Skip to main content
Philosophical Transactions of the Royal Society B: Biological Sciences logoLink to Philosophical Transactions of the Royal Society B: Biological Sciences
. 2017 Dec 4;373(1738):20160522. doi: 10.1098/rstb.2016.0522

Extracellular cell stress (heat shock) proteins—immune responses and disease: an overview

A Graham Pockley 1,, Brian Henderson 2
PMCID: PMC5717522  PMID: 29203707

Abstract

Extracellular cell stress proteins are highly conserved phylogenetically and have been shown to act as powerful signalling agonists and receptors for selected ligands in several different settings. They also act as immunostimulatory ‘danger signals’ for the innate and adaptive immune systems. Other studies have shown that cell stress proteins and the induction of immune reactivity to self-cell stress proteins can attenuate disease processes. Some proteins (e.g. Hsp60, Hsp70, gp96) exhibit both inflammatory and anti-inflammatory properties, depending on the context in which they encounter responding immune cells. The burgeoning literature reporting the presence of stress proteins in a range of biological fluids in healthy individuals/non-diseased settings, the association of extracellular stress protein levels with a plethora of clinical and pathological conditions and the selective expression of a membrane form of Hsp70 on cancer cells now supports the concept that extracellular cell stress proteins are involved in maintaining/regulating organismal homeostasis and in disease processes and phenotype. Cell stress proteins, therefore, form a biologically complex extracellular cell stress protein network having diverse biological, homeostatic and immunomodulatory properties, the understanding of which offers exciting opportunities for delivering novel approaches to predict, identify, diagnose, manage and treat disease.

This article is part of the theme issue ‘Heat shock proteins as modulators and therapeutic targets of chronic disease: an integrated perspective’.

Keywords: heat shock (stress) proteins, extracellular, immunity

1. Background

Chance favours the prepared mind.

—Louis Pasteur (1822–1895)

The presence of additional new ‘puffs’ in the polytene chromosomes of cultured Drosophila larva which were induced following their incubation at an inadvertently high temperature and observed by Ferruccio Ritossa (25 February 1936–9 January 2014) in the early 1960s was unexpected and puzzling. He realized the potential importance of this first evidence that stress can influence gene transcription and induce the synthesis of new proteins, yet found it surprisingly difficult to publish this discovery. It was eventually published in Experientia [1,2].

Ritossa's findings were extended and expanded upon during the next decade, and by the mid-to-late 1960s, it was clear that exposure of cells containing polytene chromosomes to a variety of environmental stressors resulted in the transcription of novel genes and, presumably, in the synthesis of specific proteins. However, it was not until the 1970s when Tissières at the University of Geneva and other investigators in this area [3,4] applied the newly developed technique of sodium dodecyl sulfate (SDS)–PAGE to reveal the appearance of new protein bands having distinct molecular masses in salivary glands after the application of heat shock. It was also noted that cellular levels of some proteins that were present before the application of elevated temperature either decreased or disappeared after treatment. Here was the first evidence for the existence of heat shock proteins (HSPs) or cell stress proteins, and it was then that these terms were coined. However, it is now clear that a range of different stressors, other than heat, such as viral infection, cytokines, oxidative stress, ionizing and UV irradiation, glucose deprivation or exposure to toxins and certain metals, also induce the expression of such proteins. A more descriptively correct term for these proteins is therefore ‘cell stress’ proteins [5].

The fact that research on the heat shock response was predominantly undertaken in Drosophila during the 1960s and 1970s led to the expectation that this response was specific to insects or even to Drosophila itself. However, observations that the heat shock response was present in chicken fibroblasts [6], Escherichia coli [7], yeast [8] and plants [9] indicated that the heat shock/cell stress response is a universal phenomenon. The cloning of the Drosophila genes encoding HSPs and the sequencing of many of the relevant genes by the late 1970s/early 1980s revealed the evolutionary relationships between the response and the proteins involved (e.g. [10]).

The relationships between stress-induced gene transcription and the roles of cell stress proteins in protein folding and the management of the intracellular environment took many years to be understood and consolidate [3,11,12]. Larry Hightower [13], a pioneer in studying the physiological role of cell stress proteins, first suggested that, as many of the stressors were protein chaotropes (agents able to denature proteins), then the most obvious function of this stress response was to manage and deal with improperly folded proteins within the cell. This hypothesis was tested using a simple experimental protocol which determined the influence of direct microinjection of native or denatured proteins into frog oocytes on the induction of the stress response. Only denatured proteins induced the response, thereby establishing the link between protein unfolding within the cell and the induction of the cell stress response [11].

By the late 1980s, it had been recognized that cellular proteins require help with their folding in some instances, and that this was facilitated via the actions of families of proteins termed ‘molecular chaperones’ [12], the accepted definition of which is ‘a large and diverse group of proteins that share the property of assisting the non-covalent assembly/disassembly of other macromolecular structures, but which are not permanent components of these structures when these are performing their normal biological functions’ [14]. Molecular chaperones fulfil essential cellular ‘housekeeping’ and cytoprotective functions, and thereby ensure correct functionality. They also enable cells to cope with the plethora of insults and stresses that exist in the complex and dynamic intracellular environment (table 1).

Table 1.

Mammalian cell stress response proteins, and their intracellular localization and function. ER, endoplasmic reticulum; TCP-1, tailless complex polypeptide; Grp, glucose-regulated protein; Hsp, heat shock protein; BiP, immunoglobulin heavy chain binding protein; mtHsp70, mitochondrial Hsp70; HSF1, heat shock factor 1; Apg-1, protein kinase essential for autophagy. Adapted from [15,16]. Further information on the nomenclature and individual family members has been published elsewhere [17,18].

major family, and members intracellular localization intracellular function
small Hsps
αB-crystallin
Hsp27
haeme oxygenase, Hsp32
cytoplasm
cytoplasm/nucleus
cytoplasm
cytoskeletal stabilization
actin dynamics
haeme catabolism, antioxidant properties
Hsp60 or chaperonins
Hsp60
TCP-1
mitochondria
cytoplasm
both bind to partially folded polypeptides and assist correct folding
assembly of multimeric complexes
Hsp70
Hsp70 (inducible)
Hsc70 (cognate)
Grp78/BiP
mtHsp70/Grp75
cytoplasm/nucleus
cytoplasm/peroxisome
ER
mitochondria
all bind to extended polypeptides
all prevent aggregation of unfolded peptides
all dissociate some oligomers
ATP binding
ATPase activity
Hsp70 is involved in the regulation of HSF1 activity and the repression of heat shock protein gene transcription
Hsp90
Hsp90 (α and β)
Grp94/gp96/Hsp100
cytoplasm
ER
all bind to other proteins
all regulate protein activity
all prevent aggregation of re-folded peptide
correct assembly and folding of newly synthesized protein
Hsp90 appears to be involved in maintaining the HSF1 monomeric state in non-stressful conditions. Represents 1–2% of total protein
Hsp110
Hsp110 (human)
Apg-1 (mouse)
Hsp105
nucleolus/cytoplasm
cytoplasm
cytoplasm
thermal tolerance
protein refolding

Since these early studies, a growing number of proteins that are involved in protein folding and in the cell stress response have been identified in all three of life's Kingdoms. Several families of the molecular chaperones include large numbers of proteins. For example, the Hsp40 family contains 50 members. The fact that the number of human molecular chaperones and protein-folding catalysts is probably in the region of 150 proteins underlies the enormous complexity of the cell stress response. The cell stress protein families can be subdivided into molecular chaperones, which aid protein folding without changing the client protein in any way, protein-folding catalysts such as protein disulfide isomerases, which catalyse SH-:-S-S- interconversions, and peptidyl prolyl isomerases, which catalyse cis : trans isomerization of prolines and thus induce chemical changes in their client proteins [19]. The protein-folding catalysts can also be involved in redox interactions, and this phenomenon of oxidation and reduction both within and outside the cell is now recognized as being a major modulator of biological behaviour (e.g. [20]). To confuse matters further, molecular chaperones and protein-folding catalysts can either be proteins whose genes are induced by stress, or be constitutively expressed proteins whose genes fail to be modulated by stress—only the former are classed as cell stress proteins.

Discontinuous PAGE gels enable accurate molecular masses to be identified, and informed the nomenclature for the heat shock (cell stress) proteins (e.g. Hsp60, Hsp70). However, despite the publication of nomenclature guidelines [17], the literature remains unclear, especially in the case of the 70 kDa family of molecules. The human HSP70 (gene) family consists of at least eight members, only three of which show stress-inducible expression [18]. Of the 13 protein members of the family, two closely linked genes, referred to as Hsp70-1, are the major stress-induced members [18]. Although some evidence implicates Hsp70-2 in human cancer, the cytosolic, stress-induced Hsp70-1 is the predominant form that is overexpressed in cancer [18]. It is, therefore, likely that it is this form of the molecule which is being measured in the studies that have been reported to date. However, it is important that the identity of the analyte being reported upon is verified using information on the specificity of the antibodies that are being used in the assays.

As indicated above, it is now apparent that proteins can have multiple functions, the manifestations of which are dictated by the context in which they are generated and encountered. For instance, can proteins such as stress proteins exhibit distinct profiles of physiological activities when in the intracellular and extracellular environments? If so, and this indeed appears to be the case, then this would argue against the concept of ‘one protein, one function’. Although the concept of ‘one protein, one function’ is not universally accepted, Campbell & Scanes [21] first proposed the term ‘protein moonlighting’ to describe the capacity of certain proteins to exhibit more than one biological function, specifically the apparent immunological functions of ‘endocrine peptides’. A number of prokaryote and eukaryote proteins have been shown to exhibit ‘moonlighting functions’, and this concept has been expanded upon by a number of proponents [2225]. The concept of protein moonlighting is discussed in detail by Constance Jeffery elsewhere in this issue [26].

However, it should be noted that many biologically important molecules—if not all of them—express more than one function, and the implication that a protein has only one bona fide function and that the other functions are secondary, if not superfluous, might not necessarily be the case. Another counterview to moonlighting functions of HSP (stress) in the immunological context (see below) is that these proteins might not have evolved a second function at all. Rather, it was the immune system that evolved to recognize and respond to these proteins on the basis of changed accessibility, rather than changes in physiological function.

2. Cell stress proteins are released into the extracellular environment

The concept that stress proteins can be released from cells in the absence of necrosis was highlighted by Hightower & Guidon in 1989. In this study, heat treatment was shown to increase the profile of proteins that were released from cultured rat embryo cells, from a small profile that included the constitutively expressed member of the 70 kDa family of molecules, Hsc70, to also include its inducible counterpart, Hsp70 and Hsp110 [27]. Although protein release occurred in the absence of any overt level of cellular necrosis (and was therefore likely to be an ‘active’ physiological process), it was not mediated via the common secretory pathway, as inhibitors of this pathway (colchicine, monensin) did not block it [27]. These findings, aligned with the slightly earlier study from Tytell et al. [28] in 1986, who reported the transfer of glia–axon transfer proteins (including Hsp70, Hsc70 and Hsp100) from adjacent glial cells into the squid giant axon. This response was proposed to reflect a mechanism that enabled glial cells to protect adjacent neuronal cells that exhibit a deficient ability to generate a protective response to stress.

These initial findings, and the subsequent studies reporting the presence of Hsp60 and Hsp70 in the peripheral circulation of healthy individuals by Pockley et al. in the late 1990s [29,30], were received with scepticism by the biological and biochemical communities, as it was unclear how these proteins could be released from viable cells, given that they do not express the typical N-terminal signal peptide sequences that enable secretion. However, this argument is not a strong one, as ‘non-classical’ secretion of proteins lacking such sequences has been observed for several proteins, including fibroblast growth factors 1 and 2 (FGF-1,2), interleukin-1 (IL-1) and high mobility group box 1 (HMGB-1). The mechanisms underlying non-classical secretion pathways have been reviewed elsewhere [31]. Cell stress proteins have now been reported to be released from a wide range of cells including insulin-secreting β cells, rat cortical astrocytes, a human neuroblastoma cell line, a human keratinocyte-derived cell line, cultured vascular smooth muscle cells and a broad profile of tumour cells including murine and human prostate cancer cells and B cells (reviewed in [32]), and to exist in the circulation in a number of healthy and diseased states (see below).

Extracellular Hsp70 exists either as a free protein, as a protein in association with lipid vesicles such as exosomes [33,34] and lysosomal endosomes [35] or in the context of cholesterol-rich microdomains [36]. Vesicular transport [37] and ubiquitination-triggered transport [38] have also been proposed. Recent studies have demonstrated that the minority of extracellular Hsp70 is ‘free’ Hsp70, and this is mostly derived from dying cells [39].

Exosomes are small membrane vesicles that form within late endocytic compartments called multi-vesicular bodies (MVBs). They are distinct to apoptotic vesicles in that they differ in their mode of production and protein composition [40]. The fusion of MVBs with the plasma membrane leads to the release of exosomes into the extracellular space. Various haematopoietic and non-haematopoietic cell types secrete exosomes, including reticulocytes, B and T lymphocytes, mast cells, platelets, macrophages, alveolar lung cells, tumour cells, intestinal epithelial cells and professional antigen-presenting cells (APCs) such as dendritic cells (DCs), with the function of exosomes in different physiological processes depending on their origin [41]. DC- and tumour-derived exosomes are enriched in Hsp70, Hsc70 and Hsp90 [42,43], and exosomes released from reticulocytes also contain Hsp70 [44]. Exosomal release of stress proteins and the role of exosomal-associated HSPs in cancer have been reviewed in the literature [45,46] and by Gabriele Multhoff elsewhere in this issue [47].

3. Cell stress proteins as immunomodulatory mediators

Although probably not fully appreciated at the time, the concept that stress proteins can exist in the peripheral circulation had been established in 1977 in a study that reported the presence of a protein (early pregnancy factor (EPF)) in the serum of women in the first trimester of pregnancy [48]. This protein was demonstrated to have immunosuppressive properties two years later [49] and was identified as being HSP 10 (Hsp10) in 1994 [50]. It has also been shown that Hsp10, a 10 kDa monomer that caps the Hsp60 oligomer and facilitates protein folding [51] is also present at low levels in non-pregnant individuals [52]. Hsp10 inhibits the secretion of several inflammatory mediators [53], and its immunosuppressive properties can attenuate a variety of human inflammatory diseases [5457]. The finding that circulating levels of Hsp10 in patients with periodontal disease are lower than those in matched, disease-free, controls and that levels only return to normal after effective therapy suggest the control of circulating Hsp10 levels by localized inflammation [52]. Hsp10 therefore appears to be a homeostatic controller of inflammation, in addition to being an integral component of the intracellular molecular chaperone machinery.

With regard to Hsp60 and Hsp70, the discovery of these proteins in the peripheral circulation of overtly normal individuals led to a certain degree of confusion, as these proteins were considered as being pro-inflammatory molecules when present in the extracellular environment. Indeed, one of the major issues for investigators studying the immunobiology of extracellular stress proteins is the apparently contradictory evidence that indicates both pro- and anti-inflammatory roles for these proteins. The problem is that the immunological properties of these proteins continue to be discussed in isolation and it is essential that a more systems biology approach to extracellular HSPs is adopted in order to better reflect their physiological context and roles. Although many studies indicate pro-inflammatory properties for Hsp60 and Hsp70 in their interactions with monocytes, macrophages and DCs [5862], it has been speculated that at least some of these inflammatory effects result from the presence of contaminating endotoxin in the recombinant protein preparations, especially those that have been generated using bacterial expression systems [6366]. However, much evidence argues against this being the universal explanation for these effects, as has been reviewed elsewhere [25,67]. It is, therefore, essential to ensure that reagents and experimental design(s) are beyond question when it comes to undertaking experiments in this area. The influence of HSPs on immune responses in a number of contexts has been reviewed elsewhere [15,68,69] and in this issue.

In contrast with their reported pro-inflammatory properties, a body of literature indicates that Hsp60 and Hsp70 can have profound anti-inflammatory effects. Relatively historic data have reported that the induction of T cell reactivity to self-Hsp60 and self-Hsp70 promotes the development of Th2 type CD4+ T cells producing the regulatory cytokines IL-4 and IL-10 and downregulates disease in a number of experimental models of inflammatory disease [7074]. It has also been shown that DNA vaccines encoding for these proteins inhibit experimental arthritis and diabetes [74,75]. The recognition of conserved (self) epitopes on these highly conserved molecules dominantly downregulates the inflammatory capacity of the non-conserved (non-self) epitopes [76]. Human Hsp60 can act as a co-stimulator and activator of CD4+CD25+ regulatory T cell populations by interacting with Toll-like receptor 2 (TLR2) [77] and the treatment of such cells with Hsp60 enhances their ability to regulate the CD8+ T cell populations via direct cell–cell contact and the secretion of the immunoregulatory cytokines IL-10 and TGF-β [77]. The anti-inflammatory potential of Hsp60 and Hsp60-derived peptides has also been demonstrated in studies that have used these to modulate the rejection of murine skin allografts [78,79] and autoimmune disease—the latter is discussed elsewhere [8082] and by Willem van Eden in this issue [83]. It, therefore, appears that the net outcome of any immune response is dependent on the relative strengths of these antagonistic events (reviewed in [68]). The interactions of Hsp60 with the innate and adaptive immune systems and their immunoregulatory consequences have been reviewed and considered by Quintana & Cohen [84].

4. Extracellular cell stress proteins in health and disease

The initial identification of Hsp60 and Hsp70 in the peripheral circulation [29,30] stimulated interest in this area and the development of a range of ‘in-house’ and commercial enzyme immunoassays for measuring stress proteins in extracellular compartments. Most commercially available enzyme immunoassays for cell stress proteins are optimized for free Hsp70 in buffer, but not for Hsp70 in the serum, plasma or other body fluids, and so it is essential that investigators are aware of the limitations of the assays they use. It is also a matter of debate as to whether liposomal cell stress proteins can be detected using the standard detergents that are typically included in commercial enzyme immunoassay kits. Notwithstanding the above, these studies have led to many reports associating circulating levels of cell stress proteins with healthy and diseased states (table 2), including cancer (table 3). An immediate issue relating to these studies is the need to ensure that the commercial assay kits and the ‘in-house’ assays that have been used have been properly validated for the analysis of the relevant analytes in the biological fluid that is under investigation [132]. Such information is not always apparent, and differences in the levels of HSPs in the circulation which are measured by commercial and ‘in-house’ enzyme immunoassays have been reported [133]. Serum and plasma are complex and ‘matrix’-related effects can influence measurements in biological samples. Furthermore, a clinical method comparison study has revealed that commercially available HSP27 assays are not equally useful for differentiating patients with non-small cell lung carcinoma (NSCLC) from healthy controls [134].

Table 2.

Circulating cell stress proteins in disease.

condition key findings reference
Hsp10 periodontitis lower plasma levels in periodontal disease and treatment increases these. Post-treatment levels correlate with markers of clinical improvement [52]
Hsp27 renal disease elevated serum and urine levels in chronic kidney disease [85]
autoimmunity serum levels may be a novel marker for diabetic neuropathy in patients with Type 1 diabetes [86]
chronic heart failure soluble Hsp27 is a novel candidate biomarker for diagnosing CHF with preserved ejection fraction [87]
Hsp60 stress association between elevated levels of Hsp60, low socioeconomic status and social isolation in males and females, and with psychological distress in women [88]
cardiovascular disease elevated serum levels in patients with renal and peripheral vascular disease and individuals with borderline hypertension. Serum levels in individuals with hypertension are similar to normotensive controls [8992]
elevated levels present in coronary eluates after myocardial infarction [93]
serum levels increase with accumulating features of the metabolic syndrome in postmenopausal women [94]
endothelium-dependent vasodilator function is impaired in children with detectable levels of serum Hsp60. Circulating Hsp60, or factors that stimulate the expression and systemic release of Hsp60, may contribute to the initiation of arterial disease in early life [95]
association between higher levels of plasma Hsp60 in subjects with clinically manifest cardiovascular disease and those with a history of myocardial infarction in diabetes mellitus [96]
infections plasma Hsp60 levels are elevated in HIV-infected patients. Although levels reduce after anti-retroviral therapy, they remain higher than uninfected controls. Hsp60 levels correlate with viral load, CD4+ T cell counts, and circulating soluble CD14 and lipopolysaccharide levels [97]
periodontitis a larger proportion of patients with periodontal disease exhibit intermediate levels of plasma Hsp60 than controls. Treatment has no influence on levels [52]
atherogenic dyslipidaemia and elevated circulating Hsp60 levels are linked and associated with periodontal pathology [98]
autoimmunity serum Hsp60 levels correlate with time required for remission from flare-ups in patients with juvenile idiopathic arthritis [99]
Hsp70 surgery/trauma plasma Hsp70 levels markedly increase in patients undergoing liver resection and are associated with post-operative infection, hepatic ischaemic time and the degree of post-operative organ dysfunction [100]
Hsp70 is released into the circulation following coronary artery bypass grafting [101]
cardiovascular disease elevated serum levels in patients with renal and peripheral vascular disease and individuals with borderline hypertension. By contrast, serum levels in hypertension are similar to normotensive controls [8991]
low serum levels at baseline predict the development of atherosclerosis in individuals with established hypertension [102]
increased serum levels associated with low risk of coronary artery disease [103]
increased circulating levels may be associated with the progression of atrial fibrillation and its recurrence after catheter ablation [104]
serum levels correlate with the severity of atherosclerosis in patients with carotid artery disease and chronic lower limb ischaemia. Putative role for circulating Hsp70 in the development of arterial calcification [105]
infections serum levels positively associated with the degree of inflammation in an elderly population living in a remote area in Cameroon, where infection and parasitosis are endemic [106]
positive correlations between serum levels and inflammatory markers [107]
serum Hsp70 levels in patients with chronic hepatitis are higher than controls, but lower than in patients with liver cancer [108]
pregnancy serum levels are lower in normal human pregnancy, but elevated in transient hypertension of pregnancy, in pre-eclampsia and in superimposed pre-eclampsia. Increased serum levels reflect systemic inflammation, oxidative stress and hepatocellular injury in pre-eclampsia [109111]
asthma induced sputum and plasma Hsp70 levels could serve as a useful marker for assessing airway obstruction in patients with asthma [112]
renal disease elevated urinary Hsp70 levels in stages 4 and 5 chronic kidney disease [85]
diabetes serum levels are increased in Type 1 and Type 2 diabetes [113118]
serum levels are increased and correlate with HbA1c values in women with gestational diabetes mellitus
autoimmunity plasma Hsp70 levels are high in patients with Type I diabetes [119,120]
BiP periodontitis lower circulating levels of BiP (grp78) in periodontal disease as compared to controls. Treatment has no influence on levels [52]
grp94 autoimmunity plasma grp94 (gp96) levels are high in patients with Type I diabetes [119,120]

Table 3.

Extracellular cell stress proteins in cancer. LipHsp70, liposomal Hsp70.

tumour key findings reference
Hsp27 ovarian serum Hsp27 levels are increased in epithelial ovarian cancer and correlate with peritoneal metastases. Serum Hsp27 levels may be used as a potential additional indicator for peritoneal metastases and the response of patients to treatment [121]
breast significant differences in the profiles of Annexin V+, CD66+, BCRP1+ and Hsp27+ microparticles are present in breast cancer patients with lymph node metastases, as assessed using flow cytometry [122]
lung serum levels of Hsp27 are significantly elevated in patients with non-small cell lung cancer diagnosed at an early or at an advanced stage and can distinguish between early and advanced stage disease [123]
Hsp70 leukaemia levels of plasma Hsp70 reflect overall tumour load and patients with higher levels of plasma Hsp70 have significantly shorter survival in acute myeloid leukaemia and acute lymphoblastic leukaemia circulating Hsp70 might, therefore, be a biomarker for poor prognosis? [124]
plasma Hsp70 levels above the median in chronic myeloid leukaemia are associated with a higher rate of progression to the accelerated/blast phase, and a tendency towards shorter survival. Plasma Hsp70 could be a potential marker for predicting disease progression in patients with chronic phase in chronic myeloid leukaemia [125]
colorectal serum levels of Hsp70 and mortalin are independent variables, and high serum levels of mortalin (mitochondrial Hsp70, grp75, HSPA9) are a risk factor for shorter survival patients with colorectal cancer. The concurrence of high serum Hsp70 and mortalin levels is associated with rapid disease progression [126]
serum Hsp70 levels have potential as a stage-independent prognostic marker in colorectal cancer without distant metastasis [127]
head and neck plasma Hsp70 levels are significantly higher in mice bearing membrane Hsp70-positive FaDu human squamous cell carcinomas of the head and neck, and these correlate with tumour volume. Radiation-induced tumour regression is associated with significantly decreased Hsp70 levels, and these return to those of control animals after complete remission [128]
serum Hsp70 levels are significantly higher and associated with tumour volume in patients with squamous cell carcinoma of the head and neck. Following surgery and radiotherapy, Hsp70 levels fell without tumour relapse in the follow-up period. Hsp70 is, therefore, a potential tumour biomarker for monitoring the clinical outcome of radiotherapy. High levels associated with high levels of membrane Hsp70 expression on tumour cells [129]
liver serum Hsp70 levels in patients with liver cancer are significantly higher than a control group without liver disease, and individuals with chronic hepatitis. A subgroup of patients with cirrhosis who subsequently developed liver cancer exhibited higher serum Hsp70 levels than those patients with cirrhosis that did not progress to cancer [108]
pancreatic plasma Hsp70 levels are significantly higher in mice bearing membrane Hsp70-positive spontaneous pancreatic ductal adenocarcinomas, and levels correlated with tumour volume. Radiation-induced tumour regression was associated with significantly decreased Hsp70 levels, and levels returned to those of controls after complete remission [128]
serum Hsp70 levels are significantly increased in patients and may be useful as an additional biomarker for the detection of pancreatic cancer [130]
lung serum levels of Hsp70 are significantly elevated in patients with non-small cell lung cancer diagnosed at an early or at an advanced stage when compared with healthy control groups [123]
LipHsp70 circulating lipHsp70 levels in patients with head and neck, lung, colorectal, pancreatic cancer, haematological malignancies and especially glioblastoma are significantly higher than those in healthy human volunteers [39]
membrane Hsp70 membrane Hsp70 expression correlates with an improved overall survival in patients with colon and gastric carcinomas, whereas it is negatively associated with survival in patients with lower rectal and squamous cell carcinoma [131]
Hsp90 the baseline serum HSP90 levels of melanoma patients are significantly higher than those of the control subjects, but are not associated with clinical variables or survival

Reports on relationships between circulating stress protein levels and the clinical and physiological status of an individual (tables 2 and 3) are providing insight into the role of these proteins in the maintenance of a healthy state and/or the induction, progression and resolution of diseased states. As an example, the positive association between plasma Hsp60 levels in patients with cardiovascular disease and those with a history of myocardial infarction in diabetes mellitus implicates extracellular Hsp60 in the cardiovascular pathology which is associated with diabetes [96]. Lower levels of serum Hsp70 in normal human pregnancy could provide insight into the maintenance of immune tolerance in pregnancy [109]. Although studies have associated levels of Hsp70 in infection-related inflammation, the variability of the measured levels cannot distinguish patients from healthy subjects in this context [107]. However, the variability in levels, as measured using current approaches, does not currently allow measurement of these analytes to be used as a robust discriminatory approach for the identification of disease.

Given that a number of studies have reported relationships between circulating antibodies and their corresponding antigens in the peripheral circulation and the presence, severity and progression of disease, one should consider the potential involvement of circulating immune complexes in this context. This is a commonly overlooked parameter in studies that have investigated such relationships, and this might, in some instances at least, result from potential problems that are associated with measuring the presence of antibodies in a sample that includes its cognate antigen. Our own personal experience is that HSPs and HSP antibodies coexist in the peripheral circulation [29,30,89,90,102]. Although the presence of circulating anti-cell stress protein antibodies in the peripheral circulation might impact the measurements that are made, we have not found this to be the case, in that we have not observed any direct correlation between measured levels of circulating HSPs and anti-HSP antibody levels, at least using the assays that were available at the time [29,30,89,90,102,135]. Notwithstanding this, the presence/differential presence of immune complexes has the potential to influence the inflammatory status of an individual in that the interaction of APCs with soluble immune complexes has been shown to reduce their production of the Th1-biasing cytokine IL-12 to enhance their production of the regulatory cytokine IL-10 and, consequently, to induce a Th2-like (immunoregulatory) adaptive immune T cell response [136]. By contrast, a more recent study has reported that grp94 in complexes with IgG, which is a soluble diagnostic marker of gastrointestinal tumours, exert immune-stimulating activity on peripheral blood immune cells, as demonstrated by the triggering of inflammatory cytokine secretion [137]. It is clear that more studies aimed at understanding the relationship(s) between circulating cell stress proteins in their free and lipid-associated forms and their corresponding antibodies is required.

5. Extracellular cell stress proteins in cancer

The presence of circulating cell stress proteins in cancer, and relationships with tumour volume and therapeutic response [128,129] have been reported upon in a number of studies (table 3). Importantly, Multhoff and co-workers [108] have demonstrated that serum Hsp70 levels in patients with liver cancer are significantly higher than those that are measured in a control group of individuals without liver disease, and (importantly) are also higher than in individuals with chronic hepatitis. The same study showed that serum Hsp70 levels in a subgroup of patients with liver cirrhosis who subsequently developed liver cancer were higher than those in individuals with liver cirrhosis alone [108]. Dutta et al. [130] have reported that serum Hsp70 levels are significantly elevated in patients with pancreatic cancer, compared with both healthy controls and individuals with chronic pancreatitis. These findings demonstrate the capacity of serum Hsp70 levels to distinguish between inflammatory events/disease and cancer, and suggest that circulating Hsp70 might indeed be of value as a biomarker in cancer.

Hsp90 inhibitors are being evaluated for the treatment of cancers such as myeloma, breast, prostate and lung cancer, melanoma, gastrointestinal stromal tumours and acute myeloid leukaemia. Although the activity of Hsp90 inhibitors is currently assessed based on Hsp70 induction in peripheral blood mononuclear cells using western blot analysis, this approach is laborious, only semi-quantitative and difficult to implement in the clinic [138]. Serum Hsp70 measurements are now being used to monitor responses to Hsp90 inhibitors in the clinical setting, especially when access to tumour tissue is not possible [138].

6. Therapeutic potential and biological role of extracellular cell stress proteins

The concept that extracellular cell stress proteins could have therapeutic potential originally arose from studies into cross-reactive immunity to human Hsp60 by Irun Cohen's group in Israel, which found that T cells cross-reactive with Hsp60-induced diabetes in mice. Curiously, the administration of Mycobacterium tuberculosis Hsp65 protein could either induce diabetes or prevent it [139]. Analysis of the Hsp65 sequence (epitopes) recognized by T cells identified peptide 437–460 as a major T cell recognition epitope. The same sequence was identical in mouse Hsp60, apart from K for T at position 455. Crucially, it was found that immunization of non-obese diabetic (NOD) mice with this peptide (termed p277) inhibited the induction of diabetes [140]. Some 20 years later, the evaluation of this peptide in a phase III clinical trial showed evidence of clinical benefit [141].

The mitochondrion and bacterial cytosol contains Hsp60 and the co-chaperone, Hsp10, which acts as a ‘lid’ to the Hsp60 folding chamber. This 10 kDa protein, which normally forms a heptameric structure, was identified as a circulating immunosuppressive factor that was required for inhibiting immunity to the implanted embryo (termed ‘EPF’), in 1977 [48,49]. The potential role of this factor in the maintenance of pregnancy was confirmed by studies demonstrating that pregnant mice treated with anti-EPF antibodies failed to maintain their pregnancy [142]. It was not until 1994 that EPF was identified as Hsp10. A number of years later, Hsp10 was shown to inhibit experimental immunological models such as adjuvant arthritis (in this case, the protein was M. tuberculosis Hsp10 [143]) and experimental autoimmune encephalomyelitis (EAE) [144]. Short-term administration of M. tuberculosis Hsp10 has also been shown to inhibit experimental allergic asthma in mice [145].

The findings that recombinant Hsp10 inhibited LPS-induced inflammatory changes in macrophages and in mice exposed to LPS [53] led the Brisbane-based biopharmaceutical company, CBio Ltd, to attempt the commercialization of human Hsp10 as a therapeutic, and several small-scale clinical trials of a modified human Hsp10 (termed XToll) in a small number of conditions were undertaken. In a randomized, double-blind study of 23 rheumatoid arthritis patients, the intravenous administration of Hsp10 (5, 7.5 and 10 mg twice a week) induced either clinical benefit or disease remission in a significant number of individuals, with only one serious adverse event being reported [54]. Another small randomized, double-blind study demonstrated that the administration of XToll (Hsp10, 5, 7.5 and 10 mg) to 24 patients with plaque psoriasis over a 12-week dosing regimen of two doses per week reduced disease parameters [56]. Experimental findings that Hsp10 inhibited allergic encephalomyelitis prompted a double-blind randomized, placebo-controlled, phase II trial in 50 patients with multiple sclerosis. Although the Hsp10 was well tolerated, apart from showing a decreased circulating leucocyte cytokine synthetic capacity, the changes in clinical parameters were not significant [57]. Large peptides are not natural candidates for drug therapy and it is known that XToll, which is a modified Hsp10, induces antibodies in patients [56]. The two alternatives for this cell stress protein as a therapeutic are either to: (i) couple it with an Fc receptor of with polyethylene glycol; or (ii) generate active peptide fragments. The anti-arthritic actions of synthetic M. tuberculosis Hsp10 were found to reside in the N-terminus [146]. It is, therefore, possible that smaller fragments of Hsp10 may retain activity and could be the basis for developing non-peptidic isosteres of the Hsp10 peptide.

Another stress protein, which was originally considered as being an autoantigen that drove the progression of autoimmune disease, has subsequently been characterized as being a potent immunomodulatory molecule with clinical potential. Glucose-regulated protein 78 (grp78, binding immunoglobulin protein, BiP) [147] is essential for the assembly of immunoglobulin molecules [148], and is required for the translocation of nascent polypeptides across the endoplasmic reticulum membrane and protecting cells against ER stress [149]. It can also be expressed on the cell surface and acts as regulator of coagulation [150] and cell proliferation [151,152]. It has also been shown to be a potent immunoregulator [147].

BiP is present in the circulation of healthy individuals and at lower levels in patients with rheumatoid arthritis [147]. It is also found in synovial and oviductal fluid [153,154]. In contrast with the stress proteins that have been considered above, the secretion of BiP is likely to be via a classic route as it possesses the C-terminus ER retention signal (lysine, aspartic acid, glutamic acid, leucine (KDEL) amino acid sequence), which is common to proteins that reside in the ER. The multiple activities of BiP and its potential as a therapeutic agent for the management of inflammatory disease have been eloquently and comprehensively studied elsewhere [155]. With respect to therapeutic potential, a phase I/IIA clinical trial in 24 patients with rheumatoid arthritis who received a 1 h infusion of BiP (1, 5 or 15 mg) and who were followed up for 12 weeks has reported evidence of remission in patients receiving 5 and 15 mg doses [156].

Returning to the 70 kDa family, the constitutive member Hsc70 appears to play a role in, arguably, the most important biological process for the survival of species, namely reproduction. Proteomic analysis of porcine oviductal fluid has revealed that epithelial cells in the oviductal lumen secrete several molecules in response to the presence of spermatozoa, most notable of which are HSPs (stress) [157]. HSPs have also been identified in soluble fractions of pig and cow oviductal apical plasma membranes (sAPM) and in the human apical epithelium [158160]. These are potentially important findings, as the oviduct and oviductal sperm storage play key roles in reproduction by providing a secure reservoir in which spermatozoa can attain full fertilizing properties. Hsc70 appears to interact with components of the sperm cell surface membrane [158,159], and exposure to Hsc70 prolongs the survival of boar, bull and ram sperm [158,161]. Studies have now shown that a recombinant form of Hsc70 (HSPA8) rapidly promotes the viability of uncapacitated spermatozoa, the ability of spermatozoa to bind to oviductal epithelial cells, enhances the performance of in vitro fertilization procedures, and decreases sperm mitochondrial activity [162]. The repair of membrane damage is mediated via increase in sperm membrane fluidity. The ability of HSPA8 to influence membrane stability and fluidity, alongside its conserved nature among mammalian species, supports the idea that this protein protects sperm survival through membrane repair mechanisms [162]. Ongoing studies are elucidating the mechanisms that are involved in these protective effects and their potential impact on reproductive success and potential.

7. Therapeutic potential and biological role of a typically intracellular cell stress protein

Although this article has focused on those proteins that are known to be secreted by cells and therefore to be present under normal circumstances in biological fluids, several cell stress proteins that are not as well established as being in the extracellular environment under normal conditions have also been shown to have contrasting effects. A good example of such proteins is glucose-regulated protein 94 (gp96, HSPC4). Gp96 is a 94–96 kDa member of the Hsp90 family of molecular chaperones/stress proteins which resides within the lumen of the endoplasmic reticulum. In addition to being an intracellular chaperone [163,164], the administration of tumour-derived gp96 has been shown to induce tumour-specific cytolytic T cells and a protective tumour-specific immunity, the specificity of which is defined by peptides that are associated with the administered gp96 [165167]. By contrast, no protective effect is observed when high doses (2 × 10 µg intradermally) of tumour-derived gp96 are administered to mice [166]. Furthermore, appropriate doses of gp96 purified from normal liver can suppress the onset of diabetes in NOD mice and myelin basic protein- or proteolipid protein–induced autoimmune encephalomyelitis (EAE) in SJL mice [168], as well as prolonging the survival of murine skin allografts [169] and rat cardiac allografts [170]. The mechanisms that underlie these effects were originally proposed to involve the induction, activation and/or recruitment of as yet unidentified immunoregulatory T cell populations [168,169]. In our hands, gp96 could not be shown to be an activator of DCs, but did appear to activate CD3+ T cells in vitro [171], and lead to a state of peripheral T cell hyporesponsiveness following in vivo administration to rats bearing cardiac transplants [170]. More recent work provides a better insight into the mechanisms via which gp96 elicits dichotomous immune responses by providing evidence that low and high doses of gp96 preferentially engage conventional and plasmacytoid dendritic cells (pDCs), respectively, via CD91. Global DNMT-dependent epigenetic modifications modify protein expression within these APCs leading to an upregulation of neuropilin1 by pDCs which enables long-term interactions with Treg cells, thereby enhancing suppression of Th1 anti-tumour immunity [172].

The administration of autologous tumour-derived peptides bound to gp96 (HSPPC-96) induces individual tumour-specific immunity in patients with high-grade glioma [173] and has been shown to be safe for the treatment of patients with recurrent glioblastoma multiforme (GBM) in an open-label, single-arm, phase II study of 41 adult patients with surgically resectable recurrent GBM who were treated after gross total resection [174]. In the case of patients with newly diagnosed GBM, the addition of HSPPC-96 (Prophage™) to standard radiotherapy and temozolomide chemotherapy in a phase II, single-arm multi-centre trial involving 46 patients has been shown to have the potential to improve survival [175]. That the expression of PD-L1 on circulating myeloid cells impacts on systemic immunity suggests that the inhibition of such immunological ‘checkpoint’ pathways could further enhance the efficacy of this approach [175].

8. Membrane Hsp70: a ‘third’ form of the 70 kDa cell stress protein with diagnostic, therapeutic and imaging potential

Gabriele Multhoff discovered the selective expression of a membrane form of Hsp70, the major stress-inducible member of the 70 kDa HSP family, on the plasma membrane of tumour cells (but not normal tissue) using a unique monoclonal antibody (mAb, cmHsp70.1) [176178]. The expression of membrane Hsp70 has now been detected on a broad panel of cancer cell lines, and the density of membrane Hsp70 expression on cancer cells is increased by treatments such as radio(chemo)therapy [179]. An ongoing screening programme of over 1300 patients with various solid tumours in the Multhoff laboratory is revealing that more than 50% of all patients bear a membrane Hsp70–positive tumour. Membrane Hsp70 is also highly expressed on metastatic disease [180], and its expression is associated with an unfavourable prognosis and a reduced overall survival in patients with rectal carcinoma and squamous cell carcinoma [131]. Membrane Hsp70 expression is therefore a universal, selective tumour-specific marker of ‘aggressive’ disease.

Tumour cells that express Hsp70 on their plasma membrane secrete exosomes that express Hsp70 on the surface of their plasma membranes [33]. Given that the protein composition in the exosomal lumen reflects that of the cytosol of the respective cell, it would be expected that exosomes derived from normal cells contain low levels of Hsp70, whereas exosomes from tumour cells having a high cytosolic Hsp70 contain high levels of Hsp70 in their lumen and also present it on their lipid surface [181]. This concept has been confirmed, at least in part, by studies that have reported serum Hsp70 levels to be associated with a high membrane Hsp70 expression on tumours in patients with squamous cell carcinoma of the head and neck [129].

Aligned with these studies has been the development of an enzyme immunoassay that detects liposomal Hsp70 (lipHsp70) in serum and plasma [39]. This assay was conceived and developed based on the evidence that Hsp70 membrane-positive tumour cells actively release Hsp70 in exosome-like lipid vesicles and that most commercial Hsp70 enzyme-linked immunosorbent assays (ELISAs) are not validated for the detection of liposomal Hsp70 in serum. The assay exhibits a high level of precision, and a greater recovery of ‘spiked’ Hsp70 than its commercially available counterparts. The lipHsp70 ELISA is equally suitable for serum and plasma and the measured Hsp70 concentrations are not influenced by food intake, repeated freezing and thawing of the sample or moderate haemolysis. A comparison of the Hsp70 levels in patients with head and neck, lung, colorectal, pancreatic cancer, GBM or haematological malignancies and healthy human volunteers has revealed significantly higher levels in patients bearing tumours, and especially in those bearing aggressive tumours (e.g. GBM). The lipHsp70 ELISA, therefore, provides a highly sensitive and robust method for measuring liposomal and free Hsp70 in the circulation and could provide a clinically approach for detecting tumours and monitoring therapeutic responses and clinical outcome.

From a functional perspective, Hsp70-positive tumour-derived exosomes stimulate migratory and cytolytic activity of natural killer (NK) cells [33,182] and activate macrophages [183]. In a different context, tumour-derived exosomes expressing surface Hsp72 can restrain tumour immune surveillance by promoting the suppressive functions of myeloid-derived suppressor cells and plasma-derived exosomes expressing Hsp70 have powerful cardioprotective effects in models of cardiac ischaemia–reperfusion injury via a mechanism involving a membrane Hsp70/TLR4 communication axis [184].

The significant diagnostic, therapeutic and imaging potential of membrane Hsp70-based ‘theranostics’1 is considered by Gabriele Multhoff elsewhere in this issue [47].

9. Conclusion

Levels of HSP (cell stress) in biological fluids have been associated with a plethora of clinical conditions. These proteins could, therefore, act as indicators, drivers and/or moderators of disease processes and have potential utility as biomarkers of disease. Many, if not all, of the stress proteins that are released from cells under normal physiological conditions possess a range of biological functions, the nature of which depends on the context in which they are encountered. These proteins and networks have the potential to deliver a wealth of valuable, clinically relevant diagnostic and therapeutic approaches. The current challenge is to more fully understand these networks and establish their clinical potential.

That which drugs fail to cure, the scalpel can cure. That which the scalpel fails to cure, heat can cure. If the heat cannot cure, it must be determined to be incurable

—Hippocrates

Acknowledgements

The John van Geest Cancer Research Centre is supported by the John and Lucille van Geest Foundation, the Headcase Cancer Trust, the Roger Counter Foundation, the National Institute for Health Research (NIHR), NanoString Technologies Inc. and the Qatar National Research Fund.

Endnote

1

Theranostics: combining diagnostic and therapeutic capabilities into a single agent—a key element of precision medicine.

Data accessibility

This article has no additional data.

Competing interests

We declare we have no competing interests.

Funding

We received no funding for this study.

References

  • 1.Ritossa F. 1996. Discovery of the heat shock response. Cell Stress Chaperones 1, 97–98. ( 10.1379/1466-1268(1996)001%3C0097:DOTHSR%3E2.3.CO;2) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ritossa FA. 1962. A new puffing pattern induced by temperature shock and DNP in Drosophila. Experientia 18, 571–573. ( 10.1007/BF02172188) [DOI] [Google Scholar]
  • 3.Tissières A, Mitchell HK, Tracy U. 1974. Protein synthesis in salivary glands of Drosophila melanogaster: relation to chromosome puffs. J. Mol. Biol. 84, 389–398. ( 10.1016/0022-2836(74)90447-1) [DOI] [PubMed] [Google Scholar]
  • 4.Mirault ME, Goldschmidt-Clermont M, Moran L, Arrigo AP, Tissières A. 1978. The effect of heat shock on gene expression in Drosophila melanogaster. Cold Spring Harb. Symp. Quant. Biol. 42, 819–827. ( 10.1101/SQB.1978.042.01.082) [DOI] [PubMed] [Google Scholar]
  • 5.Ashburner M. 1970. Patterns of puffing activity in the salivary gland chromosomes of Drosophila. V. Responses to environmental treatments. Chromosoma 31, 356–376. ( 10.1007/BF00321231) [DOI] [PubMed] [Google Scholar]
  • 6.Johnston D, Oppermann H, Jackson J, Levinson W. 1980. Induction of four proteins in chick embryo cells by sodium arsenite. J. Biol. Chem. 255, 6975–6980. [PubMed] [Google Scholar]
  • 7.Tilly K, McKittrick N, Zylicz M, Georgopoulos C. 1983. The dnaK protein modulates the heat-shock response of Escherichia coli. Cell 34, 641–646. ( 10.1016/0092-8674(83)90396-3) [DOI] [PubMed] [Google Scholar]
  • 8.Miller FN, Joshua IG, Anderson GL. 1982. Quantitation of vasodilator induced macromolecular leakage by in vivo fluorescent microscopy. Microvasc. Res. 24, 56–67. ( 10.1016/0026-2862(82)90042-5) [DOI] [PubMed] [Google Scholar]
  • 9.Key JL, Lin CY, Chen YM. 1981. Heat shock proteins of higher plants. Proc. Natl Acad. Sci. USA 78, 3526–3530. ( 10.1073/pnas.78.6.3526) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Bardwell JC, Craig EA. 1984. Major heat shock gene of Drosophila and the Escherichia coli heat-inducible dnaK gene are homologous. Proc. Natl Acad. Sci. USA 81, 848–852. ( 10.1073/pnas.81.3.848) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Ananthan J, Goldberg AL, Voellmy R. 1986. Abnormal proteins serve as eukaryotic stress signals and trigger the activation of heat shock genes. Science 232, 522–524. ( 10.1126/science.3083508) [DOI] [PubMed] [Google Scholar]
  • 12.Hemmingsen SM, Woolford C, van der Vies SM, Tilly K, Dennis DT, Georgopoulos CP, Hendrix RW, Ellis RJ. 1988. Homologous plant and bacterial proteins chaperone oligomeric protein assembly. Nature 333, 330–334. ( 10.1038/333330a0) [DOI] [PubMed] [Google Scholar]
  • 13.Hightower LE. 1980. Cultured animal cells exposed to amino-acid analogues or puromycin rapidly synthesise several poplypeptides. J. Cell Physiol. 102, 407–424. ( 10.1002/jcp.1041020315) [DOI] [PubMed] [Google Scholar]
  • 14.Ellis RJ. 2005. Chaperone function: the orthodox view. In Molecular chaperones and cell signalling (eds Henderson B, Pockley AG), pp. 3–21. Cambridge, UK: Cambridge University Press. [Google Scholar]
  • 15.Pockley AG. 2003. Heat shock proteins and their role as regulators of the immune response. Lancet 362, 469–476. ( 10.1016/S0140-6736(03)14075-5) [DOI] [PubMed] [Google Scholar]
  • 16.Pockley AG. 2001. Heat shock proteins in health and disease: therapeutic targets or therapeutic agents? Exp. Rev. Mol. Med. 3, 1–21. ( 10.1017/S1462399401003556) [DOI] [PubMed] [Google Scholar]
  • 17.Kampinga HH, Hageman J, Vos MJ, Kubota H, Tanguay RM, Bruford EA, Cheetham ME, Chen B, Hightower LE. 2009. Guidelines for the nomenclature of the human heat shock proteins. Cell Stress Chaperones 14, 105–111. ( 10.1007/s12192-008-0068-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Murphy ME. 2013. The HSP70 family and cancer. Carcinogenesis 34, 1181–1188. ( 10.1093/carcin/bgt111) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Gething MJ. 1997. Guidebook to molecular chaperones and protein-folding catalysts. Oxford, UK: Oxford University Press. [Google Scholar]
  • 20.Lee J, Giordano S, Zhang J. 2012. Autophagy, mitochondria and oxidative stress: cross-talk and redox signalling. Biochem. J. 441, 523–540. ( 10.1042/BJ20111451) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Campbell RM, Scanes CG. 1995. Endocrine peptides ‘moonlighting’ as immune modulators: roles for somatostatin and GH-releasing factor. J. Endocrinol. 147, 383–396. ( 10.1677/joe.0.1470383) [DOI] [PubMed] [Google Scholar]
  • 22.Jeffery CJ. 1999. Moonlighting proteins. Trends Biochem. Sci. 24, 8–11. ( 10.1016/S0968-0004(98)01335-8) [DOI] [PubMed] [Google Scholar]
  • 23.Jeffery CJ. 2003. Moonlighting proteins: old proteins learning new tricks. Trends Genet. 19, 415–417. ( 10.1016/S0168-9525(03)00167-7) [DOI] [PubMed] [Google Scholar]
  • 24.Jeffery CJ. 2009. Moonlighting proteins - an update. Mol. Biosyst. 5, 345–350. ( 10.1039/b900658n) [DOI] [PubMed] [Google Scholar]
  • 25.Henderson B, Pockley AG. 2010. Molecular chaperones and protein folding catalysts as intercellular signaling regulators in immunity and inflammation. J. Leuk. Biol. 88, 445–462. ( 10.1189/jlb.1209779) [DOI] [PubMed] [Google Scholar]
  • 26.Jeffery CJ. 2017. Protein moonlighting: what is it, and why is it important? Phil. Trans. R. Soc. B 373, 20160523 ( 10.1098/rstb.2016.0523) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hightower LE, Guidon PT. 1989. Selective release from cultured mammalian cells of heat-shock (stress) proteins that resemble glia-axon transfer proteins. J. Cell Physiol. 138, 257–266. ( 10.1002/jcp.1041380206) [DOI] [PubMed] [Google Scholar]
  • 28.Tytell M, Greenberg SG, Lasek RJ. 1986. Heat shock-like protein is transferred from glia to axon. Brain Res. 363, 161–164. ( 10.1016/0006-8993(86)90671-2) [DOI] [PubMed] [Google Scholar]
  • 29.Pockley AG, Bulmer J, Hanks BM, Wright BH. 1999. Identification of human heat shock protein 60 (Hsp60) and anti-Hsp60 antibodies in the peripheral circulation of normal individuals. Cell Stress Chaperones 4, 29–35. ( 10.1379/1466-1268(1999)004%3C0029:IOHHSP%3E2.3.CO;2) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pockley AG, Shepherd J, Corton J. 1998. Detection of heat shock protein 70 (Hsp70) and anti-Hsp70 antibodies in the serum of normal individuals. Immunol. Invest. 27, 367–377. ( 10.3109/08820139809022710) [DOI] [PubMed] [Google Scholar]
  • 31.Chimini C, Rubartelli A. 2005. Novel pathways of protein secretion. In Molecular chaperones and cell signalling (eds Henderson B, Pockley AG), pp. 45–60. New York, NY: Cambridge University Press. [Google Scholar]
  • 32.Pockley AG, Multhoff G. 2008. Cell stress proteins in extracellular fluids: friend or foe? Novartis Found Symp. 291, 86–95; discussion 6–100, 37–40 ( 10.1002/9780470754030.ch7) [DOI] [PubMed] [Google Scholar]
  • 33.Gastpar R, Gehrmann M, Bausero MA, Asea A, Gross C, Schroeder JA, Multhoff G. 2005. Heat shock protein 70 surface-positive tumor exosomes stimulate migratory and cytolytic activity of natural killer cells. Cancer Res. 65, 5238–5247. ( 10.1158/0008-5472.CAN-04-3804) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bausero MA, Gastpar R, Multhoff G, Asea A. 2005. Alternative mechanism by which IFN-g enhances tumor recognition: active release of heat shock protein 72. J. Immunol. 175, 2900–2912. ( 10.4049/jimmunol.175.5.2900) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mambula SS, Calderwood SK. 2006. Heat shock protein 70 is secreted from tumor cells by a nonclassical pathway involving lysosomal endosomes. J. Immunol. 177, 7849–7857. ( 10.4049/jimmunol.177.11.7849) [DOI] [PubMed] [Google Scholar]
  • 36.Broquet AH, Thomas G, Masliah J, Trugnan G, Bachelet M. 2003. Expression of the molecular chaperone Hsp70 in detergent-resistant microdomains correlates with its membrane delivery and release. J. Biol. Chem. 278, 21 601–21 606. ( 10.1074/jbc.M302326200) [DOI] [PubMed] [Google Scholar]
  • 37.Evdonin AL, Martynova MG, Bystrova OA, Guzhova IV, Margulis BA, Medvedeva ND. 2006. The release of Hsp70 from A431 carcinoma cells is mediated by secretory-like granules. Eur. J. Cell Biol. 85, 443–455. ( 10.1016/j.ejcb.2006.02.008) [DOI] [PubMed] [Google Scholar]
  • 38.Evdonin AL, Guzhova IV, Margulis BA, Medvedeva ND. 2004. Phospholipase C inhibitor, U73122, stimulates release of hsp-70 stress protein from A431 human carcinoma cells. Cancer Cell Int. 4, 2 ( 10.1186/1475-2867-4-2) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Breuninger S, et al. 2014. Quantitative analysis of liposomal Heat Shock Protein 70 (Hsp70) in the blood of tumor patients using a novel Liphsp70 ELISA. J. Clin. Cell Immunol. 5, 264 ( 10.4172/2155-9899.1000264) [DOI] [Google Scholar]
  • 40.Théry C, Boussac M, Véron P, Ricciardi-Castagnoli P, Raposo G, Garin G, Amigorena S. 2001. Proteomic analysis of dendritic cell-derived exosomes: a secreted subcellular compartment distinct from apoptotic vesicles. J. Immunol. 166, 7309–7318. ( 10.4049/jimmunol.166.12.7309) [DOI] [PubMed] [Google Scholar]
  • 41.Denzer K, Kleijmeer MJ, Heijnen HF, Stoorvogel W, Geuze HJ. 2000. Exosome: from internal vesicle of the multivesicular body to intercellular signalling device. J. Cell Sci. 113, 3365–3374. [DOI] [PubMed] [Google Scholar]
  • 42.Théry C, Zitvogel L, Amigorena S. 2002. Exosomes: composition, biogenesis and function. Nat. Rev. Immunol. 2, 569–579. [DOI] [PubMed] [Google Scholar]
  • 43.Chaput N, Taïeb J, Schartz NE, Andre F, Angevin E, Zitvogel L. 2004. Exosome-based immunotherapy. Cancer Immunol. Immunother. 53, 234–239. ( 10.1007/s00262-003-0472-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Mathew A, Bell A, Johnstone RM. 1995. Hsp-70 is closely associated with the transferrin receptor in exosomes from maturing reticulocytes. Biochem. J. 308, 823–830. ( 10.1042/bj3080823) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Frostegård J, Pockley AG. 2005. Heat shock protein release and naturally-occurring, exogenous heat shock proteins. In Molecular chaperones and cell signalling (eds Henderson B, Pockley AG), pp. 195–219. New York, NY: Cambridge University Press. [Google Scholar]
  • 46.Cordonnier M, Chanteloup G, Isambert N, Seigneuric R, Fumoleau P, Garrido C, Gobbo J. 2017. Exosomes in cancer theranostic: diamonds in the rough. Cell Adhes. Migr. 11, 151–163. ( 10.1080/19336918.2016.1250999) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Shevtsov M, Huile G, Multhoff G. 2017. Membrane heat shock protein 70: a theranostic target for cancer therapy. Phil. Trans. R. Soc. B 373, 20160526 ( 10.1098/rstb.2016.0526) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Morton H, Rolfe B, Clunie GJ. 1977. An early pregnancy factor detected in human serum by the rosette inhibition test. Lancet 1, 394–397. ( 10.1016/S0140-6736(77)92605-8) [DOI] [PubMed] [Google Scholar]
  • 49.Noonan FP, Halliday WJ, Morton H, Clunie GJ. 1979. Early pregnancy factor is immunosuppressive. Nature 278, 649–651. ( 10.1038/278649a0) [DOI] [PubMed] [Google Scholar]
  • 50.Cavanagh AC, Morton H. 1994. The purification of early-pregnancy factor to homogeneity from human platelets and identification as chaperonin 10. Eur. J. Biochem. 222, 551–560. ( 10.1111/j.1432-1033.1994.tb18897.x) [DOI] [PubMed] [Google Scholar]
  • 51.Richardson A, Landry SJ, Georgopoulos C. 1998. The ins and outs of a molecular chaperone machine. Trends Biochem. Sci. 23, 138–143. ( 10.1016/S0968-0004(98)01193-1) [DOI] [PubMed] [Google Scholar]
  • 52.Shamaei-Tousi A, D'Aiuto F, Nibali L, Steptoe A, Coates AR, Parkar M, Donos N, Henderson B, Pockley A. 2007. Differential regulation of circulating levels of molecular chaperones in patients undergoing treatment for periodontal disease. PLoS ONE 2, e1198 ( 10.1371/journal.pone.0001198) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Johnson BJ, et al. 2005. Heat shock protein 10 inhibits lipopolysaccharide-induced inflammatory mediator production. J. Biol. Chem. 280, 4037–4047. ( 10.1074/jbc.M411569200) [DOI] [PubMed] [Google Scholar]
  • 54.Vanags D, Williams B, Johnson B, Hall S, Nash P, Taylor A, Weiss J, Feeney D. 2006. Therapeutic efficacy and safety of chaperonin 10 in patients with rheumatoid arthritis: a double-blind randomised trial. Lancet 368, 855–863. ( 10.1016/S0140-6736(06)69210-6) [DOI] [PubMed] [Google Scholar]
  • 55.van Eden W. 2008. XToll, a recombinant chaperonin 10 as an anti-inflammatory immunomodulator. Curr. Opin Investig. Drugs 9, 523–533. [PubMed] [Google Scholar]
  • 56.Williams B, Vanags D, Hall S, McCormack C, Foley P, Weiss J, Johnson B, Latz E, Feeney D. 2008. Efficacy and safety of chaperonin 10 in patients with moderate to severe plaque psoriasis: evidence of utility beyond a single indication. Arch. Dermatol. 144, 683–685. ( 10.1001/archderm.144.5.683) [DOI] [PubMed] [Google Scholar]
  • 57.Broadley SA, et al. 2009. Results of a phase IIa clinical trial of an anti-inflammatory molecule, chaperonin 10, in multiple sclerosis. Mult. Scler. 15, 329–336. ( 10.1177/1352458508099141) [DOI] [PubMed] [Google Scholar]
  • 58.Kol A, Lichtman AH, Finberg RW, Libby P, Kurt-Jones EA. 2000. Heat shock protein (HSP) 60 activates the innate immune response: CD14 is an essential receptor for HSP60 activation of mononuclear cells. J. Immunol. 164, 13–17. ( 10.4049/jimmunol.164.1.13) [DOI] [PubMed] [Google Scholar]
  • 59.Chen W, Syldath U, Bellmann K, Burkart V, Kold H. 1999. Human 60-kDa heat-shock protein: A danger signal to the innate immune system. J. Immunol. 162, 3212–3219. [PubMed] [Google Scholar]
  • 60.Flohé SB, Bruggemann J, Lendemans S, Nikulina M, Meierhoff G, Flohé S, Kolb H. 2003. Human heat shock protein 60 induces maturation of dendritic cells versus a Th1-promoting phenotype. J. Immunol. 170, 2340–2348. ( 10.4049/jimmunol.170.5.2340) [DOI] [PubMed] [Google Scholar]
  • 61.Asea A, Kraeft S-K, Kurt-Jones EA, Stevenson MA, Chen LB, Finberg RW, Koo GC. 2000. Hsp70 stimulates cytokine production through a CD14-dependent pathway, demonstrating its dual role as a chaperone and cytokine. Nat. Med. 6, 435–442. ( 10.1038/74697) [DOI] [PubMed] [Google Scholar]
  • 62.Asea A, Rehli M, Kabingu E, Boch JA, Baré O, Auron PE, Stevenson MA, Calderwood SK. 2002. Novel signal transduction pathway utilized by extracellular HSP70. Role of Toll-like receptor (TLR) 2 and TLR4. J. Biol. Chem. 277, 15 028–15 034. ( 10.1074/jbc.M200497200) [DOI] [PubMed] [Google Scholar]
  • 63.Gao B, Tsan MF. 2003. Endotoxin contamination in recombinant human Hsp70 preparation is responsible for the induction of TNFa release by murine macrophages. J. Biol. Chem. 278, 174–179. ( 10.1074/jbc.M208742200) [DOI] [PubMed] [Google Scholar]
  • 64.Gao B, Tsan MF. 2003. Recombinant human heat shock protein 60 does not induce the release of tumor necrosis factor a from murine macrophages. J. Biol. Chem. 278, 22 523–22 529. ( 10.1074/jbc.M303161200) [DOI] [PubMed] [Google Scholar]
  • 65.Gao B, Tsan MF. 2004. Induction of cytokines by heat shock proteins and endotoxin in murine macrophages. Biochem. Biophys. Res. Commun. 317, 1149–1154. ( 10.1016/j.bbrc.2004.03.160) [DOI] [PubMed] [Google Scholar]
  • 66.Tsan MF, Gao B. 2009. Heat shock proteins and immune system. J. Leukoc. Biol. 85, 905–910. ( 10.1189/jlb.0109005) [DOI] [PubMed] [Google Scholar]
  • 67.Henderson B, Calderwood SK, Coates AR, Cohen I, van Eden W, Lehner T, Pockley AG. 2010. Caught with their PAMPs down? The extracellular signalling actions of molecular chaperones are not due to microbial contaminants. Cell Stress Chaperones 15, 123–141. ( 10.1007/s12192-009-0137-6) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pockley AG, Muthana M, Calderwood SK. 2008. The dual immunoregulatory role of stress proteins. Trends Biochem. Sci. 3, 71–79. ( 10.1016/j.tibs.2007.10.005) [DOI] [PubMed] [Google Scholar]
  • 69.Multhoff G, Pockley AG, Schmid TE, Schilling D. 2015. The role of heat shock protein 70 (Hsp70) in radiation-induced immunomodulation. Cancer Lett. 368, 179–184. ( 10.1016/j.canlet.2015.02.013) [DOI] [PubMed] [Google Scholar]
  • 70.van Eden W, van der Zee R, Prakken B. 2005. Heat shock proteins induce T-cell regulation of chronic inflammation. Nat. Immunol. 5, 318–330. ( 10.1038/nri1593) [DOI] [PubMed] [Google Scholar]
  • 71.Kingston AE, Hicks CA, Colston MJ, Billingham MEJ. 1996. A 71-kD heat shock protein (hsp) from Mycobacterium tuberculosis has modulatory effects on experimental rat arthritis. Clin. Exp. Immunol. 103, 77–82. ( 10.1046/j.1365-2249.1996.929628.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tanaka S, et al. 1999. Activation of T cells recognizing an epitope of heat-shock protein 70 can protect against rat adjuvant arthritis. J. Immunol. 163, 5560–5565. [PubMed] [Google Scholar]
  • 73.Wendling U, Paul L, van der Zee R, Prakken B, Singh M, van Eden W. 2000. A conserved mycobacterial heat shock protein (hsp) 70 sequence prevents adjuvant arthritis upon nasal administration and induces IL-10-producing T cells that cross-react with the mammalian self-hsp70 homologue. J. Immunol. 164, 2711–2717. ( 10.4049/jimmunol.164.5.2711) [DOI] [PubMed] [Google Scholar]
  • 74.Quintana FJ, Carmi P, Mor F, Cohen IR. 2004. Inhibition of adjuvant-induced arthritis by DNA vaccination with the 70-kd or the 90-kd human heat-shock protein: immune cross-regulation with the 60-kd heat-shock protein. Arthritis Rheum. 50, 3712 ( 10.1002/art.20635) [DOI] [PubMed] [Google Scholar]
  • 75.Quintana FJ, Carmi P, Mor F, Cohen IR. 2003. DNA fragments of the human 60-kDa heat shock protein (HSP60) vaccinate against adjuvant arthritis: identification of a regulatory HSP60 peptide. J. Immunol. 171, 3533–3541. ( 10.4049/jimmunol.171.7.3533) [DOI] [PubMed] [Google Scholar]
  • 76.Anderton SM, van der Zee R, Prakken B, Noordzij A, van Eden W. 1995. Activation of T cells recognizing self 60-kD heat shock protein can protect against experimental arthritis. J. Exp. Med. 181, 943–952. ( 10.1084/jem.181.3.943) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Zanin-Zhorov A, Cahalon L, Tal G, Margalit R, Lider O, Cohen IR. 2006. Heat shock protein 60 enhances CD4+CD25+ regulatory T cell function via innate TLR2 signaling. J. Clin. Invest. 116, 2022–2032. ( 10.1172/JCI28423) [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 78.Birk OS, Gur SL, Elias D, Margalit R, Mor F, Carmi P, Bockova J, Altmann DM, Cohen IR. 1999. The 60-kDa heat shock protein modulates allograft rejection. Proc. Natl Acad. Sci. USA 96, 5159–5163. ( 10.1073/pnas.96.9.5159) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Luna E, Postol E, Caldas C, Benvenuti LA, Rodrigues JM Jr, Lima K, Kalil J, Coelho V. 2007. Treatment with encapsulated Hsp60 peptide (p277) prolongs skin graft survival in a murine model of minor antigen disparity. Scand. J. Immunol. 66, 62–70. ( 10.1111/j.1365-3083.2007.01951.x) [DOI] [PubMed] [Google Scholar]
  • 80.van Eden W, van Herwijnen M, Wagenaar J, van Kooten P, Broere F, van der Zee R. 2013. Stress proteins are used by the immune system for cognate interactions with anti-inflammatory regulatory T cells. FEBS Lett. 587, 1951–1958. ( 10.1016/j.febslet.2013.05.024) [DOI] [PubMed] [Google Scholar]
  • 81.Kim EY, Durai M, Mia Y, Kim HR, Moudgil KD. 2016. Modulation of adjuvant arthritis by cellular and humoral immunity to Hsp65. Front. Immunol. 7, 203 ( 10.3389/fimmu.2016.00203) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Spierings J, van Eden W. 2017. Heat shock proteins and their immunomodulatory role in inflammatory arthritis. Rheumatology 56, 198–208. ( 10.1093/rheumatology/kew266) [DOI] [PubMed] [Google Scholar]
  • 83.van Eden W. 2017. Immune tolerance therapies for autoimmune diseases based on heat shock protein T-cell epitopes. Phil. Trans. R. Soc. B 373, 20160531 ( 10.1098/rstb.2016.0531) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Quintana FJ, Cohen IR. 2011. The HSP60 immune system network. Trends Immunol. 32, 89–95. ( 10.1016/j.it.2010.11.001) [DOI] [PubMed] [Google Scholar]
  • 85.Lebherz-Eichinger D, et al. 2012. HSP27 and HSP70 serum and urine levels in patients suffering from chronic kidney disease. Clin. Chim. Acta. 413, 282–286. ( 10.1016/j.cca.2011.10.010) [DOI] [PubMed] [Google Scholar]
  • 86.Gruden G, et al. 2008. Serum heat shock protein 27 and diabetes complications in the EURODIAB prospective complications study: a novel circulating marker for diabetic neuropathy. Diabetes 57, 1966–1970. ( 10.2337/db08-0009) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Liu S, Iskandar R, Chen W, Zhang J, Wang Y, Chen X, Xiang F. 2016. Soluble Glycoprotein 130 and Heat Shock Protein 27 as novel candidate biomarkers of chronic heart failure with preserved ejection fraction. Heart Lung Circ. 25, 1000–1006. ( 10.1016/j.hlc.2016.02.011) [DOI] [PubMed] [Google Scholar]
  • 88.Lewthwaite J, Owen N, Coates A, Henderson B, Steptoe A. 2002. Circulating human heat shock protein 60 in the plasma of British civil servants. Circulation 106, 196–201. ( 10.1161/01.CIR.0000021121.26290.2C) [DOI] [PubMed] [Google Scholar]
  • 89.Pockley AG, Wu R, Lemne C, Kiessling R, de Faire U, Frostegård J. 2000. Circulating heat shock protein 60 is associated with early cardiovascular disease. Hypertension 36, 303–307. ( 10.1161/01.HYP.36.2.303) [DOI] [PubMed] [Google Scholar]
  • 90.Pockley AG, de Faire U, Kiessling R, Lemne C, Thulin T, Frostegård J. 2002. Circulating heat shock protein and heat shock protein antibody levels in established hypertension. J. Hypertension 20, 1815–1820. ( 10.1097/00004872-200209000-00027) [DOI] [PubMed] [Google Scholar]
  • 91.Wright BH, Corton J, El-Nahas AM, Wood RFM, Pockley AG. 2000. Elevated levels of circulating heat shock protein 70 (Hsp70) in peripheral and renal vascular disease. Heart Vessels 15, 18–22. ( 10.1007/s003800070043) [DOI] [PubMed] [Google Scholar]
  • 92.Xu Q, Schett G, Perschinka H, Mayr M, Egger G, Oberhollenzer F, Willeit J, Kiechl S, Wick G. 2000. Serum soluble heat shock protein 60 is elevated in subjects with atherosclerosis in a general population. Circulation 102, 14–20. ( 10.1161/01.CIR.102.1.14) [DOI] [PubMed] [Google Scholar]
  • 93.Schett G, et al. 1999. Myocardial injury leads to a release of heat shock protein (hsp) 60 and a suppression of the anti-hsp65 immune response. Cardiovasc. Res. 42, 685–695. ( 10.1016/S0008-6363(99)00012-7) [DOI] [PubMed] [Google Scholar]
  • 94.Nahas EA, Nahas-Neto J, Orsatti CL, Tardivo AP, Uemura G, Peracoli MT, Witkin SS. 2014. The 60- and 70-kDa heat-shock proteins and their correlation with cardiovascular risk factors in postmenopausal women with metabolic syndrome. Cell Stress Chaperones 19, 559–568. ( 10.1007/s12192-013-0483-2) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Halcox JPJ, et al. 2005. Circulating human heat shock protein 60 in the blood of healthy teenagers: A novel determinant of endothelial dysfunction and early vascular injury. Arterioscler. Thromb. Vasc. Biol. 25, e141 ( 10.1161/01.ATV.0000185832.34992.ff) [DOI] [PubMed] [Google Scholar]
  • 96.Shamaei-Tousi A, Stephens JW, Bin R, Cooper JA, Steptoe A, Coates AR, Henderson B, Humphries SE. 2006. Association between plasma levels of heat shock protein 60 and cardiovascular disease in patients with diabetes mellitus. Eur. Heart J. 27, 1565–1570. ( 10.1093/eurheartj/ehl081) [DOI] [PubMed] [Google Scholar]
  • 97.Anraku I, Rajasuriar R, Dobbin C, Brown R, Lewin SR, Suhrbier A. 2012. Circulating heat shock protein 60 levels are elevated in HIV patients and are reduced by anti-retroviral therapy. PLoS ONE 7, e45291 ( 10.1371/journal.pone.0045291) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Rizzo M, et al. 2012. Heat-shock protein 60 kDa and atherogenic dyslipidemia in patients with untreated mild periodontitis: a pilot study. Cell Stress Chaperones 17, 399–407. ( 10.1007/s12192-011-0315-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Wu CT, Ou LS, Yeh KW, Lee WI, Huang JL. 2011. Serum heat shock protein 60 can predict remission of flare-up in juvenile idiopathic arthritis. Clin. Rheumatol. 30, 959–965. ( 10.1007/s10067-011-1709-2) [DOI] [PubMed] [Google Scholar]
  • 100.Kimura F, Itoh H, Ambiru S, Shimizu H, Togawa A, Yoshidome H, Kato A, Nukui Y, Miyazaki M. 2004. Circulating heat-shock protein 70 is associated with postoperative infection and organ dysfunction after liver resection. Am. J. Surg. 187, 777–784. ( 10.1016/j.amjsurg.2003.08.029) [DOI] [PubMed] [Google Scholar]
  • 101.Dybdahl B, et al. 2002. Inflammatory response after open heart surgery: release of heat-shock protein 70 and signaling through toll-like receptor-4. Circulation 105, 685–690. ( 10.1161/hc0602.103617) [DOI] [PubMed] [Google Scholar]
  • 102.Pockley AG, Georgiades A, Thulin T, de Faire U, Frostegård J. 2003. Serum heat shock protein 70 levels predict the development of atherosclerosis in subjects with established hypertension. Hypertension 42, 235–238. ( 10.1161/01.HYP.0000086522.13672.23) [DOI] [PubMed] [Google Scholar]
  • 103.Zhu J, et al. 2003. Increased serum levels of heat shock protein 70 are associated with low risk of coronary artery disease. Arterioscler. Thromb. Vasc. Biol. 23, 1055–1059. ( 10.1161/01.ATV.0000074899.60898.FD) [DOI] [PubMed] [Google Scholar]
  • 104.Kornej J, Reinhardt C, Kosiuk J, Arya A, Hindricks G, Adams V, Husser D, Bollmann A. 2013. Response of circulating heat shock protein 70 and anti-heat shock protein 70 antibodies to catheter ablation of atrial fibrillation. J. Transl. Med. 11, 49 ( 10.1186/1479-5876-11-49) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Krepuska M, et al. 2011. Serum level of soluble Hsp70 is associated with vascular calcification. Cell Stress Chaperones 16, 257–265. ( 10.1007/s12192-010-0237-3) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Njemini R, Smitz J, Demanet C, Sosso M, Mets T. 2011. Circulating heat shock protein 70 (Hsp70) in elderly members of a rural population from Cameroon: association with infection and nutrition. Arch. Gerontol. Geriatr. 53, 359–363. ( 10.1016/j.archger.2011.01.005) [DOI] [PubMed] [Google Scholar]
  • 107.Njemini R, Lambert M, Demanet C, Mets T. 2003. Elevated serum heat-shock protein 70 levels in patients with acute infection: use of an optimized enzyme-linked immunosorbent assay. Scand. J. Immunol. 58, 664–669. ( 10.1111/j.1365-3083.2003.01341.x) [DOI] [PubMed] [Google Scholar]
  • 108.Gehrmann M, Cervello M, Montalto G, Cappello F, Gulino A, Knape C, Specht HM, Multhoff G. 2014. Heat shock protein 70 serum levels differ significantly in patients with chronic hepatitis, liver cirrhosis, and hepatocellular carcinoma. Front. Immunol. 5, 307 ( 10.3389/fimmu.2014.00307) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Molvarec A, Rigo J Jr, Nagy B, Walentin S, Szalay J, Fust G, Karádi I, Prohászka Z. 2007. Serum heat shock protein 70 levels are decreased in normal human pregnancy. J. Reprod. Immunol. 74, 163–169. ( 10.1016/j.jri.2006.12.002) [DOI] [PubMed] [Google Scholar]
  • 110.Molvarec A, Prohászka Z, Nagy B, Szalay J, Fust G, Karadi I, Rigó J. 2006. Association of elevated serum heat-shock protein 70 concentration with transient hypertension of pregnancy, preeclampsia and superimposed preeclampsia: a case-control study. J. Hum. Hypertens. 20, 780–786. ( 10.1038/sj.jhh.1002060) [DOI] [PubMed] [Google Scholar]
  • 111.Molvarec A, Szarka A, Walentin S, Beko G, Karadi I, Prohaszka Z, Rigó J. 2011. Serum heat shock protein 70 levels in relation to circulating cytokines, chemokines, adhesion molecules and angiogenic factors in women with preeclampsia. Clin. Chim. Acta. 412, 1957–1962. ( 10.1016/j.cca.2011.06.042) [DOI] [PubMed] [Google Scholar]
  • 112.Hou C, et al. 2011. Increased heat shock protein 70 levels in induced sputum and plasma correlate with severity of asthma patients. Cell Stress Chaperones 16, 663–671. ( 10.1007/s12192-011-0271-9) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Morteza A, Nakhjavani M, Larry M, Nargesi AA, Esteghamati A. 2013. Heat shock protein 70 and albuminuria in patients with type 2 diabetes: a matched case control study. Cell Stress Chaperones 18, 815–819. ( 10.1007/s12192-013-0435-x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Nakhjavani M, Morteza A, Khajeali L, Esteghamati A, Khalilzadeh O, Asgarani F. 2010. Increased serum HSP70 levels are associated with the duration of diabetes. Cell Stress Chaperones 15, 959–964. ( 10.1007/s12192-010-0204-z) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Nakhjavani M, Morteza A, Meysamie A, Esteghamati A, Khalilzadeh O, Esfahanian F, Khajeali L, Feiz F. 2011. Serum heat shock protein 70 and oxidized LDL in patients with type 2 diabetes: does sex matter?. Cell Stress Chaperones 16, 195–201. ( 10.1007/s12192-010-0232-8) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Nakhjavani M, Morteza A, Asgarani F, Khalilzadeh O, Ghazizadeh Z, Bathaie SZ, Esteghamati A. 2012. The dual behavior of heat shock protein 70 and asymmetric dimethylarginine in relation to serum CRP levels in type 2 diabetes. Gene 498, 107–111. ( 10.1016/j.gene.2012.01.085) [DOI] [PubMed] [Google Scholar]
  • 117.Oglesbee MJ, Herdman AV, Passmore GG, Hoffman WH. 2005. Diabetic ketoacidosis increases extracellular levels of the major inducible 70-kDa heat shock protein. Clin. Biochem. 38, 900–904. ( 10.1016/j.clinbiochem.2005.05.011) [DOI] [PubMed] [Google Scholar]
  • 118.Garamvölgyi Z, Prohászka Z, Rigó J Jr, Kecskeméti A, Molvarec A. 2015. Increased circulating heat shock protein 70 (HSPA1A) levels in gestational diabetes mellitus: a pilot study. Cell Stress Chaperones 20, 575–581. ( 10.1007/s12192-015-0579-y) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Pagetta A, Folda A, Brunati AM, Finotti P. 2003. Identification and purification from the plasma of Type 1 diabetic subjects of a proteolytically active Grp94. Evidence that Grp94 is entirely responsible for plasma proteolytic activity. Diabetologia 46, 996–1006. ( 10.1007/s00125-003-1133-5) [DOI] [PubMed] [Google Scholar]
  • 120.Finotti P, Pagetta A. 2004. A heat shock protein 70 fusion protein with a1-antitrypsin in plasma of Type 1 diabetic subjects. Biochem. Biophys. Res. Comm. 315, 297–305. ( 10.1016/j.bbrc.2004.01.058) [DOI] [PubMed] [Google Scholar]
  • 121.Zhao M, Ding JX, Zeng K, Zhao J, Shen F, Yin YX, Chen Q. 2014. Heat shock protein 27: a potential biomarker of peritoneal metastasis in epithelial ovarian cancer? Tumour Biol. 35, 1051–1056. ( 10.1007/s13277-013-1139-7) [DOI] [PubMed] [Google Scholar]
  • 122.Liebhardt S, et al. 2010. CEA-, Her2/neu-, BCRP- and Hsp27-positive microparticles in breast cancer patients. Anticancer Res. 30, 1707–1712. [PubMed] [Google Scholar]
  • 123.Zimmermann M, et al. 2012. Discrimination of clinical stages in non-small cell lung cancer patients by serum HSP27 and HSP70: a multi-institutional case-control study. Clin. Chim. Acta. 413, 1115–1120. ( 10.1016/j.cca.2012.03.008) [DOI] [PubMed] [Google Scholar]
  • 124.Yeh CH, Tseng R, Hannah A, Estrov Z, Estey E, Kantarjian H, Albitar M. 2010. Clinical correlation of circulating heat shock protein 70 in acute leukemia. Leuk. Res. 34, 605–609. ( 10.1016/j.leukres.2009.09.014) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Yeh CH, et al. 2009. Circulating heat shock protein 70 and progression in patients with chronic myeloid leukemia. Leuk. Res. 33, 212–217. ( 10.1016/j.leukres.2008.07.012) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rozenberg P, Kocsis J, Saar M, Prohászka Z, Fust G, Fishelson Z. 2013. Elevated levels of mitochondrial mortalin and cytosolic HSP70 in blood as risk factors in patients with colorectal cancer. Int. J. Cancer 133, 514–518. ( 10.1002/ijc.28029) [DOI] [PubMed] [Google Scholar]
  • 127.Kocsis J, Madaras B, Toth EK, Fust G, Prohászka Z. 2010. Serum level of soluble 70-kD heat shock protein is associated with high mortality in patients with colorectal cancer without distant metastasis. Cell Stress Chaperones 15, 143–151. ( 10.1007/s12192-009-0128-7) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Bayer C, et al. 2014. Validation of heat shock protein 70 as a tumor-specific biomarker for monitoring the outcome of radiation therapy in tumor mouse models. Int. J. Radiat. Oncol. Biol. Phys. 88, 694–700. ( 10.1016/j.ijrobp.2013.11.008) [DOI] [PubMed] [Google Scholar]
  • 129.Gehrmann M, et al. 2014. Hsp70 - a biomarker for tumor detection and monitoring of outcome of radiation therapy in patients with squamous cell carcinoma of the head and neck. Radiat. Oncol. 9, 131 ( 10.1186/1748-717X-9-131) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Dutta SK, Girotra M, Singla M, Dutta A, Otis Stephen F, Nair PP, Merchant NB. 2012. Serum HSP70: a novel biomarker for early detection of pancreatic cancer. Pancreas 41, 530–534. ( 10.1097/MPA.0b013e3182374ace) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Pfister K, et al. 2007. Patient survival by Hsp70 membrane phenotype: association with different routes of metastasis. Cancer 110, 926–935. ( 10.1002/cncr.22864) [DOI] [PubMed] [Google Scholar]
  • 132.Multhoff G, Hightower LE. 2011. Distinguishing integral and receptor-bound heat shock protein 70 (Hsp70) on the cell surface by Hsp70-specific antibodies. Cell Stress Chaperones 16, 251–255. ( 10.1007/s12192-010-0247-1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Njemini R, Demanet C, Mets T. 2005. Comparison of two ELISAs for the determination of Hsp70 in serum. J. Immunol. Methods 306, 176–182. ( 10.1016/j.jim.2005.08.012) [DOI] [PubMed] [Google Scholar]
  • 134.Zimmermann M, et al. 2014. Circulating heat shock protein 27 as a biomarker for the differentiation of patients with lung cancer and healthy controls - a clinical comparison of different enzyme linked immunosorbent assays. Clin. Lab. 60, 999–1006. ( 10.7754/Clin.Lab.2013.130526) [DOI] [PubMed] [Google Scholar]
  • 135.Pockley AG, Frostegård J. 2005. Heat shock proteins in cardiovascular disease and the prognostic value of heat shock protein-related measurements. Heart 91, 1124–1126. ( 10.1136/hrt.2004.059220) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Anderson CF, Lucas M, Gutiérrez-Kobeh L, Field AE, Mosser DM. 2004. T cell biasing by activated dendritic cells. J. Immunol. 173, 955–961. ( 10.4049/jimmunol.173.2.955) [DOI] [PubMed] [Google Scholar]
  • 137.Tramentozzi E, et al. 2016. Grp94 in complexes with IgG is a soluble diagnostic marker of gastrointestinal tumors and displays immune-stimulating activity on peripheral blood immune cells. Oncotarget 7, 72 923–72 940. ( 10.18632/oncotarget.12141) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Dakappagari N, Neely L, Tangri S, Lundgren K, Hipolito L, Estrellado A, Burrows F, Zhang H. 2010. An investigation into the potential use of serum Hsp70 as a novel tumour biomarker for Hsp90 inhibitors. Biomarkers 15, 31–38. ( 10.3109/13547500903261347) [DOI] [PubMed] [Google Scholar]
  • 139.Elias D, Markovits D, Reshef T, van der Zee R, Cohen IR. 1990. Induction and therapy of autoimmune diabetes in the non-obese diabetic mouse by a 65-kDa heat shock protein. Proc. Natl Acad. Sci. USA. 87, 1576–1580. ( 10.1073/pnas.87.4.1576) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Elias D, Reshef T, Birk OS, van der Zee R, Walker MD, Cohen IR. 1991. Vaccination against autoimmune mouse diabetes with a T cell epitope of the human 65-kDa heat shock protein. Proc. Natl Acad. Sci. USA 88, 3088–3091. ( 10.1073/pnas.88.8.3088) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Tuccinardi D, Fioriti E, Manfrini S, D'Amico E, Pozzilli P. 2011. DiaPep277 peptide therapy in the context of other immune intervention trials in type 1 diabetes. Expert. Opin. Biol. Ther. 11, 1233–1240. ( 10.1517/14712598.2011.599319) [DOI] [PubMed] [Google Scholar]
  • 142.Athanasas-Platsis S, Quinn KA, Wong TY, Rolfe BE, Cavanagh AC, Morton H. 1989. Passive immunization of pregnant mice against early pregnancy factor causes loss of embryonic viability. J. Reprod. Fertil. 87, 495–502. ( 10.1530/jrf.0.0870495) [DOI] [PubMed] [Google Scholar]
  • 143.Agnello D, Scanziani E, Di GM, Leoni F, Modena D, Mascagni P, Introna M, Ghezzi P, Villa P. 2002. Preventive administration of Mycobacterium tuberculosis 10-kDa heat shock protein (hsp10) suppresses adjuvant arthritis in Lewis rats. Int. Immunopharmacol. 2, 463–474. ( 10.1016/S1567-5769(01)00188-6) [DOI] [PubMed] [Google Scholar]
  • 144.Harness J, Cavanagh A, Morton H, McCombe PA. 2003. A protective effect of early pregnancy factor on experimental autoimmune encephalomyelitis induced in Lewis rats by inoculation with myelin basic protein. J. Neurol. Sci. 216, 33–41. ( 10.1016/S0022-510X(03)00212-0) [DOI] [PubMed] [Google Scholar]
  • 145.Riffo-Vasquez Y, Spina D, Page C, Tormay P, Singh M, Henderson B. 2004. Effect of Mycobacterium tuberculosis chaperonins on bronchial eosinophilia and hyper-responsiveness in a murine model of allergic inflammation. Clin. Exp. Allergy 34, 712–719. ( 10.1111/j.1365-2222.2004.1931.x) [DOI] [PubMed] [Google Scholar]
  • 146.Ragno S, Winrow VR, Mascagni P, Lucietto P, Di Pierro F, Morris CJ. 1996. A synthetic 10-kD heat shock protein (hsp10) from Mycobacterium tuberculosis modulates adjuvant arthritis. Clin. Exp. Immunol. 103, 384–390. ( 10.1111/j.1365-2249.1996.tb08291.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Shields AM, Panayi GS, Corrigall VM. 2011. Resolution-associated molecular patterns (RAMP): RAMParts defending immunological homeostasis? Clin. Exp. Immunol. 165, 292–300. ( 10.1111/j.1365-2249.2011.04433.x) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Haas IG, Wabl M. 1983. Immunoglobulin heavy chain binding protein. Nature 306, 387–389. ( 10.1038/306387a0) [DOI] [PubMed] [Google Scholar]
  • 149.Gething MJ. 1999. Role and regulation of the ER chaperone BiP. Semin. Cell Dev. Biol. 10, 465–472. ( 10.1006/scdb.1999.0318) [DOI] [PubMed] [Google Scholar]
  • 150.Bhattacharjee G, et al. 2005. Regulation of tissue factor--mediated initiation of the coagulation cascade by cell surface grp78. Arterioscler. Thromb. Vasc. Biol. 25, 1737–1743. ( 10.1161/01.ATV.0000173419.31242.56) [DOI] [PubMed] [Google Scholar]
  • 151.Davidson DJ, et al. 2005. Kringle 5 of human plasminogen induces apoptosis of endothelial and tumor cells through surface-expressed glucose-regulated protein 78. Cancer Res. 65, 4663–4672. ( 10.1158/0008-5472.CAN-04-3426) [DOI] [PubMed] [Google Scholar]
  • 152.Misra UK, Deedwania R, Pizzo SV. 2006. Activation and cross-talk between Akt, NF-kappaB, and unfolded protein response signaling in 1-LN prostate cancer cells consequent to ligation of cell surface-associated GRP78. J. Biol. Chem. 281, 13 694–13 707. ( 10.1074/jbc.M511694200) [DOI] [PubMed] [Google Scholar]
  • 153.Corrigall VM, Bodman-Smith MD, Brunst M, Cornell H, Panayi GS. 2004. Inhibition of antigen-presenting cell function and stimulation of human peripheral blood mononuclear cells to express an antiinflammatory cytokine profile by the stress protein BiP: relevance to the treatment of inflammatory arthritis. Arthritis Rheum. 50, 1164–1171. ( 10.1002/art.20134) [DOI] [PubMed] [Google Scholar]
  • 154.Marin-Briggiler CI, Gonzalez-Echeverria MF, Munuce MJ, Ghersevich S, Caille AM, Hellman U, Corrigall VM, Vazquez-Levin MH. 2010. Glucose-regulated protein 78 (Grp78/BiP) is secreted by human oviduct epithelial cells and the recombinant protein modulates sperm-zona pellucida binding. Fertil. Steril. 93, 1574–1584. ( 10.1016/j.fertnstert.2008.12.132) [DOI] [PubMed] [Google Scholar]
  • 155.Shields AM, Panayi GS, Corrigall VM. 2012. A new-age for biologic therapies: Long-term drug-free therapy with BiP? Front. Immunol. 3, 17 ( 10.3389/fimmu.2012.00017) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Kirkham B, et al. 2016. Safety and patient response as indicated by biomarker changes to binding immunoglobulin protein in the phase I/IIA RAGULA clinical trial in rheumatoid arthritis. Rheumatology 55, 1993–2000. ( 10.1093/rheumatology/kew287) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Georgiou AS, Sostaric E, Wong CH, Snijders AP, Wright PC, Moore HD, Fazeli A. 2005. Gametes alter the oviductal secretory proteome. Mol. Cell. Proteomics 4, 1785–1796. ( 10.1074/mcp.M500119-MCP200) [DOI] [PubMed] [Google Scholar]
  • 158.Lloyd RE, Elliott RM, Fazeli A, Watson PF, Holt WV. 2009. Effects of oviductal proteins, including heat shock 70 kDa protein 8, on survival of ram spermatozoa over 48 h in vitro. Reprod. Fertil. Dev. 21, 408–418. ( 10.1071/RD08204) [DOI] [PubMed] [Google Scholar]
  • 159.Boilard M, Reyes-Moreno C, Lachance C, Massicotte L, Bailey JL, Sirard MA, Leclerc P. 2004. Localization of the chaperone proteins GRP78 and HSP60 on the luminal surface of bovine oviduct epithelial cells and their association with spermatozoa. Biol. Reprod. 71, 1879–1889. ( 10.1095/biolreprod.103.026849) [DOI] [PubMed] [Google Scholar]
  • 160.Lachance C, Bailey JL, Leclerc P. 2007. Expression of Hsp60 and Grp78 in the human endometrium and oviduct, and their effect on sperm functions. Hum. Reprod. 22, 2606–2614. ( 10.1093/humrep/dem242) [DOI] [PubMed] [Google Scholar]
  • 161.Elliott RM, Lloyd RE, Fazeli A, Sostaric E, Georgiou AS, Satake N, Watson PF, Holt WV. 2009. Effects of HSPA8, an evolutionarily conserved oviductal protein, on boar and bull spermatozoa. Reproduction 137, 191–203. ( 10.1530/REP-08-0298) [DOI] [PubMed] [Google Scholar]
  • 162.Moein-Vaziri N, et al. 2014. Heat-shock protein A8 restores sperm membrane integrity by increasing plasma membrane fluidity. Reproduction 147, 719–732. ( 10.1530/REP-13-0631) [DOI] [PubMed] [Google Scholar]
  • 163.Gething MJ, Sambrook J. 1992. Protein folding in the cell. Nature 355, 33–45. ( 10.1038/355033a0) [DOI] [PubMed] [Google Scholar]
  • 164.Young D, Romain E, Moreno C, O'Brien R, Born W. 1993. Molecular chaperones and the immune system response. Phil. Trans. R. Soc. Lond. B 339, 363–367. ( 10.1098/rstb.1993.0035) [DOI] [PubMed] [Google Scholar]
  • 165.Udono H, Srivastava PK. 1994. Comparison of tumor-specific immunogenicities of stress-induced proteins gp96, hsp90 and hsp70. J. Immunol. 152, 5398–5403. [PubMed] [Google Scholar]
  • 166.Chandawarkar RY, Wagh MS, Srivastava PK. 1999. The dual nature of specific immunological activity of tumor-derived gp96 preparations. J. Exp. Med. 189, 1437–1442. ( 10.1084/jem.189.9.1437) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Binder RJ, Srivastava PK. 2005. Peptides chaperoned by heat-shock proteins are a necessary and sufficient source of antigen in the cross-priming of CD8+ T cells. Nat. Immunol. 6, 593–599. ( 10.1038/ni1201) [DOI] [PubMed] [Google Scholar]
  • 168.Chandawarkar RY, Wagh MS, Kovalchin JT, Srivastava P. 2004. Immune modulation with high-dose heat-shock protein gp96: therapy of murine autoimmune diabetes and encephalomyelitis. Int. Immunol. 16, 615–624. ( 10.1093/intimm/dxh063) [DOI] [PubMed] [Google Scholar]
  • 169.Kovalchin JT, Mendonca C, Wagh MS, Wang R, Chandawarkar RY. 2006. In vivo treatment of mice with heat shock protein, gp96, improves survival of skin grafts with minor and major antigenic disparity. Transpl. Immunol. 15, 179–185. ( 10.1016/j.trim.2005.07.003) [DOI] [PubMed] [Google Scholar]
  • 170.Slack LK, Muthana M, Hopkinson K, Suvarna SK, Mirza S, Fairburn B, Pockley AG. 2007. Administration of the stress protein gp96 prolongs rat cardiac allograft survival, modifies rejection-associated inflammatory events and induces a state of peripheral T cell hyporesponsiveness. Cell Stress Chaperones 12, 71–82. ( 10.1379/CSC-237R.1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Mirza S, Muthana M, Fairburn B, Slack LK, Hopkinson K, Pockley AG. 2006. The stress protein gp96 is not an activator of resting rat bone marrow-derived dendritic cells, but is a co-stimulator and activator of CD3+ T cells. Cell Stress Chaperones 11, 364–378. ( 10.1379/CSC-208.1) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Kinner-Bibeau LB, Sedlacek AL, Messmer MN, Watkins SC, Binder RJ. 2017. HSPs drive dichotomous T-cell immune responses via DNA methylome remodelling in antigen presenting cells. Nat. Commun. 8, 15648 ( 10.1038/ncomms15648) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Crane CA, et al. 2013. Individual patient-specific immunity against high-grade glioma after vaccination with autologous tumor derived peptides bound to the 96 KD chaperone protein. Clin. Cancer Res. 19, 205–214. ( 10.1158/1078-0432.CCR-11-3358) [DOI] [PubMed] [Google Scholar]
  • 174.Bloch O, et al. 2014. Heat-shock protein peptide complex-96 vaccination for recurrent glioblastoma: a phase II, single-arm trial. Neuro. Oncol. 16, 274–279. ( 10.1093/neuonc/not203) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Bloch O, Lim M, Sughrue ME, Komotar RJ, Abrahams JM, O'Rourke DM, D'Ambrosio A, Bruce JN, Parsa AT. 2017. Autologous heat shock protein peptide vaccination for newly diagnosed glioblastoma: Impact of peripheral PD-L1 expression on response to therapy. Clin. Cancer Res. 23, 3575–3584. ( 10.1158/1078-0432.CCR-16-1369) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Multhoff G, Botzler C, Wiesnet M, Eissner G, Issels R. 1995. CD3 large granular lymphocytes recognize a heat-inducible immunogenic determinant associated with the 72-kD heat shock protein on human sarcoma cells. Blood 86, 1374–1382. [PubMed] [Google Scholar]
  • 177.Multhoff G, Botzler C, Wiesnet M, Muller E, Meier T, Wilmanns W. 1995. A stress-inducible 72-kDa heat-shock protein (HSP72) is expressed on the surface of human tumor cells, but not on normal cells. Int. J. Cancer 61, 272–279. ( 10.1002/ijc.2910610222) [DOI] [PubMed] [Google Scholar]
  • 178.Stangl S, et al. 2011. Targeting membrane heat-shock protein 70 (Hsp70) on tumors by cmHsp70.1 antibody. Proc. Natl Acad. Sci. USA 108, 733–738. ( 10.1073/pnas.1016065108) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Gehrmann M, Pfister K, Hutzler P, Gastpar R, Margulis B, Multhoff G. 2002. Effects of antineoplastic agents on cytoplasmic and membrane-bound heat shock protein 70 (Hsp70) levels. Biol. Chem. 383, 1715–1725. ( 10.1515/BC.2002.192) [DOI] [PubMed] [Google Scholar]
  • 180.Gehrmann M, et al. 2012. Immunotherapeutic targeting of membrane hsp70-expressing tumors using recombinant human granzyme B. PLoS ONE 7, e41341 ( 10.1371/journal.pone.0041341) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Mathivanan S, Ji H, Simpson RJ. 2010. Exosomes: extracellular organelles important in intercellular communication. J. Proteomics 73, 1907–1920. ( 10.1016/j.jprot.2010.06.006) [DOI] [PubMed] [Google Scholar]
  • 182.Elsner L, et al. 2007. The heat shock protein HSP70 promotes mouse NK cell activity against tumors that express inducible NKG2D ligands. J. Immunol. 179, 5523–5533. ( 10.4049/jimmunol.179.8.5523) [DOI] [PubMed] [Google Scholar]
  • 183.Vega VL, Rodríguez-Silva M, Frey T, Gehrmann M, Diaz JC, Steinem C, Multhoff G, Arispe N, De Maio A. 2008. Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages. J. Immunol. 180, 4299–4307. ( 10.4049/jimmunol.180.6.4299) [DOI] [PubMed] [Google Scholar]
  • 184.Vicencio JM, et al. 2015. Plasma exosomes protect the myocardium from ischemia-reperfusion injury. J. Am. Coll. Cardiol. 65, 1525–1536. ( 10.1016/j.jacc.2015.02.026) [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

This article has no additional data.


Articles from Philosophical Transactions of the Royal Society B: Biological Sciences are provided here courtesy of The Royal Society

RESOURCES