55. Dejeans N, et al. Overexpression of GRP94 in breast cancer cells resistant to oxidative stress promotes high levels of cancer cell proliferation and migration: implications for tumor recurrence. Free Radic Biol Med. 2012;52:993–1002. [PubMed] [Google Scholar]
56. Kaul SC, et al. Malignant transformation of NIH3T3 cells by overexpression of mot-2 protein. Oncogene. 1998;17:907–911. [PubMed] [Google Scholar]
57. Wadhwa R, et al. Inactivation of tumor suppressor p53 by mot-2, a hsp70 family member. J Biol Chem. 1998;273:29586–29591. [PubMed] [Google Scholar]
58. Mizukoshi E, et al. Fibroblast growth factor-1 interacts with the glucose-regulated protein GRP75/mortalin. Biochem J. 1999;343 Pt 2:461–466. [PMC free article] [PubMed] [Google Scholar]
59. Rao RV, et al. Coupling endoplasmic reticulum stress to the cell death program: role of the ER chaperone GRP78. FEBS Lett. 2002;514:122–128. [PMC free article] [PubMed] [Google Scholar]
60. Reddy RK, et al. Endoplasmic reticulum chaperone protein GRP78 protects cells from apoptosis induced by topoisomerase inhibitors: role of ATP binding site in suppression of caspase-7 activation. J Biol Chem. 2003;278:20915–20924. [PubMed] [Google Scholar]
61. Fu Y, Li J, Lee AS. GRP78/BiP inhibits endoplasmic reticulum BIK and protects human breast cancer cells against estrogen-starvation induced apoptosis. Cancer Res. 2007;67:3734–3740. [PubMed] [Google Scholar]
62. Zhou H, Zhang Y, Fu Y, Chan L, Lee AS. Novel mechanism of anti-apoptotic function of 78-kDa glucose-regulated protein (GRP78): endocrine resistance factor in breast cancer, through release of B-cell lymphoma 2 (BCL-2) from BCL-2-interacting killer (BIK) J Biol Chem. 2011;286:25687–25696. [PMC free article] [PubMed] [Google Scholar]
63. Ni M, Zhou H, Wey S, Baumeister P, Lee AS. Regulation of PERK signaling and leukemic cell survival by a novel cytosolic isoform of the UPR regulator GRP78/BiP. PLoS One. 2009;4:e6868. [PMC free article] [PubMed] [Google Scholar]
64. Shu CW, et al. GRP78 and Raf-1 cooperatively confer resistance to endoplasmic reticulum stress-induced apoptosis. J Cell Physiol. 2008;215:627–635. [PubMed] [Google Scholar]
65. Luo S, Mao C, Lee B, Lee AS. GRP78/BiP is required for cell proliferation and protecting the inner cell mass from apoptosis during early mouse embryonic development. Mol Cell Biol. 2006;26:5688–5697. [PMC free article] [PubMed] [Google Scholar]
66. Zhu G, et al. GRP78 plays an essential role in adipogenesis and postnatal growth in mice. FASEB J. 2013;27:955–964. [PMC free article] [PubMed] [Google Scholar]
67. Misra UK, Deedwania R, Pizzo SV. Activation and cross-talk between Akt, NF-{kappa}B, and unfolded protein response signaling in 1-LN prostate cancer cells consequent to ligation of cell surface-associated GRP78. J Biol Chem. 2006;281:13694–13707. [PubMed] [Google Scholar]
68. Davidson DJ, et al. Kringle 5 of human plasminogen induces apoptosis of endothelial and tumor cells through surface-expressed glucose-regulated protein 78. Cancer Res. 2005;65:4663–4672. [PubMed] [Google Scholar]
69. Martin-Perez R, Niwa M, Lopez-Rivas A. ER stress sensitizes cells to TRAIL through down-regulation of FLIP and Mcl-1 and PERK-dependent up-regulation of TRAIL-R2. Apoptosis. 2012;17:349–363. [PMC free article] [PubMed] [Google Scholar]
70. Cook KL, et al. Glucose-regulated protein 78 controls cross-talk between apoptosis and autophagy to determine antiestrogen responsiveness. Cancer Res. 2012;72:3337–3349. [PMC free article] [PubMed] [Google Scholar]
71. Reddy RK, Lu J, Lee AS. The endoplasmic reticulum chaperone glycoprotein GRP94 with Ca2+-binding and antiapoptotic properties is a novel proteolytic target of calpain during etoposide-induced apoptosis. J Biol Chem. 1999;274:28476–28483. [PubMed] [Google Scholar]
72. Patel PD, et al. Paralog-selective Hsp90 inhibitors define tumor-specific regulation of HER2. Nat Chem Biol. 2013;9:677–684. [PMC free article] [PubMed] [Google Scholar]
73. Namba T, et al. Up-regulation of 150-kDa oxygen-regulated protein by celecoxib in human gastric carcinoma cells. Mol Pharmacol. 2007;71:860–870. [PubMed] [Google Scholar]
74. Gao YY, et al. Implication of oxygen-regulated protein 150 (ORP150) in apoptosis induced by proteasome inhibitors in human thyroid cancer cells. J Clin Endocrinol Metab. 2010;95:E319–326. [PubMed] [Google Scholar]
75. Sanson M, et al. Oxidized low-density lipoproteins trigger endoplasmic reticulum stress in vascular cells: prevention by oxygen-regulated protein 150 expression. Circ Res. 2009;104:328–336. [PubMed] [Google Scholar]
76. Sanson M, et al. Oxygen-regulated protein-150 prevents calcium homeostasis deregulation and apoptosis induced by oxidized LDL in vascular cells. Cell Death Differ. 2008;15:1255–1265. [PubMed] [Google Scholar]
77. Virrey JJ, et al. Stress chaperone GRP78/BiP confers chemoresistance to tumor-associated endothelial cells. Mol Cancer Res. 2008;6:1268–1275. [PMC free article] [PubMed] [Google Scholar]
78. McFarland BC, et al. Plasminogen Kringle 5 induces apoptosis of brain microvessel endothelial cells: sensitization by radiation and requirement for GRP78 and LRP1. Cancer Res. 2009;69:5537–5545. [PMC free article] [PubMed] [Google Scholar]
79. Bhattacharjee G, et al. Regulation of tissue factor--mediated initiation of the coagulation cascade by cell surface grp78. Arterioscler Thromb Vasc Biol. 2005;25:1737–1743. [PubMed] [Google Scholar]
80. Philippova M, et al. Identification of proteins associating with glycosylphosphatidylinositol- anchored T-cadherin on the surface of vascular endothelial cells: role for Grp78/BiP in T-cadherin-dependent cell survival. Mol Cell Biol. 2008;28:4004–4017. [PMC free article] [PubMed] [Google Scholar]
81. Gonzalez-Gronow M, et al. Plasminogen structural domains exhibit different functions when associated with cell surface GRP78 or the voltage-dependent anion channel. J Biol Chem. 2007;282:32811–32820. [PubMed] [Google Scholar]
82. Katanasaka Y, et al. Cancer antineovascular therapy with liposome drug delivery systems targeted to BiP/GRP78. Int J Cancer. 2010;127:2685–2698. [PubMed] [Google Scholar]
83. Ozawa K, et al. Regulation of tumor angiogenesis by oxygen-regulated protein 150, an inducible endoplasmic reticulum chaperone. Cancer Res. 2001;61:4206–4213. [PubMed] [Google Scholar]
84. Fu Y, Lee AS. Glucose regulated proteins in cancer progression, drug resistance and immunotherapy. Cancer Biol Ther. 2006;5:741–744. [PubMed] [Google Scholar]
85. Sun Q, et al. Expressions of GRP78 and Bax associate with differentiation, metastasis, and apoptosis in non-small cell lung cancer. Mol Biol Rep. 2012;39:6753–6761. [PubMed] [Google Scholar]
86. Daneshmand S, et al. Glucose-regulated protein GRP78 is up-regulated in prostate cancer and correlates with recurrence and survival. Hum Pathol. 2007;38:1547–1552. [PubMed] [Google Scholar]
87. Zhang J, et al. Association of elevated GRP78 expression with increased lymph node metastasis and poor prognosis in patients with gastric cancer. Clin Exp Metastasis. 2006;23:401–410. [PubMed] [Google Scholar]
88. Li Z, et al. Cell-surface GRP78 facilitates colorectal cancer cell migration and invasion. Int J Biochem Cell Biol. 2013;45:987–994. [PubMed] [Google Scholar]
89. Dong D, et al. A critical role for GRP78/BiP in the tumor microenvironment for neovascularization during tumor growth and metastasis. Cancer Res. 2011;71:2848–2857. [PMC free article] [PubMed] [Google Scholar]
90. Misra UK, Deedwania R, Pizzo SV. Binding of activated alpha2-macroglobulin to its cell surface receptor GRP78 in 1-LN prostate cancer cells regulates PAK-2-dependent activation of LIMK. J Biol Chem. 2005;280:26278–26286. [PMC free article] [PubMed] [Google Scholar]
91. Zheng HC, et al. Overexpression of GRP78 and GRP94 are markers for aggressive behavior and poor prognosis in gastric carcinomas. Hum Pathol. 2008;39:1042–1049. [PubMed] [Google Scholar]
92. Hong F, Liu B, Chiosis G, Gewirth DT, Li Z. alpha7 helix region of alphaI domain is crucial for integrin binding to endoplasmic reticulum chaperone gp96: a potential therapeutic target for cancer metastasis. J Biol Chem. 2013;288:18243–18248. [PMC free article] [PubMed] [Google Scholar]
93. Yi X, et al. Association of mortalin (HSPA9) with liver cancer metastasis and prediction for early tumor recurrence. Mol Cell Proteomics. 2008;7:315–325. [PubMed] [Google Scholar]
94. Stojadinovic A, et al. HYOU1/Orp150 expression in breast cancer. Med Sci Monit. 2007;13:BR231–239. [PubMed] [Google Scholar]
95. Mahadevan NR, et al. Transmission of endoplasmic reticulum stress and pro-inflammation from tumor cells to myeloid cells. Proc Natl Acad Sci USA. 2011;108:6561–6566. [PMC free article] [PubMed] [Google Scholar]
96. Li Z, Li Z. Glucose regulated protein 78: A critical link between tumor microenvironment and cancer hallmarks. Biochim Biophys Acta. 2012;1826:13–22. [PubMed] [Google Scholar]
97. Hori O, et al. Exposure of astrocytes to hypoxia/reoxygenation enhances expression of glucose-regulated protein 78 facilitating astrocyte release of the neuroprotective cytokine interleukin 6. J Neurochem. 1996;66:973–979. [PubMed] [Google Scholar]
98. Jamora C, Dennert G, Lee AS. Inhibition of tumor progression by suppression of stress protein GRP78/BiP induction in fibrosarcoma B/C10ME. Proc Natl Acad Sci USA. 1996;93:7690–7694. [PMC free article] [PubMed] [Google Scholar]
99. Triantafilou M, Fradelizi D, Triantafilou K. Major histocompatibility class one molecule associates with glucose regulated protein (GRP) 78 on the cell surface. Hum Immunol. 2001;62:764–770. [PubMed] [Google Scholar]
100. Misra UK, Chu CT, Rubenstein DS, Gawdi G, Pizzo SV. Receptor-recognized alpha 2-macroglobulin-methylamine elevates intracellular calcium, inositol phosphates and cyclic AMP in murine peritoneal macrophages. Biochem J. 1993;290(Pt 3):885–891. [PMC free article] [PubMed] [Google Scholar]
101. Misra UK, Pizzo SV. Evidence for a pro-proliferative feedback loop in prostate cancer: the role of Epac1 and COX-2-dependent pathways. PLoS One. 2013;8:e63150. [PMC free article] [PubMed] [Google Scholar]
102. Oida T, Weiner HL. TGF-beta induces surface LAP expression on murine CD4 T cells independent of Foxp3 induction. PLoS One. 2010;5:e15523. [PMC free article] [PubMed] [Google Scholar]
103. Corrigall VM, Vittecoq O, Panayi GS. Binding immunoglobulin protein-treated peripheral blood monocyte-derived dendritic cells are refractory to maturation and induce regulatory T-cell development. Immunology. 2009;128:218–226. [PMC free article] [PubMed] [Google Scholar]
104. Yang Y, et al. Heat shock protein gp96 is a master chaperone for toll-like receptors and is important in the innate function of macrophages. Immunity. 2007;26:215–226. [PMC free article] [PubMed] [Google Scholar]
105. Zhang J, et al. PTEN maintains haematopoietic stem cells and acts in lineage choice and leukaemia prevention. Nature. 2006;441:518–522. [PubMed] [Google Scholar]
106. Luo B, et al. The endoplasmic reticulum chaperone protein GRP94 is required for maintaining hematopoietic stem cell interactions with the adult bone marrow niche. PLoS One. 2011;6:e20364. [PMC free article] [PubMed] [Google Scholar]
107. Luo B, Tseng CC, Adams GB, Lee AS. Deficiency of GRP94 in the hematopoietic system alters proliferation regulators in hematopoietic stem cells. Stem Cells Dev. 2013;22:3062–3073. [PMC free article] [PubMed] [Google Scholar]
108. Wu MJ, et al. Elimination of head and neck cancer initiating cells through targeting glucose regulated protein78 signaling. Mol Cancer. 2010;9:283. [PMC free article] [PubMed] [Google Scholar]
109. Chiu CC, et al. Grp78 as a therapeutic target for refractory head-neck cancer with CD24CD44 stemness phenotype. Cancer Gene Ther. 2013;20:606–615. [PubMed] [Google Scholar]
110. Galicia VA, et al. Expansion of hepatic tumor progenitor cells in Pten-null mice requires liver injury and is reversed by loss of AKT2. Gastroenterology. 2010;139:2170–2182. [PMC free article] [PubMed] [Google Scholar]
111. Chen WT, et al. GRP78 as a regulator of liver steatosis and cancer progression mediated by loss of the tumor suppressor PTEN. Oncogene. 2013 Epub ahead of print. [PMC free article] [PubMed] [Google Scholar]
112. Ji C, et al. Liver-specific loss of glucose-regulated protein 78 perturbs the unfolded protein response and exacerbates a spectrum of liver diseases in mice. Hepatology. 2011;54:229–239. [PMC free article] [PubMed] [Google Scholar]
113. Chen WT, et al. Liver-specific knockout of GRP94 in mice disrupts cell adhesion, activates liver progenitor cells, and accelerates liver tumorigenesis. Hepatology. 2013 Epub ahead of print. [PMC free article] [PubMed] [Google Scholar]
114. Cook KL, Clarke PA, Clarke R. Targeting GRP78 and antiestrogen resistance in breast cancer. Future Med Chem. 2013;5:1047–1057. [PubMed] [Google Scholar]
115. Gomer CJ, et al. Photodynamic therapy-mediated oxidative stress can induce expression of heat shock proteins. Cancer Res. 1996;56:2355–2360. [PubMed] [Google Scholar]
116. Li B, Cheng XL, Yang YP, Li ZQ. GRP78 mediates radiation resistance of a stem cell-like subpopulation within the MCF-7 breast cancer cell line. Oncol Rep. 2013;30:2119–2126. [PubMed] [Google Scholar]
117. Roller C, Maddalo D. The molecular chaperone GRP78/BiP in the development of chemoresistance: mechanism and possible treatment. Front Pharmacol. 2013;4:10. [PMC free article] [PubMed] [Google Scholar]
118. Kern J, et al. GRP-78 secreted by tumor cells blocks the antiangiogenic activity of bortezomib. Blood. 2009;114:3960–3967. [PubMed] [Google Scholar]
119. Yang L, Li H, Jiang Y, Zuo J, Liu W. Inhibition of mortalin expression reverses cisplatin resistance and attenuates growth of ovarian cancer cells. Cancer Lett. 2013;336:213–221. [PubMed] [Google Scholar]
120. Thomas S, et al. Repositioning of Verrucosidin, a purported inhibitor of chaperone protein GRP78, as an inhibitor of mitochondrial electron transport chain complex I. PLoS One. 2013;8:e65695. [PMC free article] [PubMed] [Google Scholar]
121. Su SF, et al. miR-30d, miR-181a and miR-199a-5p cooperatively suppress the endoplasmic reticulum chaperone and signaling regulator GRP78 in cancer. Oncogene. 2013;32:4694–4701. [PMC free article] [PubMed] [Google Scholar]
122. Dong D, et al. Spontaneous and controllable activation of suicide gene expression driven by the stress-inducible grp78 promoter resulting in eradication of sizable human tumors. Hum Gene Ther. 2004;15:553–561. [PubMed] [Google Scholar]
123. Azatian A, et al. Effectiveness of HSV-tk suicide gene therapy driven by the Grp78 stress-inducible promoter in esophagogastric junction and gastric adenocarcinomas. J Gastrointest Surg. 2009;13:1044–1051. [PubMed] [Google Scholar]
124. Kia A, et al. Dual systemic tumor targeting with ligand-directed phage and Grp78 promoter induces tumor regression. Mol Cancer Ther. 2012;11:2566–2577. [PMC free article] [PubMed] [Google Scholar]
125. Reddy RK, et al. Cancer-inducible transgene expression by the Grp94 promoter: spontaneous activation in tumors of various origins and cancer-associated macrophages. Cancer Res. 2002;62:7207–7212. [PubMed] [Google Scholar]
126. Paton AW, et al. AB5 subtilase cytotoxin inactivates the endoplasmic reticulum chaperone BiP. Nature. 2006;443:548–552. [PubMed] [Google Scholar]
127. Backer JM, et al. Chaperone-targeting cytotoxin and endoplasmic reticulum stress-inducing drug synergize to kill cancer cells. Neoplasia. 2009;11:1165–1173. [PMC free article] [PubMed] [Google Scholar]
128. 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. [PMC free article] [PubMed] [Google Scholar]
129. Ermakova SP, et al. (-)-Epigallocatechin gallate overcomes resistance to etoposide-induced cell death by targeting the molecular chaperone glucose-regulated protein 78. Cancer Res. 2006;66:9260–9269. [PubMed] [Google Scholar]
130. Martin S, et al. Inducing apoptosis of cancer cells using small-molecule plant compounds that bind to GRP78. Br J Cancer. 2013;109:433–443. [PMC free article] [PubMed] [Google Scholar]
131. Deng WG, Ruan KH, Du M, Saunders MA, Wu KK. Aspirin and salicylate bind to immunoglobulin heavy chain binding protein (BiP) and inhibit its ATPase activity in human fibroblasts. FASEB J. 2001;15:2463–2470. [PubMed] [Google Scholar]
132. Maddalo D, et al. A peptidic unconjugated GRP78/BiP ligand modulates the unfolded protein response and induces prostate cancer cell death. PLoS One. 2012;7:e45690. [PMC free article] [PubMed] [Google Scholar]
133. Kahali S, et al. Activation of the unfolded protein response contributes toward the antitumor activity of vorinostat. Neoplasia. 2010;12:80–86. [PMC free article] [PubMed] [Google Scholar]
134. Duerfeldt AS, et al. Development of a Grp94 inhibitor. J Am Chem Soc. 2012;134:9796–9804. [PMC free article] [PubMed] [Google Scholar]
135. Sheu ML, Liu SH, Lan KH. Honokiol induces calpain-mediated glucose-regulated protein-94 cleavage and apoptosis in human gastric cancer cells and reduces tumor growth. PLoS One. 2007;2:e1096. [PMC free article] [PubMed] [Google Scholar]
136. Han JM, et al. Identification of gp96 as a novel target for treatment of autoimmune disease in mice. PLoS One. 2010;5:e9792. [PMC free article] [PubMed] [Google Scholar]
137. Kaul SC, Aida S, Yaguchi T, Kaur K, Wadhwa R. Activation of wild type p53 function by its mortalin-binding, cytoplasmically localizing carboxyl terminus peptides. J Biol Chem. 2005;280:39373–39379. [PubMed] [Google Scholar]
138. Wadhwa R, et al. Selective toxicity of MKT-077 to cancer cells is mediated by its binding to the hsp70 family protein mot-2 and reactivation of p53 function. Cancer Res. 2000;60:6818–6821. [PubMed] [Google Scholar]
139. Lu WJ, et al. Mortalin-p53 interaction in cancer cells is stress dependent and constitutes a selective target for cancer therapy. Cell Death Differ. 2011;18:1046–1056. [PMC free article] [PubMed] [Google Scholar]
140. Utomo DH, Widodo N, Rifa'i M. Identifications small molecules inhibitor of p53-mortalin complex for cancer drug using virtual screening. Bioinformation. 2012;8:426–429. [PMC free article] [PubMed] [Google Scholar]
141. Jakobsen CG, Rasmussen N, Laenkholm AV, Ditzel HJ. Phage display derived human monoclonal antibodies isolated by binding to the surface of live primary breast cancer cells recognize GRP78. Cancer Res. 2007;67:9507–9517. [PubMed] [Google Scholar]
142. Arap MA, et al. Cell surface expression of the stress response chaperone GRP78 enables tumor targeting by circulating ligands. Cancer Cell. 2004;6:275–284. [PubMed] [Google Scholar]
143. Kim Y, et al. Targeting heat shock proteins on cancer cells: selection, characterization, and cell-penetrating properties of a peptidic GRP78 ligand. Biochemistry. 2006;45:9434–9444. [PubMed] [Google Scholar]
144. Liu Y, et al. Mechanistic studies of a peptidic GRP78 ligand for cancer cell-specific drug delivery. Mol Pharm. 2007;4:435–447. [PMC free article] [PubMed] [Google Scholar]
145. Passarella RJ, et al. Targeted nanoparticles that deliver a sustained, specific release of Paclitaxel to irradiated tumors. Cancer Res. 2010;70:4550–4559. [PMC free article] [PubMed] [Google Scholar]
146. Larson N, Ray A, Malugin A, Pike DB, Ghandehari H. HPMA copolymer-aminohexylgeldanamycin conjugates targeting cell surface expressed GRP78 in prostate cancer. Pharm Res. 2010;27:2683–2693. [PubMed] [Google Scholar]
147. Uckun FM, et al. Inducing apoptosis in chemotherapy-resistant B-lineage acute lymphoblastic leukaemia cells by targeting HSPA5, a master regulator of the anti-apoptotic unfolded protein response signalling network. Br J Haematol. 2011;153:741–752. [PubMed] [Google Scholar]
148. Kang J, et al. A peptide derived from phage display library exhibits anti-tumor activity by targeting GRP78 in gastric cancer multidrug resistance cells. Cancer Lett. 2013;339:247–259. [PubMed] [Google Scholar]
149. Delie F, Petignat P, Cohen M. GRP78-targeted nanotherapy against castrate-resistant prostate cancer cells expressing membrane GRP78. Target Oncol. 2013;8:225–230. [PubMed] [Google Scholar]
150. Li Z, et al. Reconstructed mung bean trypsin inhibitor targeting cell surface GRP78 induces apoptosis and inhibits tumor growth in colorectal cancer. Int J Biochem Cell Biol. 2014;47:68–75. [PubMed] [Google Scholar]
151. de Ridder GG, Ray R, Pizzo SV. A murine monoclonal antibody directed against the carboxyl-terminal domain of GRP78 suppresses melanoma growth in mice. Melanoma Res. 2012;22:225–235. [PubMed] [Google Scholar]
152. Rauschert N, et al. A new tumor-specific variant of GRP78 as target for antibody-based therapy. Lab Invest. 2008;88:375–386. [PubMed] [Google Scholar]
153. Rasche L, et al. The natural human IgM antibody PAT-SM6 induces apoptosis in primary human multiple myeloma cells by targeting heat shock protein GRP78. PLoS One. 2013;8:e63414. [PMC free article] [PubMed] [Google Scholar]
154. Hensel F, Eckstein M, Rosenwald A, Brandlein S. Early development of PAT-SM6 for the treatment of melanoma. Melanoma Res. 2013;23:264–275. [PubMed] [Google Scholar]
155. Cohen M, Petignat P. Purified autoantibodies against glucose-regulated protein 78 (GRP78) promote apoptosis and decrease invasiveness of ovarian cancer cells. Cancer Lett. 2011;309:104–109. [PubMed] [Google Scholar]
156. Al-Hashimi AA, et al. Binding of anti-GRP78 autoantibodies to cell surface GRP78 increases tissue factor procoagulant activity via the release of calcium from endoplasmic reticulum stores. J Biol Chem. 2010;285:28912–28923. [PMC free article] [PubMed] [Google Scholar]
157. Arnouk H, et al. Tumour secreted grp170 chaperones full-length protein substrates and induces an adaptive anti-tumour immune response in vivo. Int J Hyperthermia. 2010;26:366–375. [PMC free article] [PubMed] [Google Scholar]
158. Schreiber TH, Deyev VV, Rosenblatt JD, Podack ER. Tumor-induced suppression of CTL expansion and subjugation by gp96-Ig vaccination. Cancer Res. 2009;69:2026–2033. [PMC free article] [PubMed] [Google Scholar]
159. Randazzo M, Terness P, Opelz G, Kleist C. Active-specific immunotherapy of human cancers with the heat shock protein Gp96-revisited. Int J Cancer. 2012;130:2219–2231. [PubMed] [Google Scholar]
160. Reitsma DJ, Combest AJ. Challenges in the development of an autologous heat shock protein based anti-tumor vaccine. Hum Vaccin Immunother. 2012;8:1152–1155. [PMC free article] [PubMed] [Google Scholar]
161. Colaco C. Autologous heat-shock protein vaccines. Hum Vaccin Immunother. 2013 Epub ahead of print. [PMC free article] [PubMed] [Google Scholar]
162. Li X, et al. Induction of regulatory T cells by high-dose gp96 suppresses murine liver immune hyperactivation. PLoS One. 2013;8:e68997. [PMC free article] [PubMed] [Google Scholar]
163. Zhao B, et al. TAT-mediated gp96 transduction to APCs enhances gp96-induced antiviral and antitumor T cell responses. Vaccine. 2013;31:545–552. [PubMed] [Google Scholar]
164. Wang XY, et al. Superior antitumor response induced by large stress protein chaperoned protein antigen compared with peptide antigen. J Immunol. 2010;184:6309–6319. [PMC free article] [PubMed] [Google Scholar]
165. Wang XY, et al. Extracellular targeting of endoplasmic reticulum chaperone glucose-regulated protein 170 enhances tumor immunity to a poorly immunogenic melanoma. J Immunol. 2006;177:1543–1551. [PubMed] [Google Scholar]
166. Gao P, Sun X, Chen X, Subjeck J, Wang XY. Secretion of stress protein grp170 promotes immune-mediated inhibition of murine prostate tumor. Cancer Immunol Immunother. 2009;58:1319–1328. [PMC free article] [PubMed] [Google Scholar]
167. Yu X, et al. A multifunctional chimeric chaperone serves as a novel immune modulator inducing therapeutic antitumor immunity. Cancer Res. 2013;73:2093–2103. [PMC free article] [PubMed] [Google Scholar]
168. Tamura Y, et al. Tumor-produced secreted form of binding of immunoglobulin protein elicits antigen-specific tumor immunity. J Immunol. 2011;186:4325–4330. [PubMed] [Google Scholar]
169. Lee AS. In: Protein Discovery Technologies. Pasqualini R, Arap W, editors. CRC Press; Boca Raton: 2009. pp. 129–140. [Google Scholar]
170. Pouyssegur J, Shiu RP, Pastan I. Induction of two transformation-sensitive membrane polypeptides in normal fibroblasts by a block in glycoprotein synthesis or glucose deprivation. Cell. 1977;11:941–947. [PubMed] [Google Scholar]
171. Hightower LE. Cultured animal cells exposed to amino acid analogues or puromycin rapidly synthesize several polypeptides. J Cell Physiol. 1980;102:407–427. [PubMed] [Google Scholar]
172. Munro S, Pelham HR. An Hsp70-like protein in the ER: identity with the 78 kd glucose-regulated protein and immunoglobulin heavy chain binding protein. Cell. 1986;46:291–300. [PubMed] [Google Scholar]
173. Bole DG, Hendershot LM, Kearney JF. Posttranslational association of immunoglobulin heavy chain binding protein with nascent heavy chains in nonsecreting and secreting hybridomas. J Cell Biol. 1986;102:1558–1566. [PMC free article] [PubMed] [Google Scholar]
174. Haas IG, Wabl M. Immunoglobulin heavy chain binding protein. Nature. 1983;306:387–389. [PubMed] [Google Scholar]
175. Hendershot LM, et al. In vivo expression of mammalian BiP ATPase mutants causes disruption of the endoplasmic reticulum. Mol Biol Cell. 1995;6:283–296. [PMC free article] [PubMed] [Google Scholar]
176. Lee AS, Bell J, Ting J. Biochemical characterization of the 94- and 78-kilodalton glucose-regulated proteins in hamster fibroblasts. J Biol Chem. 1984;259:4616–4621. [PubMed] [Google Scholar]
177. Koch G, Smith M, Macer D, Webster P, Mortara R. Endoplasmic reticulum contains a common, abundant calcium-binding glycoprotein, endoplasmin. J Cell Sci. 1986;86:217–232. [PubMed] [Google Scholar]
178. Mazzarella RA, Green M. ERp99, an abundant, conserved glycoprotein of the endoplasmic reticulum, is homologous to the 90-kDa heat shock protein (hsp90) and the 94-kDa glucose regulated protein (GRP94) J Biol Chem. 1987;262:8875–8883. [PubMed] [Google Scholar]
179. Maki RG, Old LJ, Srivastava PK. Human homologue of murine tumor rejection antigen gp96: 5′-regulatory and coding regions and relationship to stress-induced proteins. Proc Natl Acad Sci USA. 1990;87:5658–5662. [PMC free article] [PubMed] [Google Scholar]
180. Johnson JL. Evolution and function of diverse Hsp90 homologs and cochaperone proteins. Biochim Biophys Acta. 2012;1823:607–613. [PubMed] [Google Scholar]
181. Schild H, Rammensee HG. gp96--the immune system's Swiss army knife. Nat Immunol. 2000;1:100–101. [PubMed] [Google Scholar]
182. Wadhwa R, Kaul SC, Ikawa Y, Sugimoto Y. Identification of a novel member of mouse hsp70 family. Its association with cellular mortal phenotype. J Biol Chem. 1993;268:6615–6621. [PubMed] [Google Scholar]
183. Sciandra JJ, Subjeck JR. The effects of glucose on protein synthesis and thermosensitivity in Chinese hamster ovary cells. J Biol Chem. 1983;258:12091–12093. [PubMed] [Google Scholar]
184. Chen X, et al. The 170 kDa glucose regulated stress protein is a large HSP70-, HSP110-like protein of the endoplasmic reticulum. FEBS Lett. 1996;380:68–72. [PubMed] [Google Scholar]
185. Kuwabara K, et al. Purification and characterization of a novel stress protein, the 150-kDa oxygen-regulated protein (ORP150), from cultured rat astrocytes and its expression in ischemic mouse brain. J Biol Chem. 1996;271:5025–5032. [PubMed] [Google Scholar]
186. Sciandra JJ, Subjeck JR, Hughes CS. Induction of glucose-regulated proteins during anaerobic exposure and of heat-shock proteins after reoxygenation. Proc Natl Acad Sci USA. 1984;81:4843–4847. [PMC free article] [PubMed] [Google Scholar]
187. Yoshida H, Haze K, Yanagi H, Yura T, Mori K. Identification of the cis-acting endoplasmic reticulum stress response element responsible for transcriptional induction of mammalian glucose-regulated proteins. Involvement of basic leucine zipper transcription factors. J Biol Chem. 1998;273:33741–33749. [PubMed] [Google Scholar]
188. Roy B, Lee AS. The mammalian endoplasmic reticulum stress response element consists of an evolutionarily conserved tripartite structure and interacts with a novel stress-inducible complex. Nucleic Acids Res. 1999;27:1437–1443. [PMC free article] [PubMed] [Google Scholar]
189. Baumeister P, et al. Endoplasmic reticulum stress induction of the Grp78/BiP promoter: activating mechanisms mediated by YY1 and its interactive chromatin modifiers. Mol Cell Biol. 2005;25:4529–4540. [PMC free article] [PubMed] [Google Scholar]
190. Hong M, et al. Transcriptional regulation of the Grp78 promoter by endoplasmic reticulum stress: role of TFII-I and its tyrosine phosphorylation. J Biol Chem. 2005;280:16821–16828. [PubMed] [Google Scholar]
191. Lee AH, Iwakoshi NN, Glimcher LH. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol. 2003;23:7448–7459. [PMC free article] [PubMed] [Google Scholar]
192. Kaneda S, Yura T, Yanagi H. Production of three distinct mRNAs of 150 kDa oxygen-regulated protein (ORP150) by alternative promoters: preferential induction of one species under stress conditions. J Biochem. 2000;128:529–538. [PubMed] [Google Scholar]
193. Giorgi C, De Stefani D, Bononi A, Rizzuto R, Pinton P. Structural and functional link between the mitochondrial network and the endoplasmic reticulum. Int J Biochem Cell Biol. 2009;41:1817–1827. [PMC free article] [PubMed] [Google Scholar]
194. Hayashi T, Rizzuto R, Hajnoczky G, Su TP. MAM: more than just a housekeeper. Trends Cell Biol. 2009;19:81–88. [PMC free article] [PubMed] [Google Scholar]
195. Hetz CA. ER stress signaling and the BCL-2 family of proteins: from adaptation to irreversible cellular damage. Antioxid Redox Signal. 2007;9:2345–2355. [PubMed] [Google Scholar]
196. Ouyang YB, Xu LJ, Emery JF, Lee AS, Giffard RG. Overexpressing GRP78 influences Ca2+ handling and function of mitochondria in astrocytes after ischemia-like stress. Mitochondrion. 2011;11:279–286. [PMC free article] [PubMed] [Google Scholar]
197. Sun FC, et al. Localization of GRP78 to mitochondria under the unfolded protein response. Biochem J. 2006;396:31–39. [PMC free article] [PubMed] [Google Scholar]
198. Ye R, et al. Grp78 heterozygosity promotes adaptive unfolded protein response and attenuates diet-induced obesity and insulin resistance. Diabetes. 2010;59:6–16. [PMC free article] [PubMed] [Google Scholar]
199. Mao C, et al. Targeted mutation of the mouse Grp94 gene disrupts development and perturbs endoplasmic reticulum stress signaling. PLoS One. 2010;5:e10852. [PMC free article] [PubMed] [Google Scholar]
-