ArticlePDF Available

Small heat shock proteins, protein degradation and protein aggregation diseases

Taylor & Francis
Autophagy
Authors:

Abstract

Small heat shock proteins have been characterized in vitro as ATP-independent molecular chaperones that can prevent aggregation of un- or mis-folded proteins and assist in their refolding with the help of ATP-dependent chaperone machines (e.g., the Hsp70 proteins). Comparison of the functionality of the 10 human members of the small HSPB family in cell models now reveals that some members function entirely differently and independently from Hsp70 machines. One member, HSPB7, has strong activities to prevent toxicity of polyglutamine-containing proteins in cells and Drosophila, and seems to act by assisting the loading of misfolded proteins or small protein aggregates into autophagosomes.
www.landesbioscience.com Autophagy 101
Autophagy 7:1, 101-103; January 2011; © 2011 Landes Bioscience
AUTOPHAGIC PUNCTUM AUTOPHAGIC PUNCTUM
Punctum to: Vos MJ, Zijstra MP, Kanon B, van
Waarde-Verhagen MAWH, Brunt ERP, Oosterveld-
Hut HMJ, et al. HSPB7 is the most potent polyQ
aggregation suppressor within the HSPB family
of molecular chaperones. Hum Mol Genet 2010;
19:4677–93; PMID: 20843828; DOI: 10.1093/hmg/
ddq398.
Key words: heat shock protein, HSPB,
polyglutamine diseases, autophagy,
Hsp70
Submitted: 10 /12/10
Revised: 10 /13/10
Accepted: 10 /13/10
Previously published online:
www.landesbioscience.com/journals/
autophagy/article/13935
DOI: 10.4161/auto.7.1.13935
*Correspondence to: Harm H. Kampinga;
Email: h.h.kampinga@med.umcg.nl
Small heat shock proteins have been
characterized in vitro as ATP-
independent molecular chaperones that
can prevent aggregation of un- or mis-
folded proteins and assist in their refold-
ing with the help of ATP-dependent
chaperone machines (e.g., the Hsp70
proteins). Comparison of the functional-
ity of the 10 human members of the small
HSPB family in cell models now reveals
that some members function entirely dif-
ferently and independently from Hsp70
machines. One member, HSPB7, has
strong activities to prevent toxicity of
polyglutamine-containing proteins in
cells and Drosophila, and seems to act
by assisting the loading of misfolded
proteins or small protein aggregates into
autophagosomes.
Polyglutamine (polyQ) diseases are char-
acterized by an unstable and abnormal
expansion of a CAG repeat within the
protein-coding region of the affected
genes, resulting in an elongation of the
glutamine repeat within the protein. This
specific mutation or elongation, promotes
aggregation of the mutant protein towards
insoluble aggregates (Fig. 1) and causes
the development of neurological disorders
like Huntington disease and spinocerebel-
lar ataxias.
It remains unclear how protein aggre-
gates lead to degeneration. In fact, the for-
mation of large(r) amyloid-like inclusions
(Fig. 1) that usually hallmark these dis-
eases, likely reflect a (last) attempt of the
protein quality control system to prevent
the detrimental effects of the more toxic
Small heat shock proteins, protein degradation and protein aggregation
diseases
Michel J. Vos, Marianne P. Zijlstra, Serena Carra, Ody C.M. Sibon and Harm H. Kampinga*
Department of Cell Biology; Section of R adiation and Stress Cell Biology; University Medical Center Groningen; University of Groningen;
Groningen, The Netherlands
These authors contributed equally to this work.
intermediate-size, amorphous aggregates.
However, by using several experimental
model systems it was shown that early pre-
vention or disposal of toxic protein aggre-
gates by the protein quality control system
can ameliorate disease. In our recent paper,
we show that members of the family of
human small heat shock proteins (HSPB)
act at different steps in protein quality
control with differential potential to pre-
vent toxic aggregation of polyQ proteins.
The family of human HSPB proteins
comprises 10 members. In cell-free assays,
some of them act as ATP-independent
“holdases,” meaning that they can main-
tain unfolded or misfolded proteins in a
folding competent, nonaggregated state,
but they cannot release them actively.
The latter implies that they alone can-
not help client refolding to an active
state; here, too, they require assistance of
ATP-regulated chaperones, in particular
the Hsp70 machines. In cellular assays,
we now confirm such a holdase activity
(Fig. 1, step I) for some of these members
(HSPB1, HSPB4 and HSPB5) and show
that they support refolding of a firefly
luciferase model protein, an action for
which they indeed depend on an active
Hsp70 machine. However, this activity is
not associated with significant prevention
of aggregation of polyQ proteins. This
suggests that maintenance of the refold-
ing-competent state of polyQ proteins,
which theoretically also may increase
the time window for degradation by, for
example, the proteasome (Fig. 1, step II),
is ultimately rather ineffective in prevent-
ing polyQ aggregation and toxicity.
102 Autophagy Volume 7 Issue 1
of HSPB7 is strongly reduced in ATG5-
/- cells that are impaired in autophagy.
Unlike HSPB8, however, HSPB7 does
not cause an increase in overall autophagic
activity, meaning that the endogenous
autophagic routes are sufficiently active to
support HSPB7-mediated protection.
How HSPB7 interacts with the auto-
phagic machinery remains to be elu-
cidated. The action of HSPB7 is not
affected by ‘poisoning’ the activity of
the cellular HSP70 machinery, suggest-
ing that a client transfer model via ATP
dependent chaperones, as suggested for
HSPB8 activates eIF2alpha, hereby induc-
ing autophagy and translational inhibition
of polyQ protein expression (Fig. 1, Step
V). Comparing all members, we show that
HSPB7 is in fact the most potent inhibi-
tor of polyQ aggregation within the HSPB
family. HSPB7 reduces polyQ toxicity in
cells and can prevent eye-degeneration
in a Drosophila model for spinocerebel-
lar ataxia type 3. We show that HSPB7
does not act via supporting proteasomal
degradation of polyQ proteins or aggre-
gates. Rather, like HSPB8, HSPB7 activ-
ity is related to autophagy, as the activity
Surprisingly, a number of small HSPB
members that were tested were unable to
support refolding. Whereas we only tested
ectopically expressed heat-denatured
luciferase as a substrate, leaving open the
possibility that these members have sub-
strate-specific refolding activities, it was
striking to note that four members that
all lacked luciferase refolding activity
(HSPB6, HSPB7, HSPB8 and HSPB9)
were all able to reduce polyQ aggregation.
Of these four chaperones, only HSPB8
has previously been identified as a suppres-
sor of polyQ aggregation. Intriguingly,
Figure 1. Model for the dierential processing of misfolded or/and unfolded proteins by dierent members of the human family of small Heat Shock
Proteins (HSPB). HSPB members are presented as dynamic oligomers (HSPB1, B4, B5) and/or dimers, the presumed minimal oligomeric structure of
most small HSP. For explanation of the various steps (I–V ) see the main text.
www.landesbioscience.com Autophagy 103
HSPB member-related actions in refold-
ing or supporting proteasomal degrada-
tion, may not apply here. Also, this makes
a role of the Hsp70-dependent chaperone
mediated autophagy (CMA) unlikely. As
we show that HSPB7 can interact directly
with polyQ aggregates, and most likely
cannot actively regulate client release as
it lacks ATPase activity, a model can be
envisioned that HSPB7 may enhance rec-
ognition of polyQ proteins or aggregates
by or enclosure into, (pre)autophagosomal
vesicles (Fig. 1, step IV). Supportive for
such a model are findings showing that
HSPB7 can bind to alpha-filamin, a pro-
tein that anchors membrane proteins to
the actin cytoskeleton. In conjunction
with HDAC6—that recruits the actin
remodeling machinery, required for the
fusion of autophagosomes to lysosomes—
this would support efficient and selec-
tive degradation of aggregates without
requiring the need for autophagy induc-
tion. Such a model also would imply that
HSPB7 is co-degraded with its clients.
Consistently, we find that the levels of
ectopically expressed HSPB7 are always
much lower than those of, for example,
the folding-supporting HSPB1 or HSPB5.
The data presented in our paper thus
show two interesting implications. First,
they demonstrate that members of the
small HSPB family are functionally dis-
tinct. This goes beyond client specificity
only, since the different HSPB members
handle the same client differently. In fact,
we have now data to show that HSPB9,
one other member that suppresses polyQ
aggregation, utilizes the proteasomal
route for client degradation (Fig. 1, step
III). A future challenge will be to unravel
what specifies this differential processing
by these various members and how these
processes are controlled and integrated
into the protein quality control network.
The second implication is that nonspecific
induction of autophagy, with all poten-
tial side effects, may not be required to
enhance clearance of polyQ aggregates.
By getting more insight in how expres-
sion and function of HSPB7 is regulated,
one might be able to activate the selec-
tive clearance of harmful proteins by the
endogenous autophagy machinery.
... Direct sHsp interactions with misfolded proteins can prevent further irreversible misfolding events and facilitate client triage by other chaperones that more actively influence client fate through specific proteostasis pathways (Hartl et al., 2011;Vos et al., 2011;Ungelenk et al., 2016). Generally, HspB1, HspB4, and HspB5, which are all heat shock responsive, are commonly reported to play a role in substrate refolding (Ehrnsperger et al., 1997;Vos et al., 2010, Vos et al., 2016, while HspB6, HspB7, and HspB8 have been shown to preferentially promote the degradation of clients through autophagy (Fuchs et al., 2009;Vos et al., 2010;Vos et al., 2011;Carra et al., 2013;Vos et al., 2016). ...
... Direct sHsp interactions with misfolded proteins can prevent further irreversible misfolding events and facilitate client triage by other chaperones that more actively influence client fate through specific proteostasis pathways (Hartl et al., 2011;Vos et al., 2011;Ungelenk et al., 2016). Generally, HspB1, HspB4, and HspB5, which are all heat shock responsive, are commonly reported to play a role in substrate refolding (Ehrnsperger et al., 1997;Vos et al., 2010, Vos et al., 2016, while HspB6, HspB7, and HspB8 have been shown to preferentially promote the degradation of clients through autophagy (Fuchs et al., 2009;Vos et al., 2010;Vos et al., 2011;Carra et al., 2013;Vos et al., 2016). However, this simple categorization of HspBs does not hold true in many instances, including many examples discussed in this review. ...
Article
Full-text available
Misfolding, aggregation, and aberrant accumulation of proteins are central components in the progression of neurodegenerative disease. Cellular molecular chaperone systems modulate proteostasis, and, therefore, are primed to influence aberrant protein-induced neurotoxicity and disease progression. Molecular chaperones have a wide range of functions from facilitating proper nascent folding and refolding to degradation or sequestration of misfolded substrates. In disease states, molecular chaperones can display protective or aberrant effects, including the promotion and stabilization of toxic protein aggregates. This seems to be dependent on the aggregating protein and discrete chaperone interaction. Small heat shock proteins (sHsps) are a class of molecular chaperones that typically associate early with misfolded proteins. These interactions hold proteins in a reversible state that helps facilitate refolding or degradation by other chaperones and co-factors. These sHsp interactions require dynamic oligomerization state changes in response to diverse cellular triggers and, unlike later steps in the chaperone cascade of events, are ATP-independent. Here, we review evidence for modulation of neurodegenerative disease-relevant protein aggregation by sHsps. This includes data supporting direct physical interactions and potential roles of sHsps in the stewardship of pathological protein aggregates in brain. A greater understanding of the mechanisms of sHsp chaperone activity may help in the development of novel therapeutic strategies to modulate the aggregation of pathological, amyloidogenic proteins. sHsps-targeting strategies including modulators of expression or post-translational modification of endogenous sHsps, small molecules targeted to sHsp domains, and delivery of engineered molecular chaperones, are also discussed.
... However, HSPB7 differs from other autophagy related suppressors like Alfy, as it cannot act on preformed aggregates to fullfill its function (15). One model that explains this finding predicts that HSPB7 is present during aggregate formation and binds these protofibrils to maintain early aggregates in a state competent for processing by the autophagic machinery (16). ...
... Alternatively early formed protofibrils are stabilized by HSPB7 to inhibit the rate of the aggregation process (41,42). Irrespective of either one of these mechnisms, aggregate growth is delayed which may facilitate their engulfment into autophagosomes to facilitate aggrephagy (8,15,16). ...
Article
Full-text available
Heat shock protein family B (small) member 7 (HSPB7) is a unique, relatively unexplored member within the family of human small heat shock proteins (HSPBs). Unlike most HSPB family members, HSPB7 does not oligomerize and so far has not been shown to associate with any other member of the HSPB family. Intriguingly, it was found to be the most potent member within the HSPB family to prevent aggregation of proteins with expanded polyglutamine (polyQ) stretches. How HSPB7 suppresses polyQ aggregation has remained elusive so far. Here, using several experimental strategies, including in vitroaggregation assays and immunoblotting and -fluorescence approaches, we show that the polyQ aggregation-inhibiting activity of HSPB7 is fully dependent on its flexible N-terminal domain (NTD). We observed that the NTD of HSPB7 is both required for association with and inhibition of polyQ aggregation. Remarkably, replacing the NTD of HSPB1, which itself cannot suppress polyQ aggregation, with the NTD of HSPB7 resulted in a hybrid protein that gained anti-polyQ aggregation activity. The hybrid NTDHSPB7-HSPB1 protein displayed a reduction in oligomer size and, unlike wildtype HSPB1, associated with polyQ. However, experiments with phospho-mimicking HSPB1 mutants revealed that de-oligomerization of HSPB1 alone does not suffice to gain polyQ aggregation-inhibiting activity. Together, our results reveal that the NTD of HSPB7 is both necessary and sufficient to bind to and suppress the aggregation of polyQ-containing proteins.
... HSPBs are able to bind unfolded/misfolded substrates and subsequently new and larger oligomers are formed, preventing irreversible client aggregation and allowing ATP-dependent chaperones recognition to assist protein refolding [6]. HSPBs contribute to protein quality control (PQC) and work to prevent protein aggregation and to generate a pool of non-native proteins that can be rapidly folded [7]. The human genome encodes 10 HSPBs named HSPB1 through HSPB10, with an apparent molecular mass of 12-43 kDa [8]. ...
Article
Full-text available
The cellular response to cancer-induced stress is one of the major aspects regulating cancer development and progression. The Heat Shock Protein B8 (HSPB8) is a small chaperone involved in chaperone-assisted selective autophagy (CASA). CASA promotes the selective degradation of proteins to counteract cell stress such as tumor-induced stress. HSPB8 is also involved in (i) the cell division machinery regulating chromosome segregation and cell cycle arrest in the G0/G1 phase and (ii) inflammation regulating dendritic cell maturation and cytokine production. HSPB8 expression and role are tumor-specific, showing a dual and opposite role. Interestingly, HSPB8 may be involved in the acquisition of chemoresistance to drugs. Despite the fact the mechanisms of HSPB8-mediated CASA activation in tumors need further studies, HSPB8 could represent an important factor in cancer induction and progression and it may be a potential target for anticancer treatment in specific types of cancer. In this review, we will discuss the molecular mechanism underlying HSPB8 roles in normal and cancer conditions. The basic mechanisms involved in anti- and pro-tumoral activities of HSPB8 are deeply discussed together with the pathways that modulate HSPB8 expression, in order to outline molecules with a beneficial effect for cancer cell growth, migration, and death.
... Molecular chaperones, in particular the heat shock proteins (HSPs), are the prime line of defense to counteract protein misfolding and the ensuing protein deposition; they recognize hydrophobic regions of proteins and promote either their refolding or their transfer to the UPS or autophagy systems [230]. The small HSPs are ATPindependent chaperones, a particular class of HSPs that bind misfolded and aggregation-prone proteins, avoiding undesired intermolecular interactions and preventing their aggregation with ensuing loss of function [231,232]. The UPS is localized both in the cytoplasm and in the nuclei; it provides rapid degradation of proteins following their polyubiquitination and subsequent delivery to the proteasome. ...
Article
Full-text available
The increasing extension in life expectancy of human beings in developed countries is accompanied by a progressively greater rate of degenerative diseases associated with lifestyle and aging, most of which are still waiting for effective, not merely symptomatic, therapies. Accordingly, at present, the recommendations aimed at reducing the prevalence of these conditions in the population are limited to a safer lifestyle including physical/mental exercise, a reduced caloric intake, and a proper diet in a convivial environment. The claimed health benefits of the Mediterranean and Asian diets have been confirmed in many clinical trials and epidemiological surveys. These diets are characterized by several features, including low meat consumption, the intake of oils instead of fats as lipid sources, moderate amounts of red wine, and significant amounts of fresh fruit and vegetables. In particular, the latter have attracted popular and scientific attention for their content, though in reduced amounts, of a number of molecules increasingly investigated for their healthy properties. Among the latter, plant polyphenols have raised remarkable interest in the scientific community; in fact, several clinical trials have confirmed that many health benefits of the Mediterranean/Asian diets can be traced back to the presence of significant amounts of these molecules, even though, in some cases, contradictory results have been reported, which highlights the need for further investigation. In light of the results of these trials, recent research has sought to provide information on the biochemical, molecular, epigenetic, and cell biology modifications by plant polyphenols in cell, organismal, animal, and human models of cancer, metabolic, and neurodegenerative pathologies, notably Alzheimer’s and Parkinson disease. The findings reported in the last decade are starting to help to decipher the complex relations between plant polyphenols and cell homeostatic systems including metabolic and redox equilibrium, proteostasis, and the inflammatory response, establishing an increasingly solid molecular basis for the healthy effects of these molecules. Taken together, the data currently available, though still incomplete, are providing a rationale for the possible use of natural polyphenols, or their molecular scaffolds, as nutraceuticals to contrast aging and to combat many associated pathologies.
... 5,6 It is now known that inside a cell, sHSPs function mainly by preventing the aggregation of nonnative, destabilized, denaturing and aggregation-prone proteins during normal as well as stress conditions, thus acting as "stability sensors" of the proteome 7 or by depositing along with the toxic aggregates of misfolded proteins within a confined location of the cell, hence functioning as "aggregases". 8 They also coordinate with the refolding 9,10 and degradation machinery 11 of the cell and contribute to proteostasis. ...
Article
Amyloid‐β (Aβ) protein aggregation in the brain is a pathological hallmark of Alzheimer's disease (AD) however, the underlying molecular mechanisms regulating amyloid aggregation are not well understood. Here, we studied the propitious role of E3 ubiquitin ligase Pirh2 in Aβ protein aggregation in view of its regulatory ligase activity in the ubiquitin‐proteasome system employing both cellular and sporadic rodent models of AD. Pirh2 protein abundance was significantly increased during Streptozotocin (STZ) induced AD conditions, and transient silencing of Pirh2 significantly inhibited the Aβ aggregation and modified the dendrite morphology along with the substantial decrease in choline level in the differentiated neurons. MALDI‐TOF/TOF, coimmunoprecipitation, and UbcH7‐linked in vitro ubiquitylation analysis confirmed the high interaction of Pirh2 with chaperone GRP78. Furthermore, Pirh2 silencing inhibits the STZ induced altered level of endoplasmic reticulum stress and intracellular Ca ²⁺ levels in neuronal N2a cells. Pirh2 silencing also inhibited the AD conditions related to the altered protein abundance of HSP90 and its co‐chaperones which may collectively involve in the reduced burden of amyloid aggregates in neuronal cells. Pirh2 silencing further stabilized the nuclear translocation of phospho‐Nrf2 and inhibited the altered level of autophagy factors. Taken together, our data indicated that Pirh2 is critically involved in STZ induced AD pathogenesis through its interaction with ER‐chaperone GRP78, improves the neuronal connectivity, affects the altered level of chaperones, co‐chaperones, & autophagic markers, and collectively inhibits the Aβ aggregation.
Preprint
Full-text available
Aging is a key risk factor for atherosclerosis (AS). However, its complex etiology and pathological mechanism are still unclear. At present, the study of cell senescence in AS has attracted wide attention, and the characteristics of immunity have also attracted more and more attention of scholars. Therefore, based on the strategy of combining bioinformatics, machine learning and single cell data analysis, this study screened out hub genes, and explored the correlation between aging and immune characteristics in atherosclerosis disease, to clarify the potential pathological mechanism of AS and explore new treatment strategies for AS. This study aims to identify and verify hub genes related to atherosclerosis by using bioinformatics analysis methods. First of all, through the intersection of the most relevant modules of Limma test and weighted correlation network analysis (WGCNA), the differentially expressed genes associated with atherosclerosis (ASDEGs) were identified. Secondly, the differential genes were extracted from 125 aging genes to classify the atherosclerotic samples, and the immune-related information was analyzed. Then, five characteristic genes, including HSPB7, MYEF2, DUSP26, TC2N and PLN, are identified by machine learning methods of support vector machine (SVM), random forest (RF), eXtreme gradient boosting (XGB) and generalized linear model (GLM). Finally, the expression of five hub genes was verified by single cell data analysis. To sum up, this study suggests that HSPB7, MYEF2, DUSP26, TC2N and PLN may play an important role in the pathological mechanism of AS, and aging may also be closely related to the influence of atherosclerotic immune microenvironment. Exploring the molecular mechanism of these hub genes and the differences of aging and different subtypes of immune cells are expected to bring new breakthroughs in the diagnosis and treatment of diseases.
Article
The small heat shock protein (sHsp) family is a group of proteins in which some are induced in response to external stimuli, such as environmental and pathological stresses, while others are constitutively expressed. They show chaperone-like activity, protect cells from apoptosis, and maintain cytoskeletal architecture. Short sequences or fragments ranging from approximately 19-20 residues in sHsps were shown to display chaperone activity in vitro. These sequences are termed sHsp-derived mini-peptides/mini-chaperones. These peptides offer an advantage in providing protective and therapeutic effects over full-length proteins owing to their small molecular weight and easy uptake into the cells. Research on sHsp mini-chaperone therapy has recently received attention and advanced tremendously. sHsp mini-chaperones have shown a wide range of therapeutic effects, such as anti-aggregation of proteins, anti-apoptotic, anti-inflammatory, anti-oxidant, senolytic, and anti-platelet activity. The administration of mini-chaperones into the several disease animal models, including experimental autoimmune encephalomyelitis, cataract, age-related macular degeneration, glaucoma, and thrombosis through various routes reduced symptoms or prevented the progression of the disease. However, it was found that the therapeutic potential of sHsp mini-chaperones is limited by their short turnover and enzymatic degradation in circulation. Nonetheless, carrier molecules approach such as nanoparticles, cell penetration peptides, and extracellular vesicles increased their efficacy by enhancing the uptake, retention time, protection from enzymatic degradation, and site-specific delivery without altering their biological activity. In this context, this review highlights the recent advances in the therapeutic potential of sHsp-derived mini-chaperones, their effect in experimental animal models, and approaches for increasing their efficacy.
Article
HspB7 is one of ten human small heat shock proteins. This protein is expressed only in insulin-dependent tissues (heart, skeletal muscle, and fat tissue), and expression of HspB7 is regulated by many different factors. Single nucleotide polymorphism is characteristic for the HspB7 gene and this polymorphism correlates with cardio-vascular diseases and obesity. HspB7 has an unusual N-terminal sequence, a conservative α-crystallin domain, and very short C-terminal domain lacking conservative IPV tripeptide involved in a small heat shock proteins oligomer formation. Nevertheless, in the isolated state HspB7 forms both small oligomers (probably dimers) and very large oligomers (aggregates). HspB7 is ineffective in suppression of amorphous aggregation of model proteins induced by heating or reduction of disulfide bonds, however it is very effective in prevention of aggregation of huntingtin fragments enriched with Gln residues. HspB7 can be an effective sensor of electrophilic agents. This protein interacts with the contractile and cytoskeleton proteins (filamin C, titin, and actin) and participates in protection of the contractile apparatus and cytoskeleton from different adverse conditions. HspB7 possesses tumor suppressive activity. Further investigations are required to understand molecular mechanisms of HspB7 participation in numerous biological processes.
Article
Photodynamic therapy (PDT) is a clinically approved treatment that exerts a selective cytotoxic activity toward cancer cells. The procedure involves the administration of a photosensitizer drug followed by its activation by visible light. In presence of oxygen, a series of events lead to tumor cell death. PDT releases different cell signals, some of these lead to death while others can lead to survival. The surviving or resistant cells contribute to the recurrence of tumors after treatment, from which the necessity to understand this molecular response induced by PDT arises. It has been shown that both Heat Shock Proteins (HSPs) and autophagy promote PDT resistance. Moreover, both of them can be stimulated by PDT treatment. However, the molecular interplay between HSP and autophagy in the photodynamic therapy context is poorly understood. We studied whether PDT induces autophagic activity through HSPs. We demonstrated that PDT promoted HSP27 expression, which in turn triggered autophagic cell survival as well as inhibited apoptosis in colon cancer cells. In addition, an overexpression of HSP27/autophagy axis was observed in skin carcinoma cells resistant to PDT.
ResearchGate has not been able to resolve any references for this publication.